Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 16 October 2017

The hygiene hypothesis in autoimmunity: the role of pathogens and commensals

  • Jean-François Bach 1 , 2 , 3  

Nature Reviews Immunology volume  18 ,  pages 105–120 ( 2018 ) Cite this article

19k Accesses

311 Citations

265 Altmetric

Metrics details

  • Autoimmune diseases
  • Toll-like receptors

The initial application of the hygiene hypothesis for autoimmune diseases proposed in the early 2000s has been confirmed and consolidated by a wealth of published data in both animal models and human autoimmune conditions.

The hygiene hypothesis probably explains the uneven geographical distribution of autoimmune diseases in the world. Individuals migrating from countries with low incidence of autoimmune diseases to countries with high incidence develop the disease with the frequency of the host country, provided that migration occurred at a young age and under a threshold that varies according to the disease.

Pathogenic bacteria, viruses and parasites are often endowed with strong protective effects on autoimmunity even when infection occurs late after birth.

Gut commensal bacteria may also have a protective role in autoimmunity when administered early in life.

Pathogens, parasites and commensals essentially act by stimulating immune regulatory pathways, implicating the innate and the adaptive immune system. Importantly, the effect is seen with both living organisms and their derivatives or purified extracts.

Both pathogens and commensals stimulate pattern recognition receptors, including Toll-like receptors (TLRs) to protect against autoimmunity. This effect may be mimicked by TLR agonists acting through pharmacological stimulation or desensitization of the target receptor.

The incidence of autoimmune diseases has been steadily rising. Concomitantly, the incidence of most infectious diseases has declined. This observation gave rise to the hygiene hypothesis, which postulates that a reduction in the frequency of infections contributes directly to the increase in the frequency of autoimmune and allergic diseases. This hypothesis is supported by robust epidemiological data, but the underlying mechanisms are unclear. Pathogens are known to be important, as autoimmune disease is prevented in various experimental models by infection with different bacteria, viruses and parasites. Gut commensal bacteria also play an important role: dysbiosis of the gut flora is observed in patients with autoimmune diseases, although the causal relationship with the occurrence of autoimmune diseases has not been established. Both pathogens and commensals act by stimulating immunoregulatory pathways. Here, I discuss the importance of innate immune receptors, in particular Toll-like receptors, in mediating the protective effect of pathogens and commensals on autoimmunity.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

hygiene hypothesis original paper

Similar content being viewed by others

hygiene hypothesis original paper

Interaction between microbiota and immunity in health and disease

hygiene hypothesis original paper

The impact of the gut microbiome on extra-intestinal autoimmune diseases

hygiene hypothesis original paper

Host–microbiota interactions in inflammatory bowel disease

Strachan, D. P. Hay fever, hygiene, and household size. BMJ 299 , 1259–1260 (1989). This is a visionary epidemiological study that paved the way for the hygiene hypothesis in atopic diseases.

CAS   PubMed   PubMed Central   Google Scholar  

Strachan, D. P. Family size, infection and atopy: the first decade of the “hygiene hypothesis”. Thorax 55 (Suppl. 1), S2–S10 (2000).

PubMed   PubMed Central   Google Scholar  

Ege, M. J. et al. Exposure to environmental microorganisms and childhood asthma. N. Engl. J. Med. 364 , 701–709 (2011).

CAS   PubMed   Google Scholar  

Greenwood, B. M., Herrick, E. M. & Voller, A. Suppression of autoimmune disease in NZB and (NZB x NZW) F1 hybrid mice by infection with malaria. Nature 226 , 266–267 (1970).

Greenwood, B. M., Herrick, E. M. & Voller, A. Can parasitic infection suppress autoimmune disease? Proc. R. Soc. Med. 63 , 19–20 (1970).

Rook, G. A. & Stanford, J. L. Give us this day our daily germs. Immunol. Today 19 , 113–116 (1998).

Sewell, D. L., Reinke, E. K., Hogan, L. H., Sandor, M. & Fabry, Z. Immunoregulation of CNS autoimmunity by helminth and mycobacterial infections. Immunol. Lett. 82 , 101–110 (2002).

Oldstone, M. B. Prevention of type I diabetes in nonobese diabetic mice by virus infection. Science 239 , 500–502 (1988). This seminal study demonstrates the protective effect of a viral infection on the development of spontaneous autoimmune IDDM in NOD mice.

Oldstone, M. B., Ahmed, R. & Salvato, M. Viruses as therapeutic agents. II. Viral reassortants map prevention of insulin-dependent diabetes mellitus to the small RNA of lymphocytic choriomeningitis virus. J. Exp. Med. 171 , 2091–2100 (1990).

Bach, J. F. The effect of infections on susceptibility to autoimmune and allergic diseases. N. Engl. J. Med. 347 , 911–920 (2002).

PubMed   Google Scholar  

Bach, J. F. Protective role of infections and vaccinations on autoimmune diseases. J. Autoimmun. 16 , 347–353 (2001).

Deckers, I. A. et al. Investigating international time trends in the incidence and prevalence of atopic eczema 1990-2010: a systematic review of epidemiological studies. PLoS ONE 7 , e39803 (2012).

Kotz, D., Simpson, C. R. & Sheikh, A. Incidence, prevalence, and trends of general practitioner-recorded diagnosis of peanut allergy in England, 2001 to 2005. J. Allergy Clin. Immunol. 127 , 623–630.e1 (2011).

Patterson, C. C. et al. Trends in childhood type 1 diabetes incidence in Europe during 1989-2008: evidence of non-uniformity over time in rates of increase. Diabetologia 55 , 2142–2147 (2012).

Karvonen, M., Pitkaniemi, J. & Tuomilehto, J. The onset age of type 1 diabetes in Finnish children has become younger. The Finnish Childhood Diabetes Registry Group. Diabetes Care 22 , 1066–1070 (1999).

Koch-Henriksen, N. & Sorensen, P. S. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurol. 9 , 520–532 (2010).

Mackenzie, I. S., Morant, S. V., Bloomfield, G. A., MacDonald, T. M. & O'Riordan, J. Incidence and prevalence of multiple sclerosis in the UK 1990-2010: a descriptive study in the General Practice Research Database. J. Neurol. Neurosurg. Psychiatry 85 , 76–84 (2014).

Grytten, N., Torkildsen, O. & Myhr, K. M. Time trends in the incidence and prevalence of multiple sclerosis in Norway during eight decades. Acta Neurol. Scand. 132 , 29–36 (2015).

Houzen, H. et al. Increased prevalence, incidence, and female predominance of multiple sclerosis in northern Japan. J. Neurol. Sci. 323 , 117–122 (2012).

Li, X. H. et al. A nine-year prospective study on the incidence of childhood type 1 diabetes mellitus in China. Biomed. Environ. Sci. 13 , 263–270 (2000).

Handel, A. E., Handunnetthi, L., Ebers, G. C. & Ramagopalan, S. V. Type 1 diabetes mellitus and multiple sclerosis: common etiological features. Nat. Rev. Endocrinol. 5 , 655–664 (2009).

Stewart, A. W., Mitchell, E. A., Pearce, N., Strachan, D. P. & Weiland, S. K. The relationship of per capita gross national product to the prevalence of symptoms of asthma and other atopic diseases in children (ISAAC). Int. J. Epidemiol. 30 , 173–179 (2001).

Patterson, C. C., Carson, D. J. & Hadden, D. R. Epidemiology of childhood IDDM in Northern Ireland 1989-1994: low incidence in areas with highest population density and most household crowding. Diabetologia 39 , 1063–1069 (1996).

Paalanen, L., Prattala, R., Palosuo, H., Helakorpi, S. & Laatikainen, T. Socio-economic differences in the use of dairy fat in Russian and Finnish Karelia, 1994–2004. Int. J. Publ. Health 55 , 325–337 (2010).

Google Scholar  

Kondrashova, A. et al. A six-fold gradient in the incidence of type 1 diabetes at the eastern border of Finland. Ann. Med. 37 , 67–72 (2005).

Laatikainen, T. et al. Allergy gap between Finnish and Russian Karelia on increase. Allergy 66 , 886–892 (2011).

Kondrashova, A. et al. Signs of beta-cell autoimmunity in nondiabetic schoolchildren: a comparison between Russian Karelia with a low incidence of type 1 diabetes and Finland with a high incidence rate. Diabetes Care 30 , 95–100 (2007).

Kuehni, C. E., Strippoli, M. P., Low, N. & Silverman, M. Asthma in young south Asian women living in the United Kingdom: the importance of early life. Clin. Exp. Allergy 37 , 47–53 (2007).

Bodansky, H. J., Staines, A., Stephenson, C., Haigh, D. & Cartwright, R. Evidence for an environmental effect in the aetiology of insulin dependent diabetes in a transmigratory population. BMJ 304 , 1020–1022 (1992).

Feltbower, R. G. et al. Trends in the incidence of childhood diabetes in south Asians and other children in Bradford. UK. Diabet. Med. 19 , 162–166 (2002).

Dean, G. & Elian, M. Age at immigration to England of Asian and Caribbean immigrants and the risk of developing multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 63 , 565–568 (1997).

Gale, C. R. & Martyn, C. N. Migrant studies in multiple sclerosis. Prog. Neurobiol. 47 , 425–448 (1995).

Kostic, A. D. et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe 17 , 260–273 (2015). This is a remarkable study reporting the detailed follow-up of the gut microbiota composition in children at risk of developing IDDM from birth to the onset of hyperglycaemia.

Ball, T. M. et al. Siblings, day-care attendance, and the risk of asthma and wheezing during childhood. N. Engl. J. Med. 343 , 538–543 (2000).

Cardwell, C. R. et al. Birth order and childhood type 1 diabetes risk: a pooled analysis of 31 observational studies. Int. J. Epidemiol. 40 , 363–374 (2011).

Almeida, M. C. et al. The effect of antihelminthic treatment on subjects with asthma from an endemic area of schistosomiasis: a randomized, double-blinded, and placebo-controlled trial. J. Parasitol. Res. 2012 , 296856 (2012).

Fleming, J. O. et al. Probiotic helminth administration in relapsing-remitting multiple sclerosis: a phase 1 study. Mult. Scler. 17 , 743–754 (2011).

Larson, J. D. et al. Murine gammaherpesvirus 68 infection protects lupus-prone mice from the development of autoimmunity. Proc. Natl Acad. Sci. USA 109 , E1092–E1100 (2012).

Alyanakian, M. A. et al. Transforming growth factor-beta and natural killer T-cells are involved in the protective effect of a bacterial extract on type 1 diabetes. Diabetes 55 , 179–185 (2006).

Finlay, C. M., Walsh, K. P. & Mills, K. H. Induction of regulatory cells by helminth parasites: exploitation for the treatment of inflammatory diseases. Immunol. Rev. 259 , 206–230 (2014).

Gause, W. C. & Maizels, R. M. Macrobiota -— helminths as active participants and partners of the microbiota in host intestinal homeostasis. Curr. Opin. Microbiol. 32 , 14–18 (2016). This study reports that the helminth H. polygyrus produces a TGFβ mimic that fully reproduces the effect of this immunoregulatory cytokine on the host immune system.

Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504 , 451–455 (2013).

De Filippo, C. et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci. USA 107 , 14691–14696 (2010).

Lin, A. et al. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS ONE 8 , e53838 (2013).

David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505 , 559–563 (2014).

Schmidt, B. et al. Establishment of normal gut microbiota is compromised under excessive hygiene conditions. PLoS ONE 6 , e28284 (2011). This study provides direct confirmation of the role of hygiene on gut microbiota composition in an original experimental model using piglets reared in conventional or clean conditions.

Vatanen, T. et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165 , 842–853 (2016). This is a comparative study of the gut microbiota composition in individuals from Finland and Karelia, two neighbouring countries with a substantial difference in the incidence of IDDM. The study demonstrates structural differences in LPS produced by 'non-protective' versus 'protective' commensals in the Finnish and the Karelian microbiota, respectively.

Alam, C. et al. Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice. Diabetologia 54 , 1398–1406 (2011).

Candon, S. et al. Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes. PLoS ONE 10 , e0125448 (2015).

Yurkovetskiy, L. et al. Gender bias in autoimmunity is influenced by microbiota. Immunity 39 , 400–412 (2013).

Brown, K. et al. Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice. ISME J. 10 , 321–332 (2016).

Berer, K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479 , 538–541 (2011).

Lee, Y. K., Menezes, J. S., Umesaki, Y. & Mazmanian, S. K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 108 (Suppl. 1), 4615–4622 (2011).

Goverman, J. et al. Transgenic mice that express a myelin basic protein-specific T cell receptor develop spontaneous autoimmunity. Cell 72 , 551–560 (1993).

Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139 , 485–498 (2009).

Gaboriau-Routhiau, V. et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31 , 677–689 (2009).

Barclay, W. & Shinohara, M. L. Inflammasome activation in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Brain Pathol. 27 , 213–219 (2017).

Dumas, A. et al. The inflammasome pyrin contributes to pertussis toxin-induced IL-1β synthesis, neutrophil intravascular crawling and autoimmune encephalomyelitis. PLoS Pathog. 10 , e1004150 (2014).

Aumeunier, A. et al. Systemic Toll-like receptor stimulation suppresses experimental allergic asthma and autoimmune diabetes in NOD mice. PLoS ONE 5 , e11484 (2010). This is the first systematic comparative study of the protective effect of different TLR agonists on autoimmunity and experimental asthma, which shows that distinct mechanisms underlie the therapeutic activity, depending on the TLR agonist used.

Calcinaro, F. et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia 48 , 1565–1575 (2005).

Falcone, M. et al. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of Lactobacillus casei . J. Diabetes Res. 105 , 643–649 (1997).

Lavasani, S. et al. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS ONE 5 , e9009 (2010).

Markle, J. G. et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339 , 1084–1088 (2013).

Wen, L. et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 455 , 1109–1113 (2008).

Peng, J. et al. Long term effect of gut microbiota transfer on diabetes development. J. Autoimmun 53 , 85–94 (2014).

Cardwell, C. R. et al. Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: a meta-analysis of observational studies. Diabetologia 51 , 726–735 (2008).

Clausen, T. D. et al. Prelabor cesarean section and risk of childhood type 1 diabetes: a nationwide register-based cohort study. Epidemiology 27 , 547–555 (2016).

Maghzi, A. H. et al. Cesarean delivery may increase the risk of multiple sclerosis. Mult. Scler. 18 , 468–471 (2012).

Knights, D. et al. Use of antibiotics in childhood and risk of Type 1 diabetes: a population-based case-control study. Nat. Microbiol. 34 , 272–277 (2017).

Boursi, B., Mamtani, R., Haynes, K. & Yang, Y. X. The effect of past antibiotic exposure on diabetes risk. Eur. J. Endocrinol. 172 , 639–648 (2015).

Clausen, T. D. et al. Broad-spectrum antibiotic treatment and subsequent childhood type 1 diabetes: a nationwide Danish cohort study. PLoS ONE 11 , e0161654 (2016).

Hviid, A. & Svanstrom, H. Antibiotic use and type 1 diabetes in childhood. Am. J. Epidemiol. 169 , 1079–1084 (2009).

Livanos, A. E. et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat. Microbiol. 1 , 16140 (2016).

Alonso, A., Jick, S. S., Jick, H. & Hernan, M. A. Antibiotic use and risk of multiple sclerosis. Am. J. Epidemiol. 163 , 997–1002 (2006).

Ljungberg, M., Korpela, R., Ilonen, J., Ludvigsson, J. & Vaarala, O. Probiotics for the prevention of beta cell autoimmunity in children at genetic risk of type 1 diabetes — the PRODIA study. Ann. NY Acad. Sci. 1079 , 360–364 (2006).

Uusitalo, U. et al. Association of early exposure of probiotics and islet autoimmunity in the TEDDY study. JAMA Pediatr. 170 , 20–28 (2016).

Pelucchi, C. et al. Probiotics supplementation during pregnancy or infancy for the prevention of atopic dermatitis: a meta-analysis. Epidemiology 23 , 402–414 (2012).

Liacopoulos, P. & Ben-Efraim, S. Antigenic competition. Prog. Allergy 18 , 97–204 (1975).

Pross, H. F. & Eidinger, D. Antigenic competition: a review of nonspecific antigen-induced suppression. Adv. Immunol. 18 , 133–168 (1974).

Buus, S., Sette, A., Colon, S. M., Miles, C. & Grey, H. M. The relation between major histocompatibility complex (MHC) restriction and the capacity of Ia to bind immunogenic peptides. Science 235 , 1353–1358 (1987).

Guillet, J. G. et al. Immunological self, nonself discrimination. Science 235 , 865–870 (1987).

Almeida, A. R., Rocha, B., Freitas, A. A. & Tanchot, C. Homeostasis of T cell numbers: from thymus production to peripheral compartmentalization and the indexation of regulatory T cells. Semin. Immunol. 17 , 239–249 (2005).

Surh, C. D. & Sprent, J. Homeostasis of naive and memory T cells. Immunity 29 , 848–862 (2008).

Qin, H. Y., Sadelain, M. W., Hitchon, C., Lauzon, J. & Singh, B. Complete Freund's adjuvant-induced T cells prevent the development and adoptive transfer of diabetes in nonobese diabetic mice. J. Immunol. 150 , 2072–2080 (1993).

Qin, H. Y. & Singh, B. BCG vaccination prevents insulin-dependent diabetes mellitus (IDDM) in NOD mice after disease acceleration with cyclophosphamide. J. Autoimmun. 10 , 271–278 (1997).

Lee, I. F., Qin, H., Trudeau, J., Dutz, J. & Tan, R. Regulation of autoimmune diabetes by complete Freund's adjuvant is mediated by NK cells. J. Immunol. 172 , 937–942 (2004).

Tian, B. et al. Upregulating CD4 + CD25 + FOXP3 + regulatory T cells in pancreatic lymph nodes in diabetic NOD mice by adjuvant immunotherapy. Transplantation 87 , 198–206 (2009).

Serreze, D. V. et al. Th1 to Th2 cytokine shifts in nonobese diabetic mice: sometimes an outcome, rather than the cause, of diabetes resistance elicited by immunostimulation. J. Immunol. 166 , 1352–1359 (2001).

Mori, Y., Kodaka, T., Kato, T., Kanagawa, E. M. & Kanagawa, O. Critical role of IFN-γ in CFA-mediated protection of NOD mice from diabetes development. Int. Immunol. 21 , 1291–1299 (2009).

Salomon, B. et al. B7/CD28 costimulation is essential for the homeostasis of the CD4 + CD25 + immunoregulatory T cells that control autoimmune diabetes. Immunity 12 , 431–440 (2000).

Caramalho, I. et al. Regulatory T cells contribute to diabetes protection in lipopolysaccharide-treated non-obese diabetic mice. Scand. J. Immunol. 74 , 585–595 (2011).

Tian, J. et al. Lipopolysaccharide-activated B cells down-regulate Th1 immunity and prevent autoimmune diabetes in nonobese diabetic mice. J. Immunol. 167 , 1081–1089 (2001).

Fillatreau, S., Sweenie, C. H., McGeachy, M. J., Gray, D. & Anderton, S. M. B cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 3 , 944–950 (2002).

Mauri, C., Gray, D., Mushtaq, N. & Londei, M. Prevention of arthritis by interleukin 10-producing B cells. J. Exp. Med. 197 , 489–501 (2003).

Mizoguchi, A., Mizoguchi, E., Takedatsu, H., Blumberg, R. S. & Bhan, A. K. Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation. Immunity 16 , 219–230 (2002).

Shen, P. & Fillatreau, S. Antibody-independent functions of B cells: a focus on cytokines. Nat. Rev. Immunol. 15 , 441–451 (2015).

Shen, P. et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 507 , 366–370 (2014).

Shen, P. & Fillatreau, S. Suppressive functions of B cells in infectious diseases. Int. Immunol. 27 , 513–519 (2015).

Filippi, C. M., Estes, E. A., Oldham, J. E. & von Herrath, M. G. Immunoregulatory mechanisms triggered by viral infections protect from type 1 diabetes in mice. J. Clin. Invest. 119 , 1515–1523 (2009).

Cooke, A. et al. Infection with Schistosoma mansoni prevents insulin dependent diabetes mellitus in non-obese diabetic mice. Parasite Immunol. 21 , 169–176 (1999).

Zaccone, P. et al. Schistosoma mansoni antigens modulate the activity of the innate immune response and prevent onset of type 1 diabetes. Eur. J. Immunol. 33 , 1439–1449 (2003).

Grainger, J. R. et al. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway. J. Exp. Med. 207 , 2331–2341 (2010).

Ince, M. N. et al. Role of T cell TGF-β signaling in intestinal cytokine responses and helminthic immune modulation. Eur. J. Immunol. 39 , 1870–1878 (2009).

Liu, Q. et al. Helminth infection can reduce insulitis and type 1 diabetes through CD25- and IL-10-independent mechanisms. Infect. Immun. 77 , 5347–5358 (2009).

Walk, S. T., Blum, A. M., Ewing, S. A., Weinstock, J. V. & Young, V. B. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus . Inflamm. Bowel Dis. 16 , 1841–1849 (2010).

Ramanan, D. et al. Helminth infection promotes colonization resistance via type 2 immunity. Science 352 , 608–612 (2016).

Hubner, M. P., Stocker, J. T. & Mitre, E. Inhibition of type 1 diabetes in filaria-infected non-obese diabetic mice is associated with a T helper type 2 shift and induction of FoxP3 + regulatory T cells. Immunology 127 , 512–522 (2009).

Hubner, M. P. et al. Helminth protection against autoimmune diabetes in nonobese diabetic mice is independent of a type 2 immune shift and requires TGF-β. J. Immunol. 188 , 559–568 (2012).

Lund, M. E. et al. Secreted proteins from the helminth Fasciola hepatica inhibit the initiation of autoreactive T cell responses and prevent diabetes in the NOD mouse. PLoS ONE 9 , e86289 (2014).

Finlay, C. M. et al. Helminth products protect against autoimmunity via innate type 2 cytokines IL-5 and IL-33, which promote eosinophilia. J. Immunol. 196 , 703–714 (2016).

Cording, S., Medvedovic, J., Aychek, T. & Eberl, G. Innate lymphoid cells in defense, immunopathology and immunotherapy. Nat. Immunol. 17 , 755–757 (2016).

Dolpady, J. et al. Oral probiotic VSL#3 prevents autoimmune diabetes by modulating microbiota and promoting indoleamine 2,3-dioxygenase-enriched tolerogenic intestinal environment. J. Diabetes Res. 2016 , 7569431 (2016).

Kobayashi, T. et al. Probiotic upregulation of peripheral IL-17 responses does not exacerbate neurological symptoms in experimental autoimmune encephalomyelitis mouse models. Immunopharmacol. Immunotoxicol. 34 , 423–433 (2012).

Ochoa-Reparaz, J. et al. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J. Immunol. 183 , 6041–6050 (2009).

Ochoa-Reparaz, J. et al. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 3 , 487–495 (2010). This is an important study describing a constituent of the commensal organism B. fragilis (that is, PSA) with remarkable protective activity in models of EAE and colitis.

Mazmanian, S. K., Round, J. L. & Kasper, D. L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453 , 620–625 (2008).

Round, J. L. & Mazmanian, S. K. Inducible Foxp3 + regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl Acad. Sci. USA 107 , 12204–12209 (2010).

Chinen, T., Volchkov, P. Y., Chervonsky, A. V. & Rudensky, A. Y. A critical role for regulatory T cell-mediated control of inflammation in the absence of commensal microbiota. J. Exp. Med. 207 , 2323–2330 (2010).

Ivanov, I. I. et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4 , 337–349 (2008).

Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331 , 337–341 (2011).

Nagano, Y., Itoh, K. & Honda, K. The induction of Treg cells by gut-indigenous Clostridium . Curr. Opin. Immunol. 24 , 392–397 (2012).

Gagliani, N. et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat. Med. 19 , 739–746 (2013).

Apetoh, L. et al. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Brain Pathol. 11 , 854–861 (2010).

CAS   Google Scholar  

Weiner, H. L., da Cunha, A. P., Quintana, F. & Wu, H. Oral tolerance. Immunol. Rev. 241 , 241–259 (2011).

Pistoia, V. & Raffaghello, L. Mesenchymal stromal cells and autoimmunity. Int. Immunol. 29 , 49–58 (2017).

Uccelli, A., Moretta, L. & Pistoia, V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 8 , 726–736 (2008).

Sica, A. & Massarotti, M. Myeloid suppressor cells in cancer and autoimmunity. J. Autoimmun. http://dx.doi.org/10.1016/j.jaut.2017.07.010 (2017).

Karumuthil-Melethil, S., Perez, N., Li, R. & Vasu, C. Induction of innate immune response through TLR2 and dectin 1 prevents type 1 diabetes. J. Immunol. 181 , 8323–8334 (2008).

Karumuthil-Melethil, S. et al. TLR2- and Dectin 1-associated innate immune response modulates T-cell response to pancreatic β-cell antigen and prevents type 1 diabetes. Diabetes 64 , 1341–1357 (2015).

Serreze, D. V., Hamaguchi, K. & Leiter, E. H. Immunostimulation circumvents diabetes in NOD/Lt mice. J. Autoimmun. 2 , 759–776 (1989).

Quintana, F. J., Rotem, A., Carmi, P. & Cohen, I. R. Vaccination with empty plasmid DNA or CpG oligonucleotide inhibits diabetes in nonobese diabetic mice: modulation of spontaneous 60-kDa heat shock protein autoimmunity. J. Immunol. 165 , 6148–6155 (2000).

Round, J. L. et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332 , 974–977 (2011).

Wang, Y. et al. An intestinal commensal symbiosis factor controls neuroinflammation via TLR2-mediated CD39 signalling. Nat. Commun. 5 , 4432 (2014).

Filippi, C. M. et al. TLR2 signaling improves immunoregulation to prevent type 1 diabetes. Eur. J. Immunol. 41 , 1399–1409 (2011).

Xiong, Y. et al. Endotoxin tolerance inhibits Lyn and c-Src phosphorylation and association with Toll-like receptor 4 but increases expression and activity of protein phosphatases. J. Innate Immun. 8 , 171–184 (2016).

Freudenberg, M. A. & Galanos, C. Induction of tolerance to lipopolysaccharide (LPS)-D-galactosamine lethality by pretreatment with LPS is mediated by macrophages. Infect. Immun. 56 , 1352–1357 (1988).

Medvedev, A. E., Kopydlowski, K. M. & Vogel, S. N. Inhibition of lipopolysaccharide-induced signal transduction in endotoxin-tolerized mouse macrophages: dysregulation of cytokine, chemokine, and Toll-like receptor 2 and 4 gene expression. J. Immunol. 164 , 5564–5574 (2000). This is a comprehensive work on changes in intracellular signalling in macrophages that lead to LPS (endotoxin) tolerance, which highlights the fundamental role of the activation of various phosphatases.

Kim, D. H. et al. Inhibition of autoimmune diabetes by TLR2 tolerance. J. Immunol. 187 , 5211–5220 (2011).

Anstadt, E. J., Fujiwara, M., Wasko, N., Nichols, F. & Clark, R. B. TLR tolerance as a treatment for central nervous system autoimmunity. J. Immunol. 197 , 2110–2118 (2016). This is an interesting report demonstrating that low doses of two different TLR2 ligands attenuate adoptively transferred EAE through receptor desensitization. One of these TLR2 ligands is a human microbiome product, which has significantly decreased serum levels in patients with multiple sclerosis compared with unaffected controls.

Hayashi, T. et al. Treatment of autoimmune inflammation by a TLR7 ligand regulating the innate immune system. PLoS ONE 7 , e45860 (2012).

Schuijs, M. J. et al. Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells. Science 349 , 1106–1110 (2015). This is a study highlighting the potential in vivo relevance of TLR desensitization by LPS in humans. The novel finding is that the LPS-induced TLR4 desensitization targets the lung epithelium and requires the ubiquitin-modifying enzyme A20.

Siebeneicher, S. et al. Epicutaneous immune modulation with Bet v 1 plus R848 suppresses allergic asthma in a murine model. Allergy 69 , 328–337 (2014).

Aryan, Z. & Rezaei, N. Toll-like receptors as targets for allergen immunotherapy. Curr. Opin. Allergy Clin. Immunol. 15 , 568–574 (2015).

Burrows, M. P., Volchkov, P., Kobayashi, K. S. & Chervonsky, A. V. Microbiota regulates type 1 diabetes through Toll-like receptors. Proc. Natl Acad. Sci. USA 112 , 9973–9977 (2015). This is a unique study that uses a genetic approach and proposes a distinct role for single TLRs in their capacity to modulate autoimmunity.

Bras, A. & Aguas, A. P. Diabetes-prone NOD mice are resistant to Mycobacterium avium and the infection prevents autoimmune disease. Immunology 89 , 20–25 (1996).

Lee, J., Reinke, E. K., Zozulya, A. L., Sandor, M. & Fabry, Z. Mycobacterium bovis bacille Calmette-Guerin infection in the CNS suppresses experimental autoimmune encephalomyelitis and Th17 responses in an IFN-γ-independent manner. J. Immunol. 181 , 6201–6212 (2008).

Newland, S. A. et al. PD-L1 blockade overrides Salmonella typhimurium -mediated diabetes prevention in NOD mice: no role for Tregs. Eur. J. Immunol. 41 , 2966–2976 (2011).

Drescher, K. M., Kono, K., Bopegamage, S., Carson, S. D. & Tracy, S. Coxsackievirus B3 infection and type 1 diabetes development in NOD mice: insulitis determines susceptibility of pancreatic islets to virus infection. Virology 329 , 381–394 (2004).

Tracy, S. et al. Toward testing the hypothesis that group B coxsackieviruses (CVB) trigger insulin-dependent diabetes: inoculating nonobese diabetic mice with CVB markedly lowers diabetes incidence. J. Virol. 76 , 12097–12111 (2002).

Davydova, B. et al. Coxsackievirus immunization delays onset of diabetes in non-obese diabetic mice. J. Med. Virol. 69 , 510–520 (2003).

Richer, M. J., Straka, N., Fang, D., Shanina, I. & Horwitz, M. S. Regulatory T-cells protect from type 1 diabetes after induction by coxsackievirus infection in the context of transforming growth factor-β. Diabetes 57 , 1302–1311 (2008).

Hermitte, L. et al. Paradoxical lessening of autoimmune processes in non-obese diabetic mice after infection with the diabetogenic variant of encephalomyocarditis virus. Eur. J. Immunol. 20 , 1297–1303 (1990).

Takei, I. et al. Suppression of development of diabetes in NOD mice by lactate dehydrogenase virus infection. J. Autoimmun. 5 , 665–673 (1992).

Wilberz, S., Partke, H. J., Dagnaes-Hansen, F. & Herberg, L. Persistent MHV (mouse hepatitis virus) infection reduces the incidence of diabetes mellitus in non-obese diabetic mice. Diabetologia 34 , 2–5 (1991).

Smith, K. A., Efstathiou, S. & Cooke, A. Murine gammaherpesvirus-68 infection alters self-antigen presentation and type 1 diabetes onset in NOD mice. J. Immunol. 179 , 7325–7333 (2007).

Mishra, P. K., Patel, N., Wu, W., Bleich, D. & Gause, W. C. Prevention of type 1 diabetes through infection with an intestinal nematode parasite requires IL-10 in the absence of a Th2-type response. Mucosal Immunol. 6 , 297–308 (2013).

Saunders, K. A., Raine, T., Cooke, A. & Lawrence, C. E. Inhibition of autoimmune type 1 diabetes by gastrointestinal helminth infection. Infect. Immun. 75 , 397–407 (2007).

Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16 , 407–420 (2016).

Inoue, M., Williams, K. L., Gunn, M. D. & Shinohara, M. L. NLRP3 inflammasome induces chemotactic immune cell migration to the CNS in experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 109 , 10480–10485 (2012).

Inoue, M. et al. Interferon-β therapy against EAE is effective only when development of the disease depends on the NLRP3 inflammasome. Sci. Signal. 5 , ra38 (2012).

Cuda, C. M., Pope, R. M. & Perlman, H. The inflammatory role of phagocyte apoptotic pathways in rheumatic diseases. Nat. Rev. Rheumatol. 12 , 543–558 (2016).

de Souza, H. S. & Fiocchi, C. Immunopathogenesis of IBD: current state of the art. Nat. Rev. Gastroenterol. Hepatol. 13 , 13–27 (2016).

von Moltke, J., Ayres, J. S., Kofoed, E. M., Chavarria-Smith, J. & Vance, R. E. Recognition of bacteria by inflammasomes. Annu. Rev. Immunol. 31 , 73–106 (2013).

Rzepecka, J. et al. Prophylactic and therapeutic treatment with a synthetic analogue of a parasitic worm product prevents experimental arthritis and inhibits IL-1beta production via NRF2-mediated counter-regulation of the inflammasome. J. Autoimmun. 60 , 59–73 (2015).

Patterson, C. et al. Diabetes in the young — a global view and worldwide estimates of numbers of children with type 1 diabetes. Diabetes Res. Clin. Pract. 103 , 161–175 (2013).

Giongo, A. et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 5 , 82–91 (2011).

Brown, C. T. et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 6 , e25792 (2011).

de Goffau, M. C. et al. Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes 62 , 1238–1244 (2013).

Murri, M. et al. Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med. 11 , 46 (2013).

Soyucen, E. et al. Differences in the gut microbiota of healthy children and those with type 1 diabetes. Pediatr. Int. 56 , 336–343 (2014).

Davis-Richardson, A. G. et al. Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front. Microbiol. 5 , 678 (2014).

Endesfelder, D. et al. Compromised gut microbiota networks in children with anti-islet cell autoimmunity. Diabetes 63 , 2006–2014 (2014).

Mejia-Leon, M. E., Petrosino, J. F., Ajami, N. J., Dominguez-Bello, M. G. & de la Barca, A. M. Fecal microbiota imbalance in Mexican children with type 1 diabetes. Sci. Rep. 4 , 3814 (2014).

Alkanani, A. K. et al. Alterations in intestinal microbiota correlate with susceptibility to type 1 diabetes. Diabetes 64 , 3510–3520 (2015).

Qi, C. J. et al. Imbalance of fecal microbiota at newly diagnosed type 1 diabetes in Chinese children. Chin. Med. J. 129 , 1298–1304 (2016).

Maffeis, C. et al. Association between intestinal permeability and faecal microbiota composition in Italian children with beta cell autoimmunity at risk for type 1 diabetes. Diabetes Metab. Res. Rev. 32 , 700–709 (2016).

Stewart, C. J. et al. Gut microbiota of Type 1 diabetes patients with good glycaemic control and high physical fitness is similar to people without diabetes: an observational study. Diabet Med. 34 , 127–134 (2017).

Miyake, S. et al. Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belonging to Clostridia XIVa and IV clusters. PLoS ONE 10 , e0137429 (2015).

Tremlett, H. et al. Gut microbiota in early pediatric multiple sclerosis: a case-control study. Eur. J. Neurol. 23 , 1308–1321 (2016).

Chen, J. et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci. Rep. 6 , 28484 (2016).

Hevia, A. et al. Intestinal dysbiosis associated with systemic lupus erythematosus. mBio 5 , e01548–e01514 (2014).

Wilson, C. S., Elizer, S. K., Marshall, A. F., Stocks, B. T. & Moore, D. J. Regulation of B lymphocyte responses to Toll-like receptor ligand binding during diabetes prevention in non-obese diabetic (NOD) mice. J. Diabetes 8 , 120–131 (2016).

Zhang, Y. et al. TLR9 blockade inhibits activation of diabetogenic CD8 + T cells and delays autoimmune diabetes. J. Immunol. 184 , 5645–5653 (2010).

Manicassamy, S. et al. Toll-like receptor 2-dependent induction of vitamin A-metabolizing enzymes in dendritic cells promotes T regulatory responses and inhibits autoimmunity. Nat. Med. 15 , 401–409 (2009).

Manoharan, I. et al. TLR2-dependent activation of beta-catenin pathway in dendritic cells induces regulatory responses and attenuates autoimmune inflammation. J. Immunol. 193 , 4203–4213 (2014).

Li, H. et al. Low dose zymosan ameliorates both chronic and relapsing experimental autoimmune encephalomyelitis. J. Neuroimmunol. 254 , 28–38 (2013).

Dillon, S. et al. Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance. J. Clin. Invest. 116 , 916–928 (2006).

Barrat, F. J. & Coffman, R. L. Development of TLR inhibitors for the treatment of autoimmune diseases. Immunol. Rev. 223 , 271–283 (2008).

Barrat, F. J., Meeker, T., Chan, J. H., Guiducci, C. & Coffman, R. L. Treatment of lupus-prone mice with a dual inhibitor of TLR7 and TLR9 leads to reduction of autoantibody production and amelioration of disease symptoms. Eur. J. Immunol. 37 , 3582–3586 (2007).

Wong, F. S. et al. The role of Toll-like receptors 3 and 9 in the development of autoimmune diabetes in NOD mice. Ann. NY Acad. Sci. 1150 , 146–148 (2008).

Gulden, E. et al. Toll-like receptor 4 deficiency accelerates the development of insulin-deficient diabetes in non-obese diabetic mice. PLoS ONE 8 , e75385 (2013).

Prinz, M. et al. Innate immunity mediated by TLR9 modulates pathogenicity in an animal model of multiple sclerosis. J. Clin. Invest. 116 , 456–464 (2006).

Cohen, S. J., Cohen, I. R. & Nussbaum, G. IL-10 mediates resistance to adoptive transfer experimental autoimmune encephalomyelitis in MyD88 −/− mice. J. Immunol. 184 , 212–221 (2010).

Marta, M., Andersson, A., Isaksson, M., Kampe, O. & Lobell, A. Unexpected regulatory roles of TLR4 and TLR9 in experimental autoimmune encephalomyelitis. Eur. J. Immunol. 38 , 565–575 (2008).

Miranda-Hernandez, S. et al. Role for MyD88, TLR2 and TLR9 but not TLR1, TLR4 or TLR6 in experimental autoimmune encephalomyelitis. J. Immunol. 187 , 791–804 (2011). This is a comprehensive study that examines the impact of TLR invalidation in the development of EAE.

Reynolds, J. M. et al. Toll-like receptor 2 signaling in CD4 + T lymphocytes promotes T helper 17 responses and regulates the pathogenesis of autoimmune disease. Immunity 32 , 692–702 (2010).

Christensen, S. R. et al. Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity 25 , 417–428 (2006).

Atlas of MS 2013. https://www.msif.org/wp-content/uploads/2014/09/Atlas-of-MS.pdf

Global tuberculosis report 2016. http://apps.who.int/iris/bitstream/10665/250441/1/9789241565394-eng.pdf?ua=1

Screening for hepatitis during the domestic medical examination for newly arrived refugees. https://www.cdc.gov/immigrantrefugeehealth/pdf/domestic-hepatitis-screening-guidelines.pdf

Caisse des Français de l'Étranger. https://www.cfe.fr/pages/votre-sante/guidespatho.php?id=126 [French]

International Monetary Fund. World Economic Outlook Database. https://www.imf.org/external/pubs/ft/weo/2015/01/weodata/index.aspx

Download references

Acknowledgements

The laboratory of the author was supported by an advanced grant from the European Research Council (ERC, Hygiene N°: 250290).

Author information

Authors and affiliations.

Université Paris Descartes, Sorbonne Paris Cité, Paris, France

Jean-François Bach

INSERM U1151, Institut Necker-Enfants Malades, Hôpital Necker-Enfants Malades, Paris, France

CNRS UMR 8253, Institut Necker-Enfants Malades, Hôpital Necker-Enfants Malades, Paris, France

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Jean-François Bach .

Ethics declarations

Competing interests.

The author declares no competing financial interests.

PowerPoint slides

Powerpoint slide for fig. 1, powerpoint slide for fig. 2, powerpoint slide for table 1, powerpoint slide for table 2, powerpoint slide for table 3.

A genetic predisposition to the cumulative development of common allergies, for example, atopic dermatitis and allergic asthma. Atopy involves phenomena of cutaneous or general hypersensitivity to allergens.

A hypothesis that postulates that an increased frequency of infections contributes to a decrease in autoimmune and allergic diseases.

An inbred mouse line that spontaneously develops an autoimmune syndrome including insulin-dependent diabetes mellitus (IDDM or type 1 diabetes).

A digestive tract disorder provoked by eating contaminated food or drinking contaminated water. In the context of our discussion, it is a self-limited pathology that illustrates the presence of a basic health environment.

Autoantibodies to various β-cell-specific autoantigens that are markers of the destruction of insulin-producing β-cells, which is the hallmark of insulin-dependent diabetes mellitus (IDDM or type 1 diabetes).

An imbalance of the microbial flora that most frequently affects the digestive tract. Dysbiosis can also be detected in other 'barrier' organs such as the skin, the lungs or the vagina.

The metabolome consists of all signalling molecules (for example, metabolites and hormones) detected in a biological sample. The metabolome thus defines a given physiological or pathological state and is therefore dynamic.

Mice born by hysterectomy under sterile conditions and raised in isolators to guarantee an environment totally devoid of pathogenic and commensal germs.

(EAE). A demyelinating allergic encephalomyelitis produced by the injection of brain tissue or purified proteins of the nervous system or their derived peptides in the presence of an adjuvant.

Germ-free mice whose intestinal microflora is reconstituted by a single commensal bacterium (monocolonized mice).

Gut commensal bacteria available as single or combined species delivered orally and putatively endowed with a health benefit.

The competition for recognition of the cognate antigen for soluble factors (cytokines) driving the proliferation and differentiation of antigen-specific lymphocytes.

Islet transplants between syngeneic (genetically identical) donor and recipient individuals, which therefore does not give rise to allograft rejection. These grafts performed in diabetic non-obese diabetic mice provide a robust model to test for recurrence of the autoimmune disease.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Bach, JF. The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol 18 , 105–120 (2018). https://doi.org/10.1038/nri.2017.111

Download citation

Published : 16 October 2017

Issue Date : February 2018

DOI : https://doi.org/10.1038/nri.2017.111

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Galectin from trichinella spiralis alleviates dss-induced colitis in mice by regulating the intestinal microbiota.

  • Jianqing Li
  • Xiangjiang Wang

Veterinary Research (2024)

Functional characterization of helminth-associated Clostridiales reveals covariates of Treg differentiation

  • Shushan Sargsian
  • Octavio Mondragón-Palomino
  • Ken Cadwell

Microbiome (2024)

The burden of dermatitis from 1990–2019 in the Middle East and North Africa region

  • Saeid Safiri
  • Mehran Jaberinezhad
  • Ali-Asghar Kolahi

BMC Public Health (2024)

Efficacy and safety of gut microbiota-based therapies in autoimmune and rheumatic diseases: a systematic review and meta-analysis of 80 randomized controlled trials

  • Liuting Zeng
  • Kailin Yang
  • Lingyun Sun

BMC Medicine (2024)

Genetic and environmental interactions contribute to immune variation in rewilded mice

  • Oyebola Oyesola
  • Alexander E. Downie

Nature Immunology (2024)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

hygiene hypothesis original paper

MINI REVIEW article

The hygiene hypothesis – learning from but not living in the past.

\nPetra I. Pfefferle,,

  • 1 Comprehensive Biobank Marburg, Medical Faculty, Philipps University of Marburg, Comprehensive Biobank Marburg, Marburg, Germany
  • 2 German Center for Lung Research (DZL), Marburg, Germany
  • 3 German Biobank Alliance, Marburg, Germany
  • 4 Institute for Pathology, Medical Faculty, Institute for Pathology, Philipps University of Marburg, Marburg, Germany
  • 5 Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Biochemical Pharmacological Center, Philipps University of Marburg, Marburg, Germany

Postulated by Strachan more than 30 years ago, the Hygiene Hypothesis has undergone many revisions and adaptations. This review journeys back to the beginnings of the Hygiene Hypothesis and describes the most important landmarks in its development considering the many aspects that have refined and generalized the Hygiene Hypothesis over time. From an epidemiological perspective, the Hygiene Hypothesis advanced to a comprehensive concept expanding beyond the initial focus on allergies. The Hygiene Hypothesis comprise immunological, microbiological and evolutionary aspects. Thus, the original postulate developed into a holistic model that explains the impact of post-modern life-style on humans, who initially evolved in close proximity to a more natural environment. Focusing on diet and the microbiome as the most prominent exogenous influences we describe these discrepancies and the resulting health outcomes and point to potential solutions to reestablish the immunological homeostasis that frequently have been lost in people living in developed societies.

Last year we celebrated the 30th anniversary of the Hygiene Hypothesis. Since Strachan framed the Hygiene Hypothesis in 1989 ( 1 ) his fundamental idea to explain the origins of allergic diseases development has survived the test of time. The basic idea of how humans, their microbiota, and a continuously modernizing environment have interacted to drive immune dysregulation has persisted and become part of the popular imagination. Here, we aim to provide an editorial overview on the history of the Hygiene Hypothesis and related topics to offer a framework for the articles collected in the special edition research topic “The Hygiene Hypothesis and its Immunological Implications.”

A Chronological Overview

The epidemiological basis for the Hygiene Hypothesis became apparent long before the Hygiene Hypothesis was postulated. Two simple observations were made in the 1960s and in the 1970s. First, a Swedish study described differences in the prevalence of asthma and socio-medical conditions between populations living in urban or rural sites ( 2 ). A few years later, in a population-based study conducted in Saskatchewan, Canada, showed that allergies were less frequent in native tribes living traditionally in rural sites compared to Caucasian Canadians living in urban habitats ( 3 ). Moreover, the authors postulated that frequent bacterial infections in childhood might be responsible for the inverse association with allergic diseases. Strachan's observations made in the late 80s in a British population corroborated these findings and he later named this concept “Hygiene Hypothesis” in 2000. Briefly, Strachan suggested that transfer of early childhood infections between siblings is associated with protection against allergies later in life ( 4 ).

The hypothesis was further substantiated and extended by studies that compared asthma and allergy prevalence directly after the “Fall of the Iron Curtain” between Western and Eastern Germany, a decade later ( 5 , 6 ). Interestingly, these studies triggered a paradigm shift in allergy research. Until then, environmental pollution was broadly regarded as the leading force for allergy development. Environmental data clearly indicated a higher level of pollution by industrial emissions in Eastern Germany compared to the Western part and the study team therefore hypothesized that the prevalence of allergic diseases was higher in children from Eastern Germany. Surprisingly, the researchers found their hypothesis disproved, as children in Western Germany showed a higher prevalence of allergies. Hence, it was postulated that other exposures than pollutants influence the development of atopic diseases. Socio-demographic and –economic factors, as well as household hygiene turned out to be further discriminatory factors between both parts of German population. Improved sanitation and hygiene were positively associated with atopic diseases. Another decade later a follow-up further validated this hypothesis and found life-style differences and the prevalence of atopic diseases began to equilibrate within 10 years after the reunification. In consequence, the Hygiene Hypothesis became the leading postulate to explain underlying relationships and mechanisms for the development of allergic diseases in a societal context ( 6 ).

Based on this paradigm shift, Rook published the “Old Friends-Hypothesis” which argues that infectious diseases have a long co-evolutionary history with human development, and appropriate levels of exposure to these microorganisms early in life might protect against immune deviation and allergic diseases. These early-life exposures to potential pathogens might educate the developing immune system from a type-2-dominated in utero -milieu toward a more defensive T helper (h)1 response ( 7 ).

The next milestone involved findings obtained from the so-called “Alpine farm studies” conducted at the turn of the millennium. Von Mutius and Braun-Fahrländer recognized the unique situation that the Alpine traditional farming environment represents a socio-cultural and ecological niche which significantly differs from the post-modern and urbanized life-style. In a number of epidemiological studies they identified traditional farming characteristics such as consumption of unprocessed farm milk and close contact with farm animals to act allergoprotective and found these parameters to be associated with a higher microbial load. These Alpine farm studies added substantial evidence to Strachan's basic idea and led to a broader view and understanding of the relationship between human health and (early life) exposure to microbes ( 8 , 9 ).

Further evidence was added by studies conducted in Northern Europe. In the late 1990s studies conducted in Scandinavian und Baltic children described microbial factors to be associated with a lower prevalence of allergic diseases in the Eastern countries ( 10 – 12 ). Next, the Karelia Study, conducted on both sides of the Finnish-Russian border, addressed the impact of the environmental microbial burden on the development of allergic diseases in Finnish and Russian Karelian children that share the same ethnic background but have different life-styles ( 13 ). These studies corroborated the Alpine farm studies and point to the microbial environment as a major factor in allergy development.

Furthermore, these studies demonstrate that the diversity and the richness of an immune-stimulating microbial world in human habitats is crucial to establish a competent, tolerogenic and defensive immune system configuration while absence or depletion of those stimuli as found in post-modern environments foster immune deviation and development of allergic diseases ( 14 ).

Moreover, two relevant studies [the cross-sectional study “Prevention of Allergy Risk factors for Sensitization In children related to Farming and Anthroposophic Lifestyle (PARSIFAL)” and the multi-center, pregnancy/birth cohort study “Protection against Allergy: Study in Rural Environments (PASTURE)”] support the idea that the “window of opportunity” in which the appropriate education of the immune system starts already in the mother's womb ( 15 – 17 ). The PARSIFAL Study demonstrated that maternal exposure to a farm environment rich in microbial compounds is inversely associated with the development of atopic sensitization and correlated with an upregulation of receptors of the innate immune system in the offspring at school age ( 15 ). Further, maternal farm activities during pregnancy were shown to modulate cord blood cytokines and allergen-specific immunoglobulin responses toward a Th1 pattern ( 16 , 17 ). These findings are in line with the Barker theory ( 18 ), postulating that pathological pathways occurring in adolescence and adulthood are paved already in prenatal life.

The Hygiene Hypothesis and the Bacterial World

Even before high-throughput sequencing techniques were established that allow a deeper view into the microbial world on our body surfaces, Noverr and Hufnagle proclaimed the “Microbiota Hypothesis” by which they claimed the microbiota to be indispensable for developing and maintaining a tolerogenic immune status ( 19 ). A similar idea concept was proposed earlier by Holt, Sly and Björkstén ( 20 ). The rediscovery of the microbiota and its powerful metabolic and immunologic interplay with the mucosal surfaces of the host underlined and complemented the principals of this basic idea ( 21 , 22 ). Microbiome research has made significant achievements over the past 15 years; here we can emphasize only a few aspects that might be relevant in the context of the Hygiene Hypothesis and the development of allergies.

Phylogenetic Impacts

An intriguing concept to better understand the complex symbiotic interplay at organ surfaces was suggested by McFall-Ngai in 2007. In her evolutionary perspective she shed light on findings made in invertebrates which not only lack an endoskeleton but also an adaptive immune system. Thus, invertebrates have to exclusively rely on their innate immune system, which to our current understanding, lacks an immunological memory. Analyses of the intestinal microbiota in such animals have shown—in contrast to vertebrates—a rather low diversity in the community of their microbial residents. Only a handful of strains could be identified as stable colonizers on the gastrointestinal surfaces while most bacteria travel through as transient visitors. Some invertebrates, like insects, separate bacterial colonies from epithelial host cells by a peritrophic matrix composed of chitin and other compounds ( 24 ). During the course of evolution, the microbial colonization of epithelia started to get more complex and in turn the host was challenged to develop new strategies to manage these diversifying communities. To permanently recognize a specific bacterium as beneficial or harmful, an adaptive immune response that provides an immunological memory over generations of immune cells was needed. Mutual adaption of both partners, the bacterial community, as well as the complex network of adaptive immune cells, led to a sophisticated metabolic and immunologic interplay with a highly digestive and defensive performance. This symbiosis is based on early education of the host's immune cells by a diverse microbial community to successfully discriminate dangerous pathogens from beneficial symbionts and own healthy cells. Finally McFall-Ngai stated, that complex systems might be prone to failure and allergies and autoimmune disorders might be a consequence of this ( 23 ).

Ontogenetic Impacts

A number of recently published reports substantiated the impact of the early life microbiota on immune maturation [recently reviewed in ( 25 )] and the development of allergic disorders in early infancy [recently reviewed in ( 26 )]. The developmental starting point of the infant gut microbiota is still unknown, but undoubtedly, the process of delivery seems to be a key point in the development of the neonatal microbiota ( 27 ). Meconium, the neonate's first intestinal discharge, was shown to contain various bacterial strains indicating that the perinatal gut is colonized by bacteria ( 28 , 29 ). In a landmark study, Dominguez-Bello et al. reported that the neonatal microbiota differs between vaginally born infants and neonates delivered by Caesarian (C)-section. The authors found a high abundance of Bacteroides, Bifidobacterium , and Lactobacillus spec . in meconium samples obtained from vaginally delivered newborns, while Staphylococcus, Streptococcus, Corynebacterium , and Propionibacterium spp . were found predominantly in meconium samples of C-section born neonates ( 30 ).

Colonization of the neonate's colon by Lactobacilli and Bifidobacteria transferred from the maternal vaginal compartment during vaginal passage might provide advantages for the newborn due to the metabolic properties of these bacteria that foster the adaptation to milk-based feeding. These bacteria are capable of metabolizing breast milk-derived lactose and human milk oligosaccharides (HMOS) ( 31 ) and were shown to provide immune-modulating short chain fatty acids (SCFAs) ( 32 ) and conjugated trans-linoleic acids (tCLAs) ( 33 ), which are shown to reduce pro-inflammatory eicosanoid production by regulating the transcription of cyclooxygenase 2 (COX-2) ( 34 ) and to induce anti-inflammatory M2-macrophage differentiation ( 35 ).

However, how sustainable and decisive are these mode of delivery-associated differences beyond the neonatal age? Chu et al. recently showed that function and composition of the microbiota significantly diversifies in all body sites within the first 6 weeks of life, resembling the corresponding maternal body site microbiota at this time point. Infant's mode of delivery or other prenatal factors seems to have no impact on this development ( 36 ). Data from the Copenhagen Prospective Studies on Asthma in Childhood 2010 (COPSAC 2010 ) cohort underlined the importance of the maturation of the microbiota on the further development of the gut microbiome and the risk of asthma later in life. In that study, Stockholm et al. compared the gut microbiome of vaginally and C-section delivered infants from birth to 1 year of life in the context of asthma development at school age. Marked differences between C-section and vaginally delivered infants were observed by 1 week and by 1 month of life, but only minor differences between these groups were found by 1 year of age. An increased risk for school-age asthma was only observed in a subgroup of C-section-born infants that maintained the C-section-associated composition for at least 1 year. The authors conclude that vaginal delivery and/or subsequent maturation of the infant microbiota might support a more robust and stable microbiota in the offspring that is more adaptive to the challenges later in life ( 37 ). Further exposure to the maternal microbiota ( 38 ), as well as nutritional impacts (e.g., cessation of breastfeeding) ( 39 ) within the first month of life, might foster the maturation of the gut microbiome in early infancy.

Nutritional Impacts

How is the microbiota linked to the rising atopic epidemic observed in the recent decades? A recently published study conducted in indigenous tribes living in the Brazilian Amazonas-Orinoco Basin may help to answer this question ( 40 ). In this study the gut microbiome of the semi-nomadic gatherer/hunter people of the Yanomami who maintained a primitive close-to-nature life-style was compared to subjects representing populations that are characterized by a westernized or non-ancestral life-style in rural and urban settings. The Yanomami microbiota was significantly more diverse than those of the westernized counterparts. Moreover, an additional study comparing Venezuelan with Brazilian Yanomami indicated a high level of adaptability to specific environmental conditions of the microbiota in these peoples. While a high taxonomic diversity was found in both sub-tribes, the composition of microbiota was significantly different ( 41 ). These findings point to environmental and life-style factors that influence the composition of the microbiota the absence of which may thus foster the loss of taxonomic and metabolic diversity in westernized societies ( 42 ).

Diet is one of the most prominent environmental factors that differ between modern and ancient life-styles. While dietary habits in indigenous people such as the Yanomami strongly depend on the sometimes limited food supply due to seasonal cycling, people living in developed societies have access to high in calories food ready at any time and in abundance. Moreover, diet in indigenous cultures is often based on high-fiber products derived from plants that are easy to culture such as plantain, manioc or sweet potatoes, all rich in inulin ( 43 ). High-fiber diet and, in particular inulin, is known as an effective enhancer of beneficial bacteria such as Bifidobacteria in the colon that stabilize gut homeostasis ( 44 ). Translating these findings into a clinical approach, McLoughlin et al. applied soluble inulin to asthmatics in a short-term placebo-controlled-trial and could report an array of beneficial effects in patients orally treated with inulin. In comparison to the placebo group, inulin-treated patients displayed a significantly reduced number of eosinophils in the sputum and, overall, reported a significantly improved asthma control. Inhibition of histone deacetylase 9 (HDAC9) in sputum cells upon a combined application of inulin and a multi-strain probiotic mixture of Lactobacillus acidophilus, Lactobacillus rhamnosus GG and Bifidobacterium animalis subspecies lactis indicated that epigenetic pathways are involved in the mechanisms by which lactic acid bacteria modulate host responses in combination with the prebiotic gavage ( 45 ).

A number of recently recognized metabolites released by beneficial symbiotic bacteria convey immunomodulatory effects, mainly in the gut but also on other mucosal surfaces ( 46 ).

In particular, SCFAs derived from dietary fibers and released in the lumen of the colon contribute to immune modulation and inhibition of pro-inflammatory cytokines when absorbed by gut epithelial cells ( 47 ). By binding to chemoattractant G protein 43 receptor, SCFAs are capable of regulating inflammatory responses ( 48 ) as shown for intestinal inflammation ( 49 ). Tryptophan, an amino acid produced by an array of beneficial microorganisms, is degraded to indole derivatives which may bind to the aryl hydrocarbon receptor (AHR) and by this regulate the activity of immune cells at the epithelial barrier. That involves AHR-dependent differentiation of regulatory T cells associated with anti-inflammatory IL-10 expression. Further, Th2-cells are inhibited on the transcription factor level in favor of a Th1 response ( 50 ).

A number of beneficial bacteria contribute to the orchestration of T cell subsets at the gut epithelial barrier. Bacteroides sp . and Clostridium clusters IV and XIVa colonizing the gut epithelium are known to stimulate intestinal epithelial cells to release thymic stromal lymphopoietin (TSLP), transforming growth factor (TGF)-ß and interleukin (IL)-25 which in combination may induce tolerogenic effects in dendritic cells (DCs) ( 51 ), e.g., by secretion of TGF-ß and retinoic acid. Both factors initiate differentiation of naïve T cells to regulatory T cells upon activation of the nuclear transcription factor forkhead box P (FoxP3) ( 52 ). These regulatory mechanisms are challenged by “pathobionts” or other damage factors. In presence of these stressors, overexpansion of Th1, Th2 and Th17 effector cell subsets might result in an inflammatory response in the infected organ or, by migration of these cells, at distant sites. Namely, Clostridium difficile , which is associated with wheezing and atopic sensitization, was shown to initially disturb the intestinal balance when acquired early in childhood ( 53 ).

Traveling from the gastrointestinal to the respiratory tract the microbiota established in the lung might also play a role in the development of allergic disorders, namely of allergic asthma. Though the gut is known to play a major role in establishing and regulating immune defense mechanisms, the “gut-lung axis” alone might not completely explain the rise of allergic asthma ( 54 ). As many studies focused on the lung microbiome, it has become clear that there is a strong relationship between frequently inhaled environmental microbes, microbial colonization of the respiratory tract, and the prevalence of allergic asthma ( 55 ). For example, results from the “Multidisciplinary Study to Identify the Genetic and Environmental Causes of Asthma in the European Community (GABRIEL) Advanced Studies (GABRIELA)” study suggested a transfer of built-environment-associated bacteria into the respiratory tract. Indoor dust samples from farm houses and nasal swabs from farm children displayed a higher bacterial diversity than those samples collected in rural non-farm children ( 56 ). New evidence was added recently by studies conducted in the Finnish part of the PASTURE-study. Kirjavainen et al. reported that the ecological diversity of the so-called “indoor microbiota” is inversely linked to the prevalence of allergic asthma. Substantiating former farm studies, this report further validated the hypothesis that microbial diversity and composition in the natural environment is linked to a reduced risk of early-onset allergic asthma and that traditional farming is a proxy for this effect ( 57 ).

But what are the cellular and molecular mechanisms associated with high microbial diversity? Interestingly, the farm studies consistently showed an inverse association between a highly diverse environmental microbiota and allergic asthma, but this did not account to other allergic manifestations such as hay fever or atopic sensitization. On the other hand, endotoxin exposure protects against atopy but fosters the risk of non-allergic asthma and early onset of wheeze when inhaled in higher concentrations. These findings derived from the farm studies still challenge the Hygiene Hypothesis and might point out that microbial colonization and exposure to microbial compounds have to be considered separately ( 58 ). Integration of beneficial environmental bacteria into the microbial community of the respiratory tract leads to a tolerogenic mucosal symbiosis that establishes a local T-cell balanced anti-inflammatory milieu at the epithelium, probably enhanced by a well-balanced gut microbiota. Endotoxins are potent activators of innate TLR-signaling and can attenuate B cell driven sensitization and formation of IgE-antibodies ( 59 ). Already in 2003, Vercelli postulated a switch from Th2-driven allergic responses at low endotoxin exposure to a pronounced Th1 response in the lung under high levels of environmental endotoxin. This might explain the elevated prevalence of non-allergic asthma in environments overloaded with endotoxin ( 60 ).

Conclusions

The many aspects and facets of the Hygiene Hypothesis have been supported by concepts and findings coming from a variety of scientific disciplines such as epidemiology, immunology, microbiology and anthropology. Within the last three decades we obtained a multiplicity of new insights into the complexity and plasticity of T cell networks which led us to recognize the complexity and significance of a powerful and well-regulated adaptive immune response in relation to exogenous factors ( 61 ). Early developmental findings characterizing pre and postnatal life events highlighted the initial role of the innate immune system as an early warning system that orchestrates, educates and shapes subsequent immune responses ( 62 , 63 ). Evidence from evolutionary biology and anthropology enabled us to understand how host-environment interactions are refined throughout evolutionary adaption ( 58 , 64 ). Microbiology added fundamental knowledge about the micro-ecosystem that is established throughout the human body as a unique symbiosis between humans and microbes. And finally, coming back to the introductory statement, epidemiological observations such as those initially made by Strachan and von Mutius about 30 years ago still challenge and refine the hypothesis.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

This work was funded by the Library of the Philipps-University of Marburg.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

1. Strachan DP. Hay fever, hygiene, and household size. BMJ. (1989) 299:1259–60. doi: 10.1136/bmj.299.6710.1259

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Irnell L, Kiviloog J. Bronchial asthma and chronic bronchitis in a Swedish urban and rural population. With special reference to prevalence, respiratory function and socio-medical condition. Scand J Respir Dis Suppl. (1968) 66:1–86.

PubMed Abstract | Google Scholar

3. Gerrard J. Serum IgE levels in white and metis communities in saskatchewan. Ann Allergy. (1976) 37:91–100.

4. Strachan DP. Family size, infection and atopy: the first decade of the “Hygiene Hypothesis”. Thorax. (2000) 55(Suppl. 1):S2–10. doi: 10.1136/thorax.55.suppl_1.S2

5. Weiland SK, von Mutius E, Hirsch T, Duhme H, Fritzsch C, Werner B, et al. Prevalence of respiratory and atopic disorders among children in the East and West of Germany five years after unification. Eur Respir J. (1999) 14:862–70. doi: 10.1034/j.1399-3003.1999.14d23.x

6. Heinrich J, Hoelscher B, Frye C, Meyer I, Wjst M, Wichmann HE. Trends in prevalence of atopic diseases and allergic sensitization in children in Eastern Germany. Eur Respir J. (2002) 19:1040–6. doi: 10.1183/09031936.02.00261802

7. Rook GA, Brunet LR. Microbes, immunoregulation, and the gut. Gut. (2005) 54:317–20. doi: 10.1136/gut.2004.053785

CrossRef Full Text | Google Scholar

8. Riedler J, Braun-Fahrländer C, Eder W, Schreuer M, Waser M, Maisch S, et al. Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet. (2001) 358:1129–33. doi: 10.1016/S0140-6736(01)06252-3

9. Braun-Fahrländer C, Gassner M, Grize L, Neu U, Sennhauser FH, Varonier HS, et al. Prevalence of hay fever and allergic sensitization in farmer's children and their peers living in the same rural community. SCARPOL team. swiss study on childhood allergy and respiratory symptoms with respect to air pollution. Clin Exp Allergy . (1999) 29:28–34. doi: 10.1046/j.1365-2222.1999.00479.x

10. Björkstén B, Dumitrascu D, Foucard T, Khetsuriani N, Khaitov R, Leja M, et al. Prevalence of childhood asthma, rhinitis and eczema in Scandinavia and Eastern Europe. Eur Resp J. (1998) 12:432–7. doi: 10.1183/09031936.98.12020432

11. Bråbäck L, Breborowicz A, Julge K, Knutsson A, Riikjärv M, Vasar M, et al. Risk factors for respiratory symptoms and atopic sensitization in the Baltic area. Arch Dis Child. (1995) 72:487–93. doi: 10.1136/adc.72.6.487

12. Björkstén B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic estonian and swedish 2-year old children. Clin Exper Allergy . (1999) 29:342–6. doi: 10.1046/j.1365-2222.1999.00560.x

13. Seiskari T, Kondrashova A, Viskari H, Kaila M, Haapala AM, Aittoniemi J, et al. Allergic sensitization and microbial load–a comparison between Finland and Russian Karelia. Clin Exp Immunol. (2007) 148:47–52. doi: 10.1111/j.1365-2249.2007.03333.x

14. Ege MJ, Mayer M, Normand AC, Genuneit J, Cookson WO, Braun-Fahrländer C, et al. Exposure to environmental microorganisms and childhood asthma. N Engl J Med. (2011) 364:701–9. doi: 10.1056/NEJMoa1007302

15. Ege MJ, Bieli C, Frei R, van Strien RT, Riedler J, Ublagger E, et al. Prenatal farm exposure is related to the expression of receptors of the innate immunity and to atopic sensitization in school-age children. J Allergy Clin Immunol. (2006) 117:817–23. doi: 10.1016/j.jaci.2005.12.1307

16. Pfefferle PI, Sel S, Ege MJ, Büchele G, Blümer N, Krauss-Etschmann S, et al. Cord blood allergen-specific IgE is associated with reduced IFN-gamma production by cord blood cells: the protection against allergy-study in rural environments (pasture) study. J Allergy Clin Immunol. (2008) 122:711–16. doi: 10.1016/j.jaci.2008.06.035

17. Pfefferle PI, Büchele G, Blümer N, Roponen M, Ege MJ, Krauss-Etschmann S, et al. Cord blood cytokines are modulated by maternal farming activities and consumption of farm dairy products during pregnancy: the PASTURE study. J Allergy Clin Immunol. (2010) 125:108–15.e1-3. doi: 10.1016/j.jaci.2009.09.019

18. Barker DJ. The fetal and infant origins of disease. Eur J Clin Invest. (1995) 25:457–63. doi: 10.1111/j.1365-2362.1995.tb01730.x

19. Noverr MC, Huffnagle GB. The ‘microflora hypothesis' of allergic diseases. Clin Exp Allergy. (2005) 35:1511–20. doi: 10.1111/j.1365-2222.2005.02379.x

20. Holt P, Sly P, Björkstén B. Atopic versus infectious disease in childhood: a question of balance? Pediatr Allergy Immunol. (1997) 8:1–5 doi: 10.1111/j.1399-3038.1997.tb00145.x

21. Fyhrquist N. The human microbiota and its relationship with allergies. Gastroenterol Clin North Am. (2019) 48:377–87. doi: 10.1016/j.gtc.2019.04.005

22. von Mutius E. A fascinating look at the world with a new microscope. J Allergy Clin Immunol. (2012) 129:1202–3. doi: 10.1016/j.jaci.2011.12.994

23. McFall-Ngai M. Adaptive immunity: care for the community. Nature. (2007) 445:153. doi: 10.1038/445153a

24. Wong ACN, Vanhove AS, Watnick PI. The interplay between intestinal bacteria and host metabolism in health and disease: lessons from drosophila melanogaster. Dis Model Mech. (2016) 9:271–81. doi: 10.1242/dmm.023408

25. Al Nabhani Z, Eberl G. Imprinting of the immune system by the microbiota early in life. Mucosal Immunol. (2020) 13:183–18. doi: 10.1038/s41385-020-0257-y

26. Peroni DG, Nuzzi G, Trambusti I, Di Cicco ME, Comberiati P. Microbiome composition and its impact on the development of allergic diseases. Front Immunol. (2020) 11:700. doi: 10.3389/fimmu.2020.00700

27. Mesa MD, Loureiro B, Iglesia I, Fernandez Gonzalez S, Llurba Olivé E, García Algar O, et al. The evolving microbiome from pregnancy to early infancy: a comprehensive review. Nutrients. (2020) 12:133–54. doi: 10.3390/nu12010133

28. Jimenez E, Marín ML, Martín R, Odriozola JM, Olivares M, Xaus J, et al. Is meconium from healthy newborns actually sterile? Res Microbiol. (2008) 159:187–93. doi: 10.1016/j.resmic.2007.12.007

29. Hansen R, Scott KP, Khan S, Martin JC, Berry SH, Stevenson M. First-pass meconium samples from healthy term vaginally-delivered neonates: an analysis of the microbiota. PLoS ONE. (2015) 10:e0133320. doi: 10.1371/journal.pone.0133320

30. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. (2010) 107:11971–75. doi: 10.1073/pnas.1002601107

31. Plaza-Díaz J, Fontana L, Gil A. Human milk oligosaccharides and immune system development. Nutrients. (2018) 10:1038–55. doi: 10.3390/nu10081038

32. Rivière A, Selak M, Lantin D. Butyrate-producing colon bacteria: importance and strategies for their stimulation in the human gut. Front Microbiol. (2016) 7:979–80. doi: 10.3389/fmicb.2016.00979

33. Kishino S, Takeuchi M, Park SB, Hirata A, Kitamura N, Kunisawa J. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci USA. (2013) 110:17808–13. doi: 10.1073/pnas.1312937110

34. Ringseis R, Müller A, Herter C, Gahler S, Steinhart H, Eder K, et al. CLA isomers inhibit TNFalpha-induced eicosanoid release from human vascular smooth muscle cells via a PPARgamma ligand-like action. Biochim Biophys Acta. (2006) 1760:290–300. doi: 10.1016/j.bbagen.2005.12.002

35. Ohue-Kitano R, Yasuoka Y, Goto T, Kitamura N, Park SB, Kishino S, et al. α-Linolenic acid-derived metabolites from gut lactic acid bacteria induce differentiation of anti-inflammatory M2 macrophages through G protein-coupled receptor 40. FASEB J. (2018) 32:304–18. doi: 10.1096/fj.201700273R

36. Chu DM, Ma J, Prince A, Antony KM, Sefrovic MD, Aagaard KM. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med. (2017) 23:314–26. doi: 10.1038/nm.4272

37. Stokholm J, Thorsen J, Blaser MJ, Rasmussen MA, Hjelmsø M, Shah S, et al. Delivery mode and gut microbial changes correlate with an increased risk of childhood asthma. Sci Transl Med. (2020) 12:eaax9929. doi: 10.1126/scitranslmed.aax9929

38. Mortensen MS, Rasmussen MA, Stokholm J, Brejnrod AD, Balle C, Thorsen J, et al. Modeling transfer of vaginal microbiota from mother to infant in early life. Elife. (2021) 10:e57051. doi: 10.7554/eLife.57051

39. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. (2015) 17:690–703. doi: 10.1016/j.chom.2015.04.004

40. Clemente JC, Pehrsson EC, Blaser MJ, Sandhu K, Gao Z, Wang B, et al. The microbiome of uncontacted amerindians Sci Adv . (2015) 1:e1500183. doi: 10.1126/sciadv.1500183

41. Conteville LC, Oliveira-Ferreira J, Vicente ACP. Gut microbiome biomarkers and functional diversity within an amazonian semi-nomadic hunter-gatherer group. Front Microbiol. (2019) 10:1743. doi: 10.3389/fmicb.2019.01743

42. Moeller AH. The shrinking human gut microbiome. Curr Opin Microbiol. (2017) 38:30–5. doi: 10.1016/j.mib.2017.04.002

43. Lizot J. Resources and warfare among the Yanomami. Man New Ser. (1977) 12:497–17. doi: 10.2307/2800552

44. Gibson GR, Beatty ER, Wang X, Cummings JH. Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology. (1995) 108:975–82. doi: 10.1016/0016-5085(95)90192-2

45. McLoughlin R, Berthon BS, Rogers GB, Baines KJ, Lex EX, Leong LEX, et al. Soluble fibre supplementation with and without a probiotic in adults with asthma: a 7-day randomised, double blind, three way cross-over trial. EBioMedicine. (2019) 46:473–85. doi: 10.1016/j.ebiom.2019.07.048

46. O'Keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol. (2016) 13:691–706. doi: 10.1038/nrgastro.2016.165

47. Smith PM, Howitt MR, Panikoc N, Michaud M, Gallini CA, Bohlooly-Y M, et al. The microbial metabolites, short-chain fatty acids, regulate colonic treg cell homeostasis. Science. (2013) 341 569–73. doi: 10.1126/science.1241165

48. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. (2009) 461:1282–86. doi: 10.1038/nature08530

49. Sina C, Gavrilova O, Förster M, Till A, Derer S, Hildebrand F, et al. G protein- coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation. J. Immunol. (2009) 183:7514–22. doi: 10.4049/jimmunol.0900063

50. Hao N, Whitelaw ML. The emerging roles of AhR in physiology and immunity. Biochem Pharmacol. (2013) 86:561–70. doi: 10.1016/j.bcp.2013.07.004

51. Brown EM, Arrieta MC, Finlay BB. A fresh look at the hygiene hypothesis: how intestinal microbial exposure drives immune effector responses in atopic disease. Semin Immunol. (2013) 25:378–87. doi: 10.1016/j.smim.2013.09.003

52. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. (2011) 331:337–4. doi: 10.1126/science.1198469

53. van Nimwegen FA, Penders J, Stobberingh EE, Postma DS, Koppelman GH, Kerkhof M, et al. Mode and place of delivery, gastrointestinal microbiota, and their influence on asthma and atopy. J Allergy Clin Immunol. (2011) 128:948–55. doi: 10.1016/j.jaci.2011.07.027

54. Marsland BJ, Trompette A, Gollwitzer ES. The gut-lung axis in respiratory disease. Ann Am Thorac Soc. (2015) 12(Suppl. 2):S150–6. doi: 10.1513/AnnalsATS.201503-133AW

55. Fabbrizzi A, Amedei A, Lavorini F, Renda T, Fontana G. The lung microbiome: clinical and therapeutic implications. Intern Emerg Med. (2019) 14:1241–50. doi: 10.1007/s11739-019-02208-y

56. Birzele LT, Depner M, Ege MJ, Engel M, Kublik S, Bernau C, et al. Environmental and mucosal microbiota and their role in childhood asthma. Allergy. (2017) 72:109–19. doi: 10.1111/all.13002

57. Kirjavainen PV, Karvonen AM, Adams RI, Täubel M, Roponen M, Tuoresmäki P, et al. Farm-like indoor microbiota in non-farm homes protects children from asthma development. Nat Med. (2019) 25:1089–95. doi: 10.1038/s41591-019-0469-4

58. Ege MJ. The hygiene hypothesis in the age of the microbiome. Ann Am Thorac Soc. (2017) 14(Suppl. S3):48–53. doi: 10.1513/AnnalsATS.201702-139AW

59. Vatanen T, Kostic AD, d'Hennezel E, Siljander H, Franzosa EA, Yassour M, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. (2016) 165:1551. doi: 10.1016/j.cell.2016.05.056

60. Vercelli D. Learning from discrepancies: CD14 polymorphisms, atopy and the endotoxin switch. Clin Exp Allergy. (2003) 33:153–5. doi: 10.1046/j.1365-2222.2003.01606.x

61. Haspeslagh E, Heyndrickx I, Hammad H, Lambrecht BN. The hygiene hypothesis: immunological mechanisms of airway tolerance. Curr Opin Immunol. (2018) 54:102–8. doi: 10.1016/j.coi.2018.06.007

62. Conrad ML, Ferstl R, Teich R, Brand S, Blümer N, Yildirim AO, et al. Maternal TLR signaling is required for prenatal asthma protection by the nonpathogenic microbe acinetobacter lwoffii F78. J Exp Med. (2009) 206:2869–77. doi: 10.1084/jem.20090845

63. Pivniouk V, Gimenes Junior JA, Honeker LK, Vercelli D. The role of innate immunity in asthma development and protection: lessons from the environment. Clin Exp Allergy. (2020) 50:282–90. doi: 10.1111/cea.13508

64. Parker W, Ollerton J. Evolutionary biology and anthropology suggest biome reconstitution as a necessary approach toward dealing with immune disorders. Evol Med Public Health. (2013) 2013:89–103. doi: 10.1093/emph/eot008

Keywords: hygiene hypothesis, allergy, asthma, immune tolerance, T cell-response, microbiome

Citation: Pfefferle PI, Keber CU, Cohen RM and Garn H (2021) The Hygiene Hypothesis – Learning From but Not Living in the Past. Front. Immunol. 12:635935. doi: 10.3389/fimmu.2021.635935

Received: 30 November 2020; Accepted: 17 February 2021; Published: 16 March 2021.

Reviewed by:

Copyright © 2021 Pfefferle, Keber, Cohen and Garn. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) . The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Petra I. Pfefferle, petraina.pfefferle@uni-marburg.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

The Hygiene Hypothesis - Learning From but Not Living in the Past

Affiliations.

  • 1 Comprehensive Biobank Marburg, Medical Faculty, Philipps University of Marburg, Comprehensive Biobank Marburg, Marburg, Germany.
  • 2 German Center for Lung Research (DZL), Marburg, Germany.
  • 3 German Biobank Alliance, Marburg, Germany.
  • 4 Institute for Pathology, Medical Faculty, Institute for Pathology, Philipps University of Marburg, Marburg, Germany.
  • 5 Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Biochemical Pharmacological Center, Philipps University of Marburg, Marburg, Germany.
  • PMID: 33796103
  • PMCID: PMC8007786
  • DOI: 10.3389/fimmu.2021.635935

Postulated by Strachan more than 30 years ago, the Hygiene Hypothesis has undergone many revisions and adaptations. This review journeys back to the beginnings of the Hygiene Hypothesis and describes the most important landmarks in its development considering the many aspects that have refined and generalized the Hygiene Hypothesis over time. From an epidemiological perspective, the Hygiene Hypothesis advanced to a comprehensive concept expanding beyond the initial focus on allergies. The Hygiene Hypothesis comprise immunological, microbiological and evolutionary aspects. Thus, the original postulate developed into a holistic model that explains the impact of post-modern life-style on humans, who initially evolved in close proximity to a more natural environment. Focusing on diet and the microbiome as the most prominent exogenous influences we describe these discrepancies and the resulting health outcomes and point to potential solutions to reestablish the immunological homeostasis that frequently have been lost in people living in developed societies.

Keywords: T cell-response; allergy; asthma; hygiene hypothesis; immune tolerance; microbiome.

Copyright © 2021 Pfefferle, Keber, Cohen and Garn.

PubMed Disclaimer

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Similar articles

  • Microbial ambassadors against food allergies. Eisenstein M. Eisenstein M. Nature. 2020 Dec;588(7836):S11-S13. doi: 10.1038/d41586-020-02781-9. Nature. 2020. PMID: 33268856 No abstract available.
  • AllergoOncology: Microbiota in allergy and cancer-A European Academy for Allergy and Clinical Immunology position paper. Untersmayr E, Bax HJ, Bergmann C, Bianchini R, Cozen W, Gould HJ, Hartmann K, Josephs DH, Levi-Schaffer F, Penichet ML, O'Mahony L, Poli A, Redegeld FA, Roth-Walter F, Turner MC, Vangelista L, Karagiannis SN, Jensen-Jarolim E. Untersmayr E, et al. Allergy. 2019 Jun;74(6):1037-1051. doi: 10.1111/all.13718. Epub 2019 Mar 6. Allergy. 2019. PMID: 30636005 Free PMC article.
  • Revisiting the Hygiene Hypothesis in the Context of Autoimmunity. Bach JF. Bach JF. Front Immunol. 2021 Jan 28;11:615192. doi: 10.3389/fimmu.2020.615192. eCollection 2020. Front Immunol. 2021. PMID: 33584703 Free PMC article. Review.
  • Revisiting the hygiene hypothesis for allergy and asthma. Liu AH. Liu AH. J Allergy Clin Immunol. 2015 Oct;136(4):860-5. doi: 10.1016/j.jaci.2015.08.012. J Allergy Clin Immunol. 2015. PMID: 26449798
  • Hygiene Hypothesis in Asthma Development: Is Hygiene to Blame? van Tilburg Bernardes E, Arrieta MC. van Tilburg Bernardes E, et al. Arch Med Res. 2017 Nov;48(8):717-726. doi: 10.1016/j.arcmed.2017.11.009. Epub 2017 Dec 7. Arch Med Res. 2017. PMID: 29224909 Review.
  • Presence of sputum IgG against eosinophilic inflammatory proteins in asthma. Qin R, Long F, Zhang P, Huang R, Hu H, Guo Y, Zheng Z, Xiao J, He L, Peng T, Li J. Qin R, et al. Front Immunol. 2024 Jul 18;15:1423764. doi: 10.3389/fimmu.2024.1423764. eCollection 2024. Front Immunol. 2024. PMID: 39091502 Free PMC article.
  • The Early Appearance of Asthma and Its Relationship with Gut Microbiota: A Narrative Review. Suárez-Martínez C, Santaella-Pascual M, Yagüe-Guirao G, García-Marcos L, Ros G, Martínez-Graciá C. Suárez-Martínez C, et al. Microorganisms. 2024 Jul 19;12(7):1471. doi: 10.3390/microorganisms12071471. Microorganisms. 2024. PMID: 39065238 Free PMC article. Review.
  • The Prevention of House Dust Mite Allergies in Pediatric Asthma. Klain A, Senatore AA, Licari A, Galletta F, Bettini I, Tomei L, Manti S, Mori F, Miraglia Del Giudice M, Indolfi C. Klain A, et al. Children (Basel). 2024 Apr 15;11(4):469. doi: 10.3390/children11040469. Children (Basel). 2024. PMID: 38671686 Free PMC article. Review.
  • Airways Type-2 Related Disorders: Multiorgan, Systemic or Syndemic Disease? Giombi F, Pace GM, Pirola F, Cerasuolo M, Ferreli F, Mercante G, Spriano G, Canonica GW, Heffler E, Ferri S, Puggioni F, Paoletti G, Malvezzi L. Giombi F, et al. Int J Mol Sci. 2024 Jan 5;25(2):730. doi: 10.3390/ijms25020730. Int J Mol Sci. 2024. PMID: 38255804 Free PMC article. Review.
  • Alternatives to antibiotics in pig production: looking through the lens of immunophysiology. Liu HY, Zhu C, Zhu M, Yuan L, Li S, Gu F, Hu P, Chen S, Cai D. Liu HY, et al. Stress Biol. 2024 Jan 2;4(1):1. doi: 10.1007/s44154-023-00134-w. Stress Biol. 2024. PMID: 38163818 Free PMC article. Review.
  • Strachan DP. Hay fever, hygiene, and household size. BMJ. (1989) 299:1259–60. 10.1136/bmj.299.6710.1259 - DOI - PMC - PubMed
  • Irnell L, Kiviloog J. Bronchial asthma and chronic bronchitis in a Swedish urban and rural population. With special reference to prevalence, respiratory function and socio-medical condition. Scand J Respir Dis Suppl. (1968) 66:1–86. - PubMed
  • Gerrard J. Serum IgE levels in white and metis communities in saskatchewan. Ann Allergy. (1976) 37:91–100. - PubMed
  • Strachan DP. Family size, infection and atopy: the first decade of the “Hygiene Hypothesis”. Thorax. (2000) 55(Suppl. 1):S2–10. 10.1136/thorax.55.suppl_1.S2 - DOI - PMC - PubMed
  • Weiland SK, von Mutius E, Hirsch T, Duhme H, Fritzsch C, Werner B, et al. . Prevalence of respiratory and atopic disorders among children in the East and West of Germany five years after unification. Eur Respir J. (1999) 14:862–70. 10.1034/j.1399-3003.1999.14d23.x - DOI - PubMed

Publication types

  • Search in MeSH

LinkOut - more resources

Full text sources.

  • Europe PubMed Central
  • Frontiers Media SA
  • PubMed Central

Other Literature Sources

  • scite Smart Citations

Miscellaneous

  • NCI CPTAC Assay Portal

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

The Hygiene Hypothesis

  • First Online: 04 February 2016

Cite this chapter

hygiene hypothesis original paper

  • Caroline Roduit 2 , 3 ,
  • Remo Frei 4 , 5 ,
  • Erika von Mutius 6 &
  • Roger Lauener 4 , 7  

1666 Accesses

3 Citations

3 Altmetric

Over the past decades, an increase of the prevalence of allergic diseases has been observed in Western countries. The hygiene hypothesis proposes that viral, bacterial, or helminth infections; environments with high levels of microbial components, such as farms; and the nutrition are preventive against the development of allergies despite the same genetic predisposition. The timing of these exposures is crucial. The critical window of time starts already in utero and ends in school age depending on the kind of exposure. The underlying immunological mechanism of such exposures seems rather to include the induction of regulatory processes to control the allergic reaction than to prevent the production of IgE.

In this chapter, we review the best understood exposures together with the timing and the immunological mechanisms they induce to get the most preventive effect on the development of allergic diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
  • Available as EPUB and PDF
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
  • Durable hardcover edition

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

hygiene hypothesis original paper

Hygiene Hypothesis: What Is the Current Thinking?

hygiene hypothesis original paper

The immunology of the allergy epidemic and the hygiene hypothesis

hygiene hypothesis original paper

Skewed Exposure to Environmental Antigens Complements Hygiene Hypothesis in Explaining the Rise of Allergy

Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand L, Jenmalm MC (2014) Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy 44:842–850

Article   CAS   PubMed   Google Scholar  

Akkoc T, Aydogan M, Yildiz A, Karakoc-Aydiner E, Eifan A, Keles S et al (2010) Neonatal BCG vaccination induces IL-10 production by CD4+ CD25+ T cells. Pediatr Allergy Immunol 21:1059–1063

Article   PubMed   Google Scholar  

Alfven T, Braun-Fahrlander C, Brunekreef B, von Mutius E, Riedler J, Scheynius A et al (2006) Allergic diseases and atopic sensitization in children related to farming and anthroposophic lifestyle--the PARSIFAL study. Allergy 61:414–421

Alm JS, Lilja G, Pershagen G, Scheynius A (1997) Early BCG vaccination and development of atopy. Lancet 350:400–403

Bach JF (2002) The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 347:911–920

Ball TM, Castro-Rodriguez JA, Griffith KA, Holberg CJ, Martinez FD, Wright AL (2000) Siblings, day-care attendance, and the risk of asthma and wheezing during childhood. N Engl J Med 343:538–543

Bieli C, Eder W, Frei R, Braun-Fahrlander C, Klimecki W, Waser M et al (2007) A polymorphism in CD14 modifies the effect of farm milk consumption on allergic diseases and CD14 gene expression. J Allergy Clin Immunol 120:1308–1315

Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M (2001) Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol 108:516–520

Bodner C, Godden D, Seaton A (1998) Family size, childhood infections and atopic diseases. The Aberdeen WHEASE Group. Thorax 53:28–32

Article   PubMed Central   CAS   PubMed   Google Scholar  

Boehm G, Lidestri M, Casetta P, Jelinek J, Negretti F, Stahl B et al (2002) Supplementation of a bovine milk formula with an oligosaccharide mixture increases counts of faecal bifidobacteria in preterm infants. Arch Dis Child Fetal Neonatal Ed 86:F178–F181

Brand S, Teich R, Dicke T, Harb H, Yildirim AO, Tost J et al (2011) Epigenetic regulation in murine offspring as a novel mechanism for transmaternal asthma protection induced by microbes. J Allergy Clin Immunol 128:618–25.e1–7

Braun-Fahrlander C, Gassner M, Grize L, Neu U, Sennhauser FH, Varonier HS et al (1999) Prevalence of hay fever and allergic sensitization in farmer’s children and their peers living in the same rural community. SCARPOL team. Swiss Study on Childhood Allergy and Respiratory Symptoms with Respect to Air Pollution. Clin Exp Allergy 29:28–34

Braun-Fahrlander C, Riedler J, Herz U, Eder W, Waser M, Grize L et al (2002) Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med 347:869–877

Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, Bleharski JR et al (1999) Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science 285:732–736

Brown EM, Arrieta MC, Finlay BB (2013) A fresh look at the hygiene hypothesis: how intestinal microbial exposure drives immune effector responses in atopic disease. Semin Immunol 25:378–387

Celedon JC, Wright RJ, Litonjua AA, Sredl D, Ryan L, Weiss ST et al (2003) Day care attendance in early life, maternal history of asthma, and asthma at the age of 6 years. Am J Respir Crit Care Med 167:1239–1243

Clemente JC, Ursell LK, Parfrey LW, Knight R (2012) The impact of the gut microbiota on human health: an integrative view. Cell 148:1258–1270

Conrad ML, Ferstl R, Teich R, Brand S, Blumer N, Yildirim AO et al (2009) Maternal TLR signaling is required for prenatal asthma protection by the nonpathogenic microbe Acinetobacter lwoffii F78. J Exp Med 206:2869–2877

Douwes J, Cheng S, Travier N, Cohet C, Niesink A, McKenzie J et al (2008) Farm exposure in utero may protect against asthma, hay fever and eczema. Eur Respir J 32:603–611

Eder W, Klimecki W, Yu L, von Mutius E, Riedler J, Braun-Fahrlander C et al (2004) Toll-like receptor 2 as a major gene for asthma in children of European farmers. J Allergy Clin Immunol 113:482–488

Ege MJ, von Mutius E (2013) Can genes forecast asthma risk? Lancet Respir Med 1:425–426

Ege MJ, Bieli C, Frei R, van Strien RT, Riedler J, Ublagger E et al (2006) Prenatal farm exposure is related to the expression of receptors of the innate immunity and to atopic sensitization in school-age children. J Allergy Clin Immunol 117:817–823

Ege MJ, Frei R, Bieli C, Schram-Bijkerk D, Waser M, Benz MR et al (2007) Not all farming environments protect against the development of asthma and wheeze in children. J Allergy Clin Immunol 119:1140–1147

Ege MJ, Mayer M, Normand AC, Genuneit J, Cookson WO, Braun-Fahrlander C et al (2011a) Exposure to environmental microorganisms and childhood asthma. N Engl J Med 364:701–709

Ege MJ, Strachan DP, Cookson WO, Moffatt MF, Gut I, Lathrop M et al (2011b) Gene-environment interaction for childhood asthma and exposure to farming in Central Europe. J Allergy Clin Immunol 127:138–144, 44.e1–4

Eyerich K, Novak N (2013) Immunology of atopic eczema: overcoming the Th1/Th2 paradigm. Allergy 68:974–982

Fageras Bottcher M, Hmani-Aifa M, Lindstrom A, Jenmalm MC, Mai XM, Nilsson L et al (2004) A TLR4 polymorphism is associated with asthma and reduced lipopolysaccharide-induced interleukin-12(p70) responses in Swedish children. J Allergy Clin Immunol 114:561–567

Article   PubMed   CAS   Google Scholar  

Feillet H, Bach JF (2004) Increased incidence of inflammatory bowel disease: the price of the decline of infectious burden? Curr Opin Gastroenterol 20:560–564

Frei R, Lauener RP, Crameri R, O’Mahony L (2012) Microbiota and dietary interactions: an update to the hygiene hypothesis? Allergy 67:451–461

Frei R, Roduit C, Bieli C, Loeliger S, Waser M, Scheynius A et al (2014) Expression of genes related to anti-inflammatory pathways are modified among farmers’ children. PLoS One 9:e91097

Article   PubMed Central   PubMed   CAS   Google Scholar  

Gehring U, Bolte G, Borte M, Bischof W, Fahlbusch B, Wichmann HE et al (2001) Exposure to endotoxin decreases the risk of atopic eczema in infancy: a cohort study. J Allergy Clin Immunol 108:847–854

Hanski I, von Hertzen L, Fyhrquist N, Koskinen K, Torppa K, Laatikainen T et al (2012) Environmental biodiversity, human microbiota, and allergy are interrelated. Proc Natl Acad Sci U S A 109:8334–8339

Hoffjan S, Nicolae D, Ostrovnaya I, Roberg K, Evans M, Mirel DB et al (2005) Gene-environment interaction effects on the development of immune responses in the 1st year of life. Am J Hum Genet 76:696–704

Illi S, von Mutius E, Lau S, Bergmann R, Niggemann B, Sommerfeld C et al (2001) Early childhood infectious diseases and the development of asthma up to school age: a birth cohort study. BMJ 322:390–395

Illi S, Depner M, Genuneit J, Horak E, Loss G, Strunz-Lehner C et al (2012) Protection from childhood asthma and allergy in Alpine farm environments-the GABRIEL Advanced Studies. J Allergy Clin Immunol 129:1470–1477.e6

Kabesch M (2014) Epigenetics in asthma and allergy. Curr Opin Allergy Clin Immunol 14:62–68

Kalliomaki M, Isolauri E (2003) Role of intestinal flora in the development of allergy. Curr Opin Allergy Clin Immunol 3:15–20

Kim YS, Kwon KS, Kim DK, Choi IW, Lee HK (2004) Inhibition of murine allergic airway disease by Bordetella pertussis. Immunology 112:624–630

Kim HY, DeKruyff RH, Umetsu DT (2010) The many paths to asthma: phenotype shaped by innate and adaptive immunity. Nat Immunol 11:577–584

Kondo N, Kobayashi Y, Shinoda S, Takenaka R, Teramoto T, Kaneko H et al (1998) Reduced interferon gamma production by antigen-stimulated cord blood mononuclear cells is a risk factor of allergic disorders--6-year follow-up study. Clin Exp Allergy 28:1340–1344

Krämer U, Heinrich J, Wjst M, Wichmann HE (1999) Age of entry to day nursery and allergy in later childhood. Lancet 353:450–454

Google Scholar  

Kudo M, Melton AC, Chen C, Engler MB, Huang KE, Ren X et al (2012) IL-17A produced by alpha beta T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med 18:547–554

Langan SM, Flohr C, Williams HC (2007) The role of furry pets in eczema: a systematic review. Arch Dermatol 143:1570–1577

Lauener RP, Birchler T, Adamski J, Braun-Fahrlander C, Bufe A, Herz U et al (2002) Expression of CD14 and Toll-like receptor 2 in farmers’ and non-farmers’ children. Lancet 360:465–466

Li XM, Srivastava K, Huleatt JW, Bottomly K, Burks AW, Sampson HA (2003) Engineered recombinant peanut protein and heat-killed Listeria monocytogenes coadministration protects against peanut-induced anaphylaxis in a murine model. J Immunol 170:3289–3295

Litonjua AA, Belanger K, Celedon JC, Milton DK, Bracken MB, Kraft P et al (2005) Polymorphisms in the 5′ region of the CD14 gene are associated with eczema in young children. J Allergy Clin Immunol 115:1056–1062

Lluis A, Ballenberger N, Illi S, Schieck M, Kabesch M, Illig T et al (2014a) Regulation of TH17 markers early in life through maternal farm exposure. J Allergy Clin Immunol 133:864–871

Lluis A, Depner M, Gaugler B, Saas P, Casaca VI, Raedler D et al (2014b) Increased regulatory T-cell numbers are associated with farm milk exposure and lower atopic sensitization and asthma in childhood. J Allergy Clin Immunol 133:551–559

Lodge CJ, Allen KJ, Lowe AJ, Hill DJ, Hosking CS, Abramson MJ et al (2012) Perinatal cat and dog exposure and the risk of asthma and allergy in the urban environment: a systematic review of longitudinal studies. Clin Dev Immunol 2012:176484

Article   PubMed Central   PubMed   Google Scholar  

Loftus EV Jr (2004) Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences. Gastroenterology 126:1504–1517

Loss G, Apprich S, Waser M, Kneifel W, Genuneit J, Buchele G et al (2011) The protective effect of farm milk consumption on childhood asthma and atopy: the GABRIELA study. J Allergy Clin Immunol 128:766–773.e4

Loss G, Bitter S, Wohlgensinger J, Frei R, Roduit C, Genuneit J et al (2012) Prenatal and early-life exposures alter expression of innate immunity genes: the PASTURE cohort study. J Allergy Clin Immunol 130:523–530.e9

Lyons A, O’Mahony D, O’Brien F, MacSharry J, Sheil B, Ceddia M et al (2010) Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy 40:811–819

CAS   PubMed   Google Scholar  

Marshall AL, Chetwynd A, Morris JA, Placzek M, Smith C, Olabi A et al (2004) Type 1 diabetes mellitus in childhood: a matched case control study in Lancashire and Cumbria, UK. Diabet Med 21:1035–1040

Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D et al (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461:1282–1286

Matricardi PM, Rosmini F, Ferrigno L, Nisini R, Rapicetta M, Chionne P et al (1997) Cross sectional retrospective study of prevalence of atopy among Italian military students with antibodies against hepatitis A virus. BMJ 314:999–1003

McDade TW (2012) Early environments and the ecology of inflammation. Proc Natl Acad Sci U S A 109(Suppl 2):17281–17288

McDade TW, Kuzawa CW, Adair LS, Beck MA (2004) Prenatal and early postnatal environments are significant predictors of total immunoglobulin E concentration in Filipino adolescents. Clin Exp Allergy 34:44–50

McKeever TM, Lewis SA, Smith C, Collins J, Heatlie H, Frischer M et al (2002) Early exposure to infections and antibiotics and the incidence of allergic disease: a birth cohort study with the West Midlands General Practice Research Database. J Allergy Clin Immunol 109:43–50

Michel S, Busato F, Genuneit J, Pekkanen J, Dalphin JC, Riedler J et al (2013) Farm exposure and time trends in early childhood may influence DNA methylation in genes related to asthma and allergy. Allergy 68:355–364

Michelson PH, Williams LW, Benjamin DK, Barnato AE (2009) Obesity, inflammation, and asthma severity in childhood: data from the National Health and Nutrition Examination Survey 2001-2004. Ann Allergy Asthma Immunol 103:381–385

Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G (2006) A mixture of prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the first six months of age. Arch Dis Child 91:814–819

Mrabet-Dahbi S, Maurer M (2010) Does allergy impair innate immunity? Leads and lessons from atopic dermatitis. Allergy 65:1351–1356

Mustonen K, Keski-Nisula L, Vaarala O, Pfefferle PI, Renz H, Riedler J et al (2012) Few associations between high-sensitivity C-reactive protein and environmental factors in 4.5-year-old children. Pediatr Allergy Immunol 23:522–528

Neaville WA, Tisler C, Bhattacharya A, Anklam K, Gilbertson-White S, Hamilton R et al (2003) Developmental cytokine response profiles and the clinical and immunologic expression of atopy during the first year of life. J Allergy Clin Immunol 112:740–746

Novak N, Yu CF, Bussmann C, Maintz L, Peng WM, Hart J et al (2007) Putative association of a TLR9 promoter polymorphism with atopic eczema. Allergy 62:766–772

Noverr MC, Falkowski NR, McDonald RA, McKenzie AN, Huffnagle GB (2005) Development of allergic airway disease in mice following antibiotic therapy and fungal microbiota increase: role of host genetics, antigen, and interleukin-13. Infect Immun 73:30–38

Nwaru BI, Takkinen HM, Kaila M, Erkkola M, Ahonen S, Pekkanen J et al (2014) Food diversity in infancy and the risk of childhood asthma and allergies. J Allergy Clin Immunol 133:1084–1091

Olesen AB, Juul S, Thestrup-Pedersen K (2003) Atopic dermatitis is increased following vaccination for measles, mumps and rubella or measles infection. Acta Derm Venereol 83:445–450

Ong MS, Umetsu DT, Mandl KD (2014) Consequences of antibiotics and infections in infancy: bugs, drugs, and wheezing. Ann Allergy Asthma Immunol 112:441–445.e1

Paunio M, Heinonen OP, Virtanen M, Leinikki P, Patja A, Peltola H (2000) Measles history and atopic diseases: a population-based cross-sectional study. JAMA 283:343–346

Pfefferle PI, Buchele G, Blumer N, Roponen M, Ege MJ, Krauss-Etschmann S et al (2010) Cord blood cytokines are modulated by maternal farming activities and consumption of farm dairy products during pregnancy: the PASTURE Study. J Allergy Clin Immunol 125:108–15.e1–3

Phipatanakul W, Celedon JC, Raby BA, Litonjua AA, Milton DK, Sredl D et al (2004) Endotoxin exposure and eczema in the first year of life. Pediatrics 114:13–18

Prescott SL, Smith P, Tang M, Palmer DJ, Sinn J, Huntley SJ et al (2008) The importance of early complementary feeding in the development of oral tolerance: concerns and controversies. Pediatr Allergy Immunol 19:375–380

Riedler J, Eder W, Oberfeld G, Schreuer M (2000) Austrian children living on a farm have less hay fever, asthma and allergic sensitization. Clin Exp Allergy 30:194–200

Riedler J, Braun-Fahrlander C, Eder W, Schreuer M, Waser M, Maisch S et al (2001) Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet 358:1129–1133

Robinson DS, Larche M, Durham SR (2004) Tregs and allergic disease. J Clin Invest 114:1389–1397

Roduit C, Wohlgensinger J, Frei R, Bitter S, Bieli C, Loeliger S et al (2011) Prenatal animal contact and gene expression of innate immunity receptors at birth are associated with atopic dermatitis. J Allergy Clin Immunol 127:179–185, 85.e1

Roduit C, Frei R, Loss G, Buchele G, Weber J, Depner M et al (2012) Development of atopic dermatitis according to age of onset and association with early-life exposures. J Allergy Clin Immunol 130:130–136.e5

Roduit C, Frei R, Depner M, Schaub B, Loss G, Genuneit J et al (2014) Increased food diversity in the first year of life is inversely associated with allergic diseases. J Allergy Clin Immunol 133:1056–1064

Romagnani S (2004a) Immunologic influences on allergy and the TH1/TH2 balance. J Allergy Clin Immunol 113:395–400

Romagnani S (2004b) The increased prevalence of allergy and the hygiene hypothesis: missing immune deviation, reduced immune suppression, or both? Immunology 112:352–363

Russell DA, Ross RP, Fitzgerald GF, Stanton C (2011) Metabolic activities and probiotic potential of bifidobacteria. Int J Food Microbiol 149:88–105

Saemann MD, Bohmig GA, Osterreicher CH, Burtscher H, Parolini O, Diakos C et al (2000) Anti-inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL-12 and up-regulation of IL-10 production. FASEB J 14:2380–2382

Schaub B, Liu J, Hoppler S, Schleich I, Huehn J, Olek S et al (2009) Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J Allergy Clin Immunol 123:774–782.e5

Schmitt J, Schmitt NM, Kirch W, Meurer M (2010) Early exposure to antibiotics and infections and the incidence of atopic eczema: a population-based cohort study. Pediatr Allergy Immunol 21:292–300

Schumann A, Nutten S, Donnicola D, Comelli EM, Mansourian R, Cherbut C et al (2005) Neonatal antibiotic treatment alters gastrointestinal tract developmental gene expression and intestinal barrier transcriptome. Physiol Genomics 23:235–245

Shirakawa T, Enomoto T, Shimazu S, Hopkin JM (1997) The inverse association between tuberculin responses and atopic disorder. Science 275:77–79

Silverberg JI, Kleiman E, Silverberg NB, Durkin HG, Joks R, Smith-Norowitz TA (2012) Chickenpox in childhood is associated with decreased atopic disorders, IgE, allergic sensitization, and leukocyte subsets. Pediatr Allergy Immunol 23:50–58

Smit LA, Siroux V, Bouzigon E, Oryszczyn MP, Lathrop M, Demenais F et al (2009) CD14 and toll-like receptor gene polymorphisms, country living, and asthma in adults. Am J Respir Crit Care Med 179:363–368

Sozanska B, Pearce N, Dudek K, Cullinan P (2013) Consumption of unpasteurized milk and its effects on atopy and asthma in children and adult inhabitants in rural Poland. Allergy 68:644–650

Strachan DP (1989) Hay fever, hygiene, and household size. BMJ 299:1259–1260

Strachan DP, Harkins LS, Golding J (1997a) Sibship size and self-reported inhalant allergy among adult women. ALSPAC Study Team. Clin Exp Allergy 27:151–155

Strachan DP, Harkins LS, Johnston ID, Anderson HR (1997b) Childhood antecedents of allergic sensitization in young British adults. J Allergy Clin Immunol 99:6–12

Takeda K, Akira S (2005) Toll-like receptors in innate immunity. Int Immunol 17:1–14

Takkouche B, Gonzalez-Barcala FJ, Etminan M, Fitzgerald M (2008) Exposure to furry pets and the risk of asthma and allergic rhinitis: a meta-analysis. Allergy 63:857–864

Tedelind S, Westberg F, Kjerrulf M, Vidal A (2007) Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol 13:2826–2832

PubMed Central   CAS   PubMed   Google Scholar  

Townley RG, Barlan IB, Patino C, Vichyanond P, Minervini MC, Simasathien T et al (2004) The effect of BCG vaccine at birth on the development of atopy or allergic disease in young children. Ann Allergy Asthma Immunol 92:350–355

Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C et al (2014) Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20:159–166

Tukenmez F, Bahceciler NN, Barlan IB, Basaran MM (1999) Effect of pre-immunization by killed Mycobacterium bovis and vaccae on immunoglobulin E response in ovalbumin-sensitized newborn mice. Pediatr Allergy Immunol 10:107–111

van den Elsen LW, Nusse Y, Balvers M, Redegeld FA, Knol EF, Garssen J et al (2013) n-3 Long-chain PUFA reduce allergy-related mediator release by human mast cells in vitro via inhibition of reactive oxygen species. Br J Nutr 109:1821–1831

van Strien RT, Engel R, Holst O, Bufe A, Eder W, Waser M et al (2004) Microbial exposure of rural school children, as assessed by levels of N-acetyl-muramic acid in mattress dust, and its association with respiratory health. J Allergy Clin Immunol 113:860–867

Velasco G, Campo M, Manrique OJ, Bellou A, He H, Arestides RS et al (2005) Toll-like receptor 4 or 2 agonists decrease allergic inflammation. Am J Respir Cell Mol Biol 32:218–224

Vercelli D (2006) Mechanisms of the hygiene hypothesis--molecular and otherwise. Curr Opin Immunol 18:733–737

Vercelli D, Gozdz J, von Mutius E (2014) Innate lymphoid cells in asthma: when innate immunity comes in a Th2 flavor. Curr Opin Allergy Clin Immunol 14:29–34

Von Ehrenstein OS, Von Mutius E, Illi S, Baumann L, Bohm O, von Kries R (2000) Reduced risk of hay fever and asthma among children of farmers. Clin Exp Allergy 30:187–193

Article   Google Scholar  

von Mutius E, Vercelli D (2010) Farm living: effects on childhood asthma and allergy. Nat Rev Immunol 10:861–868

Article   CAS   Google Scholar  

von Mutius E, Pearce N, Beasley R, Cheng S, von Ehrenstein O, Bjorksten B et al (2000a) International patterns of tuberculosis and the prevalence of symptoms of asthma, rhinitis, and eczema. Thorax 55:449–453

von Mutius E, Braun-Fahrlander C, Schierl R, Riedler J, Ehlermann S, Maisch S et al (2000b) Exposure to endotoxin or other bacterial components might protect against the development of atopy. Clin Exp Allergy 30:1230–1234

Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP et al (2008) Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol 121:129–134

Watanabe S, Narisawa Y, Arase S, Okamatsu H, Ikenaga T, Tajiri Y et al (2003) Differences in fecal microflora between patients with atopic dermatitis and healthy control subjects. J Allergy Clin Immunol 111:587–591

Werner M, Topp R, Wimmer K, Richter K, Bischof W, Wjst M et al (2003) TLR4 gene variants modify endotoxin effects on asthma. J Allergy Clin Immunol 112:323–330

Wiria AE, Djuardi Y, Supali T, Sartono E, Yazdanbakhsh M (2012) Helminth infection in populations undergoing epidemiological transition: a friend or foe? Semin Immunopathol 34:889–901

Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema: ISAAC. The International Study of Asthma and Allergies in Childhood (ISAAC) Steering Committee (1998) Lancet. 351:1225–1232

Yazdanbakhsh M, Kremsner PG, van Ree R (2002) Allergy, parasites, and the hygiene hypothesis. Science 296:490–494

Download references

Author information

Authors and affiliations.

Department of Allergy/Immunology, Children’s Hospital, University of Zurich, Zurich, Switzerland

Caroline Roduit

Department of Allergy/Immunology, Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland

Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland

Remo Frei & Roger Lauener

Molecular Immunology, Swiss Institute of Allergy and Asthma Research, University of Zurich, Zurich, Switzerland

Department of Asthma and Allergy, Dr von Hauner Children’s Hospital, Ludwig Maximilians University Munich, Munich, Germany

Erika von Mutius

Children’s Hospital of Eastern Switzerland, St. Gallen, Switzerland

Roger Lauener

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Caroline Roduit .

Editor information

Editors and affiliations.

University of Düsseldorf, IUF–Leibniz Res Inst for Env Med, Düsseldorf, Germany

Charlotte Esser

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer-Verlag Wien

About this chapter

Roduit, C., Frei, R., von Mutius, E., Lauener, R. (2016). The Hygiene Hypothesis. In: Esser, C. (eds) Environmental Influences on the Immune System. Springer, Vienna. https://doi.org/10.1007/978-3-7091-1890-0_4

Download citation

DOI : https://doi.org/10.1007/978-3-7091-1890-0_4

Published : 04 February 2016

Publisher Name : Springer, Vienna

Print ISBN : 978-3-7091-1888-7

Online ISBN : 978-3-7091-1890-0

eBook Packages : Biomedical and Life Sciences Biomedical and Life Sciences (R0)

Share this chapter

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Publish with us

Policies and ethics

  • Find a journal
  • Track your research
  • Biochemistry and Molecular Biology
  • Biostatistics
  • Environmental Health and Engineering
  • Epidemiology
  • Health Policy and Management
  • Health, Behavior and Society
  • International Health
  • Mental Health
  • Molecular Microbiology and Immunology
  • Population, Family and Reproductive Health
  • Program Finder
  • Admissions Services
  • Course Directory
  • Academic Calendar
  • Hybrid Campus
  • Lecture Series
  • Convocation
  • Strategy and Development
  • Implementation and Impact
  • Integrity and Oversight
  • In the School
  • In the Field
  • In Baltimore
  • Resources for Practitioners
  • Articles & News Releases
  • In The News
  • Statements & Announcements
  • At a Glance
  • Student Life
  • Strategic Priorities
  • Inclusion, Diversity, Anti-Racism, and Equity (IDARE)
  • What is Public Health?

Is the Hygiene Hypothesis True?

Did Covid shutdowns stunt kids' immune systems?

Caitlin Rivers

The hygiene hypothesis is the idea that kids need to be exposed to germs in order to develop healthy immune systems. We know that many common viruses did not circulate as widely during the pandemic, thanks to social distancing, masking, and other COVID mitigation measures. Are there downsides to those missed infections? 

In this Q&A, Caitlin Rivers speaks with Marsha Wills-Karp, PhD, MHS , professor and chair of Environmental Health and Engineering , about the role of household microbiomes, birth, and vaccines in the development of kids’ immune systems—and whether early exposure really is the best medicine.

This Q&A is adapted from Rivers’ Substack blog, Force of Infection .

I think there’s some concern among parents who have heard about the hygiene hypothesis that there is a downside to all those stuffy noses that didn’t happen [during the COVID-19 pandemic]. Are there any upsides to viral infections? Do they help the immune system in some meaningful way?

I don’t think so.

You mentioned the hygiene hypothesis, which was postulated back in the ‘80s. German scientists noticed that families with fewer children tended to have more allergic disease. This was interpreted [to mean] that allergic disease was linked to experiencing fewer infections. I have explored this idea in my research for a couple of decades now.

This phenomenon has helped us to understand the immune system, but our interpretation of it has grown and expanded—particularly with respect to viruses. Almost no virus is protective against allergic disease or other immune diseases. In fact, infections with viruses mostly either contribute to the development of those diseases or worsen them.

The opposite is true of bacteria. There are good bacteria and there are bad bacteria. The good bacteria we call commensals . Our bodies actually have more bacterial cells than human cells. What we’ve learned over the years is that the association with family life and the environment probably has more to do with the microbiome. So one thing I would say is sanitizing every surface in your home to an extreme is probably not a good thing. Our research team showed in animals that sterile environments don’t allow the immune system to develop at all. We don’t want that.

What does contribute to the development of the immune system, if not exposure to viruses?

There are a number of factors that we’ve associated with the hygiene hypothesis over the last 20 years, and these exposures start very early in life. Cesarean sections, which do not allow the baby to travel through the birth canal and get exposed to the mother’s really healthy bacterial content, is a risk factor for many different immune diseases. Getting that early seeding with good bacteria is critical for setting up the child going forward. Breastfeeding also contributes to the development of a healthy immune system.

There are other factors. Our diets have changed dramatically over the years. We eat a lot of processed food that doesn’t have the normal components of a healthy microbiome, like fiber. These healthy bacteria in our gut need that fiber to maintain themselves. They not only are important for our immune system but they’re absolutely critical to us deriving calories and nutrients from our food. All these things contribute to a healthy child.

We’ve also noticed that people who live on farms have fewer of these diseases because they’re exposed to—for lack of a better term—the fecal material of animals. And what we have found is that it’s due to these commensal bacteria. That is one of the components that help us keep a healthy immune system. Most of us will probably not adopt farm life. But we can have a pet, we can have a dog.

I think all the pet lovers out there will be pleased to hear that.

There’s a lot of evidence that owning a pet in early childhood is very protective.

What about the idea that you need to be exposed to viruses in early life because if you get them as an adult, you’ll get more severely ill? We know that’s true for chickenpox, for example. Do you have any concerns about that?

We should rely on vaccines for those exposures because we can never predict who is going to be susceptible to severe illness, even in early childhood. If we look back before vaccines, children under 4 often succumbed to infections. I don’t think we want to return to that time in history.

Let me just give you one example. There’s a virus called RSV, it’s a respiratory virus. Almost all infants are positive for it by the age of 2. But those who get severe disease are more likely to develop allergic disease and other problems. So this idea that we must become infected with a pathogenic virus to be healthy is not a good one.

Even rhinovirus, which is the common cold, most people recover fine. But there’s a lot of evidence that for somebody who is allergic, rhinovirus exposures make them much worse. In fact, most allergic or asthmatic kids suffer through the winter months when these viruses are more common.

And that’s particularly salient because there is a lot of rhinovirus and enterovirus circulating right now.

From my point of view, right now, avoiding flu and COVID-19 is a priority. Those are not going to help you develop a healthy immune response, and in fact, they can do a lot of damage to the lungs during that critical developmental time. Data [show] that children that have more infections in the first 6 months to a year of life go on to have more problems.

It’s always surprising to me when I look at the data of the fraction of time that young children spend with these common colds—and this is pre-pandemic—it’s not uncommon for kids to be sick 50% of the time. That feels right as a parent, but it’s startling.

The other thing people don’t know is that the GI tract is where you get tolerized to all of your foods, allergens and things. Without those healthy bacteria in your gut, you can’t tolerate common allergens.

How does that relate to the guidance that’s changed over the years—that you should withhold peanuts in early life and now you’re supposed to offer them in early life?

The guidance to delay exposure to peanuts didn’t consider the fact that oral exposure to peanuts was not the only exposure kids were getting. There were peanut oils in all kinds of skin creams and other things. So kids got exposed through their skin, but they had no gut protection—and the GI tract is important for a tolerant system. If you have a healthy immune response, you get tolerized in early life.

This concept is a little bit different for those families who may already have a predisposition to allergies. But for the general public, exposure is key to protecting them in early life.

I think some parents look at the guidance that you should now offer peanuts in early life and say, “Are we not doing that with rhinovirus by masking kids or improving ventilation?” How should people think about the development of the immune system for food allergies compared to infections?

The thing about rhinoviruses is that after recovering, you’re not protected from the next infection. There is no real immune protection there. Most of us suffer from colds throughout our whole life. Like I said, bacterial exposure is what’s key to priming the immune response. 

Also, we forget that a lot of kids die from the flu. Unlike COVID-19, where younger kids are not quite as susceptible to severe illness, that’s not true for flu. RSV, too, can be quite severe in young children and older adults.

Caitlin Rivers, PhD, MPH , is a senior scholar at the Johns Hopkins Center for Health Security and an assistant professor in Environmental Health and Engineering at the Johns Hopkins Bloomberg School of Public Health.

RELATED: 

  • Study Finds That Children’s Antibody Responses to COVID-19 Are Stronger Than Adults’
  • Back to School: COVID, CDC Guidance, Monkeypox, and More
  • A New Shot Prevents Serious Illness from RSV

Related Content

A microscopic image of fungus

A ‘Critical Pathogen’: The Rise of Drug-Resistant Fungal Diseases

covid vaccine vials, syringes, and bandaids on a metal tray

What to Know About the Updated COVID Vaccine for Fall, Winter 2024–25

A world globe with a mask on

Why We’re Still Waiting for a Pandemic Treaty

An illustration of a wave in the ocean covered in COVID molecules with a person swimming underneath it holding a vaccine

Why COVID Surges in the Summer

A vehicle is left abandoned in floodwater on a highway after Hurricane Beryl swept through the area on July 8, 2024, in Houston, Texas.

The Visible and Unseen Dangers Lurking in Floodwater

  • A-Z Publications

Annual Review of Anthropology

Volume 51, 2022, review article, the ecoimmunology of health and disease: the hygiene hypothesis and plasticity in human immune function.

  • Aaron D. Blackwell 1
  • View Affiliations Hide Affiliations Affiliations: Department of Anthropology, Washington State University, Pullman, Washington, USA; email: [email protected]
  • Vol. 51:401-418 (Volume publication date October 2022) https://doi.org/10.1146/annurev-anthro-101819-110236
  • First published as a Review in Advance on July 27, 2022
  • Copyright © 2022 by Annual Reviews. All rights reserved This article is part of a special theme on Global Health. For a list of other articles in this theme, see https://www.annualreviews.org/toc/anthro/51/1

The original hygiene hypothesis proposed that certain diseases derive from low levels of early-life microbial exposure. Since then, the hypothesis has been applied to numerous inflammatory, autoimmune, and allergic conditions. The changes in hygiene linked to these diseases include numerous changes in biotic exposure and lifestyle. To this end, some scholars have called for abandonment of the term or have suggested alternate labels, e.g., the old friends hypothesis. However, neither of these terms encompasses the complexity of plasticity in immune response and host–parasite/commensal interactions that influence these conditions. Here, I review this complexity, with particular regard to the factors affecting immunological strategies, the development of tolerance, immune dysfunction, and ecological interactions among organisms. I discuss the biotic factors that affect immune plasticity and how these interact with abiotic factors such as nutrition, as well as how transgenerational exposures may affect immune plasticity. Finally, I review the general features of diseases linked to biotic exposures.

Article metrics loading...

Full text loading...

Literature Cited

  • Ahmed SA. 2000 . The immune system as a potential target for environmental estrogens (endocrine disrupters): a new emerging field. Toxicology 150 : 1–3 191– 206 [Google Scholar]
  • Alcock J , Franklin ML , Kuzawa CW. 2012 . Nutrient signaling: evolutionary origins of the immune-modulating effects of dietary fat. Q. Rev. Biol. 87 : 3 187– 223 [Google Scholar]
  • Anderson AS , Trumble BC , Hové C , Kraft TS , Kaplan H et al. 2020 . Old friends and friendly fire: pregnancy, hookworm infection, and anemia among tropical horticulturalists. Am. J. Hum. Biol. 32 : 2 e23337 [Google Scholar]
  • Angelucci F , Cechova K , Amlerova J , Hort J. 2019 . Antibiotics, gut microbiota, and Alzheimer's disease. J. Neuroinflamm. 16 : 1 108 [Google Scholar]
  • Bach J-F. 2018 . The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat. Rev. Immunol. 18 : 2 105– 20 [Google Scholar]
  • Barker DJ. 1985 . Acute appendicitis and dietary fibre: an alternative hypothesis. BMJ 290 : 6475 1125– 27 [Google Scholar]
  • Barker DJ , Morris JA , Simmonds SJ , Oliver RH. 1988 . Appendicitis epidemic following introduction of piped water to Anglesey. J. Epidemiol. Community Health 42 : 2 144– 48 [Google Scholar]
  • Barker DJP , Morris J. 1988 . Acute appendicitis, bathrooms, and diet in Britain and Ireland. BMJ 296 : 6627 953– 55 [Google Scholar]
  • Becker KG. 2007 . Autism, asthma, inflammation, and the hygiene hypothesis. Med. Hypotheses 69 : 4 731– 40 [Google Scholar]
  • Begum K , Cooper GD , Akhter N , Nahar P , Kasim A , Bentley GR. 2021 . Early life, life course and gender influences on levels of C-reactive protein among migrant Bangladeshis in the UK. Evol. Med. Public Health 10 : 21– 35 [Google Scholar]
  • Belkaid Y , Rouse BT. 2005 . Natural regulatory T cells in infectious disease. Nat. Immunol. 6 : 4 353– 60 [Google Scholar]
  • Bentham J , Di Cesare M , Bilano V , Bixby H , Zhou B et al. 2017 . Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet 390 : 10113 2627– 42 [Google Scholar]
  • Bergström EMK , Short DJ , Frankel HL , Henderson NJ , Jones PRM. 1999 . The effect of childhood spinal cord injury on skeletal development: a retrospective study. Spinal Cord 37 : 12 838– 46 [Google Scholar]
  • Blackwell AD. 2016 . Helminth infection during pregnancy: insights from evolutionary ecology. Int. J. Womens Health 8 : 651– 61 [Google Scholar]
  • Blackwell AD , Gurven M , Sugiyama LS , Madimenos FC , Liebert MA et al. 2011 . Evidence for a peak shift in a humoral response to helminths: age profiles of IgE in the Shuar of Ecuador, the Tsimane of Bolivia, and the U.S. NHANES. PLOS Negl. Trop. Dis. 5 : 6 e1218 [Google Scholar]
  • Blackwell AD , Martin M , Kaplan H , Gurven M. 2013 . Antagonism between two intestinal parasites in humans: the importance of co-infection for infection risk and recovery dynamics. Proc. R. Soc. B 280 : 1769 . 20131671 [Google Scholar]
  • Blackwell AD , Snodgrass JJ , Madimenos FC , Sugiyama LS. 2010 . Life history, immune function, and intestinal helminths: trade-offs among immunoglobulin E, C-reactive protein, and growth in an Amazonian population. Am. J. Hum. Biol. 22 : 6 836– 48 [Google Scholar]
  • Blackwell AD , Tamayo MA , Beheim B , Trumble BC , Stieglitz J et al. 2015 . Helminth infection, fecundity, and age of first pregnancy in women. Science 350 : 6263 970– 72 [Google Scholar]
  • Blackwell AD , Trumble BC , Maldonado Suarez I , Stieglitz J , Beheim B et al. 2016 . Immune function in Amazonian horticulturalists. Ann. Hum. Biol. 43 : 4 382– 96 [Google Scholar]
  • Bloomfield SF , Rook GAW , Scott EA , Shanahan F , Stanwell-Smith R , Turner P. 2016 . Time to abandon the hygiene hypothesis: new perspectives on allergic disease, the human microbiome, infectious disease prevention and the role of targeted hygiene. Perspect. Public Health 136 : 4 213– 24 [Google Scholar]
  • Borghesi J , Mario LC , Rodrigues MN , Favaron PO , Miglino MA. 2014 . Immunoglobulin transport during gestation in domestic animals and humans—a review. Open J. Anim. Sci. 4 : 5 323– 36 [Google Scholar]
  • Borkow G , Leng Q , Weisman Z , Stein M , Galai N et al. 2000 . Chronic immune activation associated with intestinal helminth infections results in impaired signal transduction and anergy. J. Clin. Invest. 106 : 8 1053– 60 [Google Scholar]
  • Breslau J , Borges G , Hagar Y , Tancredi D , Gilman S. 2009 . Immigration to the USA and risk for mood and anxiety disorders: variation by origin and age at immigration. Psychol. Med. 39 : 7 1117– 27 [Google Scholar]
  • Briggs N , Weatherhead J , Sastry KJ , Hotez PJ. 2016 . The hygiene hypothesis and its inconvenient truths about helminth infections. PLOS Negl. Trop. Dis. 10 : 9 e0004944 [Google Scholar]
  • Burt TD. 2013 . Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease. Am. J. Reprod. Immunol. 69 : 4 346– 58 [Google Scholar]
  • Cepon-Robins TJ , Gildner TE 2020 . Old friends meet a new foe: a potential role for immune-priming parasites in mitigating COVID-19 morbidity and mortality. Evol. Med. Public Health 2020 : 1 234– 48 [Google Scholar]
  • Chalubinski M , Kowalski ML. 2006 . Endocrine disrupters—potential modulators of the immune system and allergic response. Allergy Eur. J. Allergy Clin. Immunol. 61 : 11 1326– 35 [Google Scholar]
  • Charnov EL. 1991 . Evolution of life history variation among female mammals. PNAS 88 : 4 1134– 37 [Google Scholar]
  • Chatterjee B , Karandikar RL , Mande SC. 2021 . Mortality due to COVID-19 in different countries is associated with their demographic character and prevalence of autoimmunity. Curr. Sci. 120 : 3 501– 8 [Google Scholar]
  • de Ruiter K , Tahapary DL , Sartono E , Soewondo P , Supali T et al. 2017 . Helminths, hygiene hypothesis and type 2 diabetes. Parasite Immunol 39 : 5 e12404 [Google Scholar]
  • Dean G , Elian M. 1997 . Age at immigration to England of Asian and Caribbean immigrants and the risk of developing multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 63 : 5 565– 68 [Google Scholar]
  • Demas GE , Nelson RJ. 2012 . Ecoimmunology Oxford, UK: Oxford Univ. Press [Google Scholar]
  • Effros RB , Dagarag M , Spaulding C , Man J 2005 . The role of CD8+ T-cell replicative senescence in human aging. Immunol. Rev. 205 : 147– 57 [Google Scholar]
  • Eisenstein EM , Williams CB. 2009 . The T reg /Th17 cell balance: a new paradigm for autoimmunity. Pediatr. Res. 65 : 5 Pt. 2 26R– 31 R [Google Scholar]
  • Fleming J , Hernandez G , Hartman L , Maksimovic J , Nace S et al. 2019 . Safety and efficacy of helminth treatment in relapsing-remitting multiple sclerosis: results of the HINT 2 clinical trial. Mult. Scler. J. 25 : 1 81– 91 [Google Scholar]
  • Fox M , Knapp LA , Andrews PW , Fincher CL. 2013 . Hygiene and the world distribution of Alzheimer's disease: epidemiological evidence for a relationship between microbial environment and age-adjusted disease burden. Evol. Med. Public Health 2013 : 1 173– 86 [Google Scholar]
  • Giacomin P , Agha Z , Loukas A. 2016 . Helminths and intestinal flora team up to improve gut health. Trends Parasitol 32 : 9 664– 66 [Google Scholar]
  • Graham AL , Shuker DM , Pollitt LC , Auld SKJR , Wilson AJ , Little TJ. 2011 . Fitness consequences of immune responses: strengthening the empirical framework for ecoimmunology. Funct. Ecol. 25 : 1 5– 17 [Google Scholar]
  • Gurven M , Stieglitz J , Trumble B , Blackwell AD , Beheim B et al. 2017 . The Tsimane Health and Life History Project: integrating anthropology and biomedicine. Evol. Anthropol. 26 : 2 54– 73 [Google Scholar]
  • Gurven MD , Kaplan H , Winking J , Eid Rodriguez D , Vasunilashorn S et al. 2009 . Inflammation and infection do not promote arterial aging and cardiovascular disease risk factors among lean horticulturalists. PLOS ONE 4 : 8 e6590 [Google Scholar]
  • Gurven MD , Trumble BC , Stieglitz J , Blackwell AD , Michalik DE et al. 2016 . Cardiovascular disease and type 2 diabetes in evolutionary perspective: a critical role for helminths?. Evol. Med. Public Health 2016 : 1 338– 57 [Google Scholar]
  • Harrington WE , Kanaan SB , Muehlenbachs A , Morrison R , Stevenson P et al. 2017 . Maternal micro-chimerism predicts increased infection but decreased disease due to Plasmodium falciparum during early childhood. J. Infect. Dis. 215 : 9 1445– 51 [Google Scholar]
  • Hays R , Pierce D , Giacomin P , Loukas A , Bourke P , McDermott R. 2020 . Helminth coinfection and COVID-19: an alternate hypothesis. PLOS Negl. Trop. Dis. 14 : 8 e0008628 [Google Scholar]
  • Helmby H. 2015 . Human helminth therapy to treat inflammatory disorders – where do we stand?. BMC Immunol 16 : 1 12 [Google Scholar]
  • Hotez PJ , Brindley PJ , Bethony JM , King CH , Pearce EJ , Jacobson J. 2008 . Helminth infections: the great neglected tropical diseases. J. Clin. Investig. 118 : 4 1311– 21 [Google Scholar]
  • Hunter P. 2020 . Public health struggles to square hygiene with diversity: research on the link between microbiomes and immune function puts the “hygiene hypothesis” to rest. EMBO Rep 21 : 10 e51540 [Google Scholar]
  • Iancovici Kidon M , Stein M , Geller-Bernstein C , Weisman Z , Steinberg S et al. 2005 . Serum immunoglobulin E levels in Israeli-Ethiopian children: environment and genetics. Isr. Med. Assoc. J. 7 : 12 799– 802 [Google Scholar]
  • Jackson JA , Hall AJ , Friberg IM , Ralli C , Lowe A et al. 2014 . An immunological marker of tolerance to infection in wild rodents. PLOS Biol 12 : 7 e1001901 [Google Scholar]
  • Jiang TT , Chaturvedi V , Ertelt JM , Kinder JM , Clark DR et al. 2014 . Regulatory T cells: new keys for further unlocking the enigma of fetal tolerance and pregnancy complications. J. Immunol. 192 : 11 4949– 56 [Google Scholar]
  • Johnson CSC , Shively CA , Michalson KT , Lea AJ , DeBo RJ et al. 2021 . Contrasting effects of Western vs Mediterranean diets on monocyte inflammatory gene expression and social behavior in a primate model. eLife 10 : e68293 [Google Scholar]
  • Kalinkovich A , Weisman Z , Greenberg Z , Nahmias J , Eitan S et al. 1998 . Decreased CD4 and increased CD8 counts with T cell activation is associated with chronic helminth infection. Clin. Exp. Immunol. 114 : 3 414– 21 [Google Scholar]
  • Kehrmann J , Effenberg L , Wilk C , Schoemer D , Ngo Thi , Phuong N et al. 2020 . Depletion of Foxp3+ regulatory T cells is accompanied by an increase in the relative abundance of Firmicutes in the murine gut microbiome. Immunology 159 : 3 344– 53 [Google Scholar]
  • Kinder JM , Jiang TT , Ertelt JM , Xin L , Strong BS et al. 2015 . Cross-generational reproductive fitness enforced by microchimeric maternal cells. Cell 162 : 3 505– 15 [Google Scholar]
  • Knochelmann HM , Dwyer CJ , Bailey SR , Amaya SM , Elston DM et al. 2018 . When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell. Mol. Immunol. 15 : 5 458– 69 [Google Scholar]
  • Kuehni CE , Strippoli M-PF , Low N , Silverman M. 2007 . Asthma in young South Asian women living in the United Kingdom: the importance of early life. Clin. Exp. Allergy 37 : 1 47– 53 [Google Scholar]
  • Kutzer MAM , Armitage SAO. 2016 . Maximising fitness in the face of parasites: a review of host tolerance. Zoology 119 : 4 281– 89 [Google Scholar]
  • Kuzawa CW , Adair L , Bechayda SA , Borja JRB , Carba DB et al. 2020 . Evolutionary life history theory as an organising framework for cohort studies: insights from the Cebu Longitudinal Health and Nutrition Survey. Ann. Hum. Biol. 47 : 2 94– 105 [Google Scholar]
  • La Rocca C , Carbone F , Longobardi S , Matarese G 2014 . The immunology of pregnancy: regulatory T cells control maternal immune tolerance toward the fetus. Immunol. Lett. 162 : 1 41– 48 [Google Scholar]
  • Lee GR. 2018 . The balance of Th17 versus Treg cells in autoimmunity. Int. J. Mol. Sci. 19 : 3 730 [Google Scholar]
  • Lee SC , Tang MS , Lim YAL , Choy SH , Kurtz ZD et al. 2014 . Helminth colonization is associated with increased diversity of the gut microbiota. PLOS Negl. Trop. Dis. 8 : 5 e2880 [Google Scholar]
  • Liu AH. 2015 . Revisiting the hygiene hypothesis for allergy and asthma. J. Allergy Clin. Immunol. 136 : 4 860– 65 [Google Scholar]
  • Lochmiller RL , Deerenberg C. 2000 . Trade-offs in evolutionary immunology: Just what is the cost of immunity?. Oikos 88 : 1 87– 98 [Google Scholar]
  • Lopata AL , Kleine-Tebbe J , Kamath SD. 2016 . Allergens and molecular diagnostics of shellfish allergy. Allergo J. Int. 25 : 7 210– 18 [Google Scholar]
  • Lynch NR , Hagel I , Perez M , Di Prisco MC , Lopez R , Alvarez N. 1993 . Effect of anthelmintic treatment on the allergic reactivity of children in a tropical slum. J. Allergy Clin. Immunol. 92 : 3 404– 11 [Google Scholar]
  • Macpherson AJ , Harris NL. 2004 . Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 4 : 6 478– 85 [Google Scholar]
  • Maes M. 2011 . Depression is an inflammatory disease, but cell-mediated immune activation is the key component of depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 35 : 3 664– 75 [Google Scholar]
  • Magid K , Chatterton RT , Ahamed FU , Bentley GR. 2018 . Childhood ecology influences salivary testosterone, pubertal age and stature of Bangladeshi UK migrant men. Nat. Ecol. Evol. 2 : 1146– 54 [Google Scholar]
  • Mahajan NN , Gajbhiye RK , Bahirat S , Lokhande PD , Mathe A et al. 2021 . Co-infection of malaria and early clearance of SARS-CoV-2 in healthcare workers. J. Med. Virol. 93 : 4 2431– 38 [Google Scholar]
  • Maizels RM. 2005 . Infections and allergy – helminths, hygiene and host immune regulation. Curr. Opin. Immunol. 17 : 6 656– 61 [Google Scholar]
  • Maizels RM , Lawrence RA. 1991 . Immunological tolerance: the key feature in human filariasis?. Parasitol. Today 7 : 10 271– 76 [Google Scholar]
  • Maizels RM , McSorley HJ. 2016 . Regulation of the host immune system by helminth parasites. J. Allergy Clin. Immunol. 138 : 666– 75 [Google Scholar]
  • Maizels RM , Yazdanbakhsh M. 2003 . Immune regulation by helminth parasites: cellular and molecular mechanisms. Nat. Rev. Immunol. 3 : 9 733– 44 [Google Scholar]
  • Malhotra I , Mungai PL , Wamachi AN , Tisch D , Kioko JM et al. 2006 . Prenatal T cell immunity to Wuchereria bancrofti and its effect on filarial immunity and infection susceptibility during childhood. J. Infect. Dis. 193 : 7 1005– 13 [Google Scholar]
  • Maloney S , Smith A , Furst DE , Myerson D , Rupert K et al. 1999 . Microchimerism of maternal origin persists into adult life. J. Clin. Invest. 104 : 1 41– 47 [Google Scholar]
  • Martin LB , Hawley DM , Ardia DR. 2011 . An introduction to ecological immunology. Funct. Ecol. 25 : 1 1– 4 [Google Scholar]
  • Martin LB II , Weil ZM , Nelson RJ. 2008 . Fever and sickness behaviour vary among congeneric rodents. Funct. Ecol. 22 : 1 67– 77 [Google Scholar]
  • Martin M , Blackwell AD , Gurven M , Kaplan H 2013 . Make new friends and keep the old? Parasite coinfection and comorbidity in Homo Sapiens . Primates, Pathogens, and Evolution JF Brinkworth, K Pechenkina 363– 88 New York: Springer [Google Scholar]
  • Martínez-Colón GJ , Ratnasiri K , Chen H , Jiang S , Zanley E et al. 2021 . SARS-CoV-2 infects human adipose tissue and elicits an inflammatory response consistent with severe COVID-19. bioRxiv. https://doi.org/10.1101/2021.10.24.465626 [Crossref]
  • Maurer D , Lewis TL , Mondloch CJ. 2005 . Missing sights: consequences for visual cognitive development. Trends Cogn. Sci. 9 : 3 144– 51 [Google Scholar]
  • May K , Grube M , Malhotra I , Long CA , Singh S et al. 2009 . Antibody-dependent transplacental transfer of malaria blood-stage antigen using a human ex vivo placental perfusion model. PLOS ONE 4 : 11 e7986 [Google Scholar]
  • Mbow M , Lell B , Jochems SP , Cisse B , Mboup S et al. 2020 . COVID-19 in Africa: dampening the storm?. Science 369 : 6504 624– 26 [Google Scholar]
  • McDade TW. 2003 . Life history theory and the immune system: steps toward a human ecological immunology. Yearb. Phys. Anthropol. 122 : S37 100– 25 [Google Scholar]
  • McDade TW. 2005a . The ecologies of human immune function. Annu. Rev. Anthropol. 34 : 495– 521 [Google Scholar]
  • McDade TW. 2005b . Life history, maintenance, and the early origins of immune function. Am. J. Hum. Biol. 17 : 1 81– 94 [Google Scholar]
  • McDade TW , Georgiev AV , Kuzawa CW. 2016 . Trade-offs between acquired and innate immune defenses in humans. Evol. Med. Public Health 2016 : 1 1– 16 [Google Scholar]
  • Medzhitov R , Schneider DS , Soares MP. 2012 . Disease tolerance as a defense strategy. Science 335 : 6071 936– 41 [Google Scholar]
  • Muehlenbein MP , Hirschtick JL , Bonner JZ , Swartz AM. 2010 . Toward quantifying the usage costs of human immunity: altered metabolic rates and hormone levels during acute immune activation in men. Am. J. Hum. Biol. 22 : 4 546– 56 [Google Scholar]
  • Natri H , Garcia AR , Buetow KH , Trumble BC , Wilson MA. 2019 . The pregnancy pickle: evolved immune compensation due to pregnancy underlies sex differences in human diseases. Trends Genet 35 : 7 478– 88 [Google Scholar]
  • Nemazee D. 2017 . Mechanisms of central tolerance for B cells. Nat. Rev. Immunol. 17 : 5 281– 94 [Google Scholar]
  • Nikolich-Žugich J. 2008 . Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat. Rev. Immunol. 8 : 7 512– 22 [Google Scholar]
  • Núñez-de la Mora A , Chatterton RT , Choudhury OA , Napolitano DA , Bentley GR. 2007 . Childhood conditions influence adult progesterone levels. PLOS Med 4 : 5 e167 [Google Scholar]
  • Olszak T , An D , Zeissig S , Vera MP , Richter J et al. 2012 . Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336 : 6080 489– 93 [Google Scholar]
  • Panda AK , Tripathy R , Das BK. 2020 . Plasmodium falciparum infection may protect a population from severe acute respiratory syndrome coronavirus 2 infection. J. Infect. Dis. 222 : 9 1570– 71 [Google Scholar]
  • Pandiyan P , Bhaskaran N , Zou M , Schneider E , Jayaraman S , Huehn J. 2019 . Microbiome dependent regulation of T regs and Th17 cells in mucosa. Front. Immunol. 10 : 426 [Google Scholar]
  • Panelli S , Epis S , Cococcioni L , Perini M , Paroni M et al. 2020 . Inflammatory bowel diseases, the hygiene hypothesis and the other side of the microbiota: parasites and fungi. Pharmacol. Res. 159 : 104962 [Google Scholar]
  • Parham P. 2015 . The Immune System New York: Garland Sci., Taylor & Francis Group. , 4th ed.. [Google Scholar]
  • Parker W , Sarafian JT , Broverman SA , Laman JD. 2021 . Between a hygiene rock and a hygienic hard place: avoiding SARS-CoV-2 while needing environmental exposures for immunity. Evol. Med. Public Health 9 : 1 120– 30 [Google Scholar]
  • Pawelec G , Derhovanessian E. 2011 . Role of CMV in immune senescence. Virus Res 157 : 175– 79 [Google Scholar]
  • Pentsuk N , van der Laan JW. 2009 . An interspecies comparison of placental antibody transfer: new insights into developmental toxicity testing of monoclonal antibodies. Birth Defects Res. B Dev. Reprod. Toxicol. 86 : 4 328– 44 [Google Scholar]
  • Permar SR , Moss WJ , Ryon JJ , Douek DC , Monze M , Griffin DE. 2003 . Increased thymic output during acute measles virus infection. J. Virol. 77 : 14 7872– 79 [Google Scholar]
  • Pontzer H. 2018 . Energy constraint as a novel mechanism linking exercise and health. Physiology 33 : 6 384– 93 [Google Scholar]
  • Råberg L , Sim D , Read AF. 2007 . Disentangling genetic variation for resistance and tolerance to infectious diseases in animals. Science 318 : 5851 812– 14 [Google Scholar]
  • Redman CWG , Sacks GP , Sargent IL. 1999 . Preeclampsia: an excessive maternal inflammatory response to pregnancy. Am. J. Obstet. Gynecol. 180 : 2 Pt. I 499– 506 [Google Scholar]
  • Rook G , Bäckhed F , Levin B , McFall-Ngai M , McLean A. 2017 . Evolution, human-microbe interactions and life history plasticity. Lancet 390 : 10093 521– 30 [Google Scholar]
  • Rook GAW. 2009 . Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis. Immunology 126 : 1 3– 11 [Google Scholar]
  • Rook GAW. 2012 . Hygiene hypothesis and autoimmune diseases. Clin. Rev. Allergy Immunol. 42 : 1 5– 15 [Google Scholar]
  • Rook GAW , Brunet LR. 2005 . Old friends for breakfast. Clin. Exp. Allergy 35 : 7 841– 42 [Google Scholar]
  • Saito S , Shima T , Nakashima A , Shiozaki A , Ito M , Sasaki Y. 2007 . What is the role of regulatory T cells in the success of implantation and early pregnancy?. J. Assist. Reprod. Genet. 24 : 9 379– 86 [Google Scholar]
  • Sansoni P , Vescovini R , Fagnoni F , Biasini C , Zanni F et al. 2008 . The immune system in extreme longevity. Exp. Gerontol. 43 : 2 61– 65 [Google Scholar]
  • Schneider-Crease IA , Blackwell AD , Kraft TS , Emery Thompson M , Maldonado Suarez I et al. 2021 . Helminth infection is associated with dampened cytokine responses to viral and bacterial stimulations in Tsimane forager-horticulturalists. Evol. Med. Public Health 9 : 1 349– 59 [Google Scholar]
  • Sear R. 2020 . Do human ‘life history strategies’ exist?. Evol. Hum. Behav. 41 : 6 513– 26 [Google Scholar]
  • Sheldon BC , Verhulst S. 1996 . Ecological immunology: costly parasite defences and trade-offs in evolutionary ecology. Trends Ecol. Evol. 11 : 8 317– 21 [Google Scholar]
  • Shivappa N , Steck SE , Hurley TG , Hussey JR , Hébert JR. 2014 . Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr 17 : 8 1689– 96 [Google Scholar]
  • Stearns SC. 1976 . Life-history tactics: a review of the ideas. Q. Rev. Biol. 51 : 1 3– 47 [Google Scholar]
  • Stearns SC. 1992 . The Evolution of Life Histories New York: Oxford Univ. Press [Google Scholar]
  • Stearns SC , Rodrigues AMM. 2020 . On the use of “life history theory” in evolutionary psychology. Evol. Hum. Behav. 41 : 6 474– 85 [Google Scholar]
  • Strachan DP. 1989 . Hay fever, hygiene, and household size. BMJ 299 : 6710 1259– 60 [Google Scholar]
  • Tai P , Wang J , Jin H , Song X , Yan J et al. 2008 . Induction of regulatory T cells by physiological level estrogen. J. Cell. Physiol. 214 : 2 456– 64 [Google Scholar]
  • Targonski PV , Jacobson RM , Poland GA. 2007 . Immunosenescence: role and measurement in influenza vaccine response among the elderly. Vaccine 25 : 16 3066– 69 [Google Scholar]
  • Tesmer LA , Lundy SK , Sarkar S , Fox DA. 2008 . Th17 cells in human disease. Immunol. Rev. 223 : 1 87– 113 [Google Scholar]
  • Thompson PA , Khatami M , Baglole CJ , Sun J , Harris SA et al. 2015 . Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 36 : S232– 53 [Google Scholar]
  • Thurber C , Dugas LR , Ocobock C , Carlson B , Speakman JR , Pontzer H. 2019 . Extreme events reveal an alimentary limit on sustained maximal human energy expenditure. Sci. Adv. 5 : 6 eaaw0341 [Google Scholar]
  • Tilg H , Kaser A. 2011 . Gut microbiome, obesity, and metabolic dysfunction. J. Clin. Investig. 121 : 6 2126– 32 [Google Scholar]
  • Tsegaye A , Wolday D , Otto S , Petros B , Assefa T et al. 2003 . Immunophenotyping of blood lymphocytes at birth, during childhood, and during adulthood in HIV-1-uninfected Ethiopians. Clin. Immunol. 109 : 3 338– 46 [Google Scholar]
  • Tyagi N , Farnell EJ , Fitzsimmons CM , Ryan S , Tukahebwa E et al. 2015 . Comparisons of allergenic and metazoan parasite proteins: allergy the price of immunity. PLOS Comput. Biol. 11 : 10 e1004546 [Google Scholar]
  • Urlacher SS , Blackwell AD , Liebert MA , Madimenos FC , Cepon-Robins TJ et al. 2016 . Physical growth of the shuar: height, weight, and BMI references for an indigenous Amazonian population. Am. J. Hum. Biol. 28 : 1 16– 30 [Google Scholar]
  • Urlacher SS , Ellison PT , Sugiyama LS , Pontzer H , Eick G et al. 2018 . Tradeoffs between immune function and childhood growth among Amazonian forager-horticulturalists. PNAS 115 : E3914– 21 [Google Scholar]
  • Velasquez-Manoff M. 2013 . An Epidemic of Absence: A New Way of Understanding Allergies and Autoimmune Diseases New York: Scribner [Google Scholar]
  • Wasko NJ , Nichols F , Clark RB 2020 . Multiple sclerosis, the microbiome, TLR2, and the hygiene hypothesis. Autoimmun. Rev. 19 : 1 102430 [Google Scholar]
  • Wells JCK , Cortina-Borja M. 2013 . Different associations of subscapular and triceps skinfold thicknesses with pathogen load: an ecogeographical analysis. Am. J. Hum. Biol. 25 : 594– 605 [Google Scholar]
  • West-Eberhard MJ. 2019 . Nutrition, the visceral immune system, and the evolutionary origins of pathogenic obesity. PNAS 116 : 3 723– 31 [Google Scholar]
  • White DW , Beard RS , Barton ES. 2012 . Immune modulation during latent herpesvirus infection. Immunol. Rev. 245 : 1 189– 208 [Google Scholar]
  • Wiley AS. 2021 . Pearl lecture: Biological normalcy: a new framework for biocultural analysis of human population variation. Am. J. Hum. Biol. 33 : 5 e23563 [Google Scholar]
  • Wiley AS , Cullin JM. 2020 . Biological normalcy. Evol. Med. Public Health 2020 : 1 https://doi.org/10.1093/emph/eoz035 [Crossref] [Google Scholar]
  • Wilson BC , Butler ÉM , Grigg CP , Derraik JGB , Chiavaroli V et al. 2021 . Oral administration of maternal vaginal microbes at birth to restore gut microbiome development in infants born by caesarean section: a pilot randomised placebo-controlled trial. EBioMedicine 69 : 103443 [Google Scholar]
  • Wiria AE , Hamid F , Wammes LJ , Prasetyani MA , Dekkers OM et al. 2015 . Infection with soil-transmitted helminths is associated with increased insulin sensitivity. PLOS ONE 10 : 6 e0127746 [Google Scholar]
  • Wiria AE , Sartono E , Supali T , Yazdanbakhsh M. 2014 . Helminth infections, type-2 immune response, and metabolic syndrome. PLOS Pathog . 10 : 7 e1004140 [Google Scholar]
  • Wolday D , Gebrecherkos T , Arefaine ZG , Kiros YK , Gebreegzabher A et al. 2021a . Effect of co-infection with intestinal parasites on COVID-19 severity: a prospective observational cohort study. EClinicalMedicine 39 : 101054 [Google Scholar]
  • Wolday D , Tasew G , Amogne W , Urban B , Schallig HDFH et al. 2021b . Interrogating the impact of intestinal parasite-microbiome on pathogenesis of COVID-19 in Sub-Saharan Africa. Front. Microbiol. 12 : 614522 [Google Scholar]
  • Wolfs IMJ , Stöger JL , Goossens P , Pöttgens C , Gijbels MJJ et al. 2014 . Reprogramming macrophages to an anti-inflammatory phenotype by helminth antigens reduces murine atherosclerosis. FASEB J 28 : 1 288– 99 [Google Scholar]
  • Yazdanbakhsh M , Kremsner PG , van Ree R. 2002 . Allergy, parasites, and the hygiene hypothesis. Science 296 : 5567 490– 94 [Google Scholar]
  • Ye Q , Wang B , Mao J 2020 . The pathogenesis and treatment of the ‘cytokine storm’ in COVID-19. J. Infect. 80 : 6 607– 13 [Google Scholar]
  • Yudkin JS , Stehouwer CD , Emeis JJ , Coppack SW. 1999 . C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose. Aterioscler. Thromb. Vasc. Biol. 19 : 4 972– 78 [Google Scholar]

Data & Media loading...

  • Article Type: Review Article

Most Read This Month

Most cited most cited rss feed, ethnography in/of the world system: the emergence of multi-sited ethnography, the politics and poetics of infrastructure, migrant “illegality” and deportability in everyday life, poetics and performances as critical perspectives on language and social life, language and agency, refugees and exile: from "refugee studies" to the national order of things, parks and peoples: the social impact of protected areas, language ideology, think practically and look locally: language and gender as community-based practice, the local and the global: the anthropology of globalization and transnationalism.

  • - Google Chrome

Intended for healthcare professionals

  • My email alerts
  • BMA member login
  • Username * Password * Forgot your log in details? Need to activate BMA Member Log In Log in via OpenAthens Log in via your institution

Home

Search form

  • Advanced search
  • Search responses
  • Search blogs
  • Hay fever, hygiene,...

Hay fever, hygiene, and household size.

  • Related content
  • Peer review

PDF extract preview

This is a PDF-only article. The first page of the PDF of this article appears above.

hygiene hypothesis original paper

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • SAGE - PMC COVID-19 Collection

Logo of phesage

Time to abandon the hygiene hypothesis: new perspectives on allergic disease, the human microbiome, infectious disease prevention and the role of targeted hygiene

Sally f bloomfield.

London School of Hygiene & Tropical Medicine and International Scientific Forum on Home Hygiene, The Old Dairy Cottage, Montacute, Somerset TA15 6XL, UK

Graham AW Rook

Centre for Clinical Microbiology, Department of Infection, University College London (UCL), London, UK

Elizabeth A Scott

Center for Hygiene and Health, Department of Biology, Simmons College, Boston, MA, USA

Fergus Shanahan

APC Microbiome Institute, University College Cork – National University of Ireland, Cork, Ireland

Rosalind Stanwell-Smith

London School of Hygiene & Tropical Medicine, London, UK

Paul Turner

Section of Paediatrics (Allergy & Infectious Diseases) and MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK; Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, NSW, Australia

To review the burden of allergic and infectious diseases and the evidence for a link to microbial exposure, the human microbiome and immune system, and to assess whether we could develop lifestyles which reconnect us with exposures which could reduce the risk of allergic disease while also protecting against infectious disease.

Using methodology based on the Delphi technique, six experts in infectious and allergic disease were surveyed to allow for elicitation of group judgement and consensus view on issues pertinent to the aim.

Key themes emerged where evidence shows that interaction with microbes that inhabit the natural environment and human microbiome plays an essential role in immune regulation. Changes in lifestyle and environmental exposure, rapid urbanisation, altered diet and antibiotic use have had profound effects on the human microbiome, leading to failure of immunotolerance and increased risk of allergic disease. Although evidence supports the concept of immune regulation driven by microbe–host interactions, the term ‘hygiene hypothesis’ is a misleading misnomer. There is no good evidence that hygiene, as the public understands, is responsible for the clinically relevant changes to microbial exposures.

Conclusion:

Evidence suggests a combination of strategies, including natural childbirth, breast feeding, increased social exposure through sport, other outdoor activities, less time spent indoors, diet and appropriate antibiotic use, may help restore the microbiome and perhaps reduce risks of allergic disease. Preventive efforts must focus on early life. The term ‘hygiene hypothesis’ must be abandoned. Promotion of a risk assessment approach (targeted hygiene) provides a framework for maximising protection against pathogen exposure while allowing spread of essential microbes between family members. To build on these findings, we must change public, public health and professional perceptions about the microbiome and about hygiene. We need to restore public understanding of hygiene as a means to prevent infectious disease.

Introduction

Allergic diseases including asthma, hay fever, eczema and food allergies have dramatically increased over the last century, initially in high-income communities but now elsewhere. At the same time, threats of infectious disease pandemics, antibiotic resistance and numbers of immune-compromised people living in the community have increased. Taken together, these diseases are a significant burden on health and prosperity.

The idea that there might be a link between the rise in allergic disease and reduced microbial exposure as a result of measures introduced to protect against infection was first proposed in 1989. 1 , 2 This so-called hygiene hypothesis, as outlined by Dr David Strachan, proposed that a lower incidence of infection in early childhood could be an explanation for the 20 th century rise in atopic diseases. Although a simple idea in itself, it raised the thought that rising allergies may be an inevitable price to be paid for freedom from the burden of killer infectious diseases. Although evidence still supports the concept that immune regulation is driven by microbe–host interactions, the term ‘hygiene hypothesis’ is now being seen by many as a misleading misnomer for a concept with far-reaching consequences for public health and an issue which needs to be addressed. 3 , 4

Humans are ecosystems, where the microbes that live on and within us (the human microbiome) constitute an organ at least as essential to health as our liver or kidneys. 5 The immune system is a learning device, and at birth it resembles a computer with hardware and software but few data. Additional data must be supplied during the first years of life, through contact with microorganisms from other humans and the natural environment. If these inputs are inadequate or inappropriate, the regulatory mechanisms of the immune system can fail. As a result, the system attacks not only harmful organisms which cause infections but also innocuous targets such as pollen, house dust and food allergens resulting in allergic diseases.

Despite this new understanding, the hygiene hypothesis concept – that we have become too clean – still persists in the minds of the public. As a result, the public has lost confidence in hygiene. This is happening at a time when infectious disease issues mean that hygiene is becoming more, rather than less, important.

The aim of this study is to review the burden of allergic and infectious diseases and the evidence for a link to microbial exposure, the human microbiome and immune system. Also, it is to assess whether and to what extent we could develop lifestyles which reconnect us with exposures and thereby reduce the risks of allergic disease while also protecting against infectious disease.

Using methodology based on the Delphi technique, 6 – 9 six experts in infectious and allergic disease were surveyed to allow for elicitation of group judgement in order to arrive at a consensus view on issues pertinent to the aim of the study.

Key themes emerged: first, the extent of the health burden of allergic and hygiene-related diseases; second, the most recent evidence regarding the nature of the link between reduced microbial exposure and its impact on the human microbiome and the immune regulatory system; third, the question of relationship between lifestyles and protection against infectious diseases. The Delphi technique is a qualitative research method that relies on the judgement of individuals presumed to be knowledgeable and expert at what they do. When a sufficient degree of consensus is achieved, the Delphi process is curtailed and the resulting judgement is published. Six experts in infectious diseases and allergies were invited to participate, and the issues to be addressed were agreed via online communication. The authors participated in a conference in which each presented evidence related to their area of expertise. Following this, authors submitted a written contribution. These were analysed and key themes were integrated into a paper which was made available online to all authors for review. This included further questions soliciting the author’s views. After further rounds of questions and revision, a consensus position was obtained.

Why hygiene is important in the 21 st century

In the 1950s and 1960s, there was optimism that, with vaccination and antibiotics freely available, conquest of most infections would follow. During the last four decades, this opinion has been reversed. Infectious disease continues to exert a heavy burden on health and prosperity. The various infectious disease issues are most often considered in isolation, but when viewed together, they represent a powerful argument for renewed emphasis on hygiene, which alongside vaccination strategies remain key to containing infectious disease. 10

During the 1980s, there was a rapid increase in reported cases of food poisoning in the United Kingdom, particularly related to Salmonella and Campylobacter. 11 Although reported cases have somewhat declined, food, waterborne, and non-food-related infectious intestinal diseases (IIDs) remain at unacceptable levels. The latest study of IID (food and non-foodborne IID) reported that the true incidence in the community is 43% higher than in the mid-1990s: this study estimated 17 million cases a year in the United Kingdom. 12 The estimated cost of food-related IID is £1.5 billion a year, including resource and welfare losses. 12 Norovirus, mainly spread from person-to-person, is the most significant cause of intestinal infections in the developed world, including 3 million cases per year in the United Kingdom. 12

Evidence shows that respiratory hygiene involving hands and surfaces can limit spread of respiratory infections, particularly colds, and also influenza. 13 – 15 Since respiratory and intestinal viral infections are not treatable by antibiotics, prevention through hygiene is key.

In developed countries, about 7% of inpatients acquire an infection in hospital. 16 Recent figures show a decline in health-care-associated infection (HCAI), in the United Kingdom, particularly of Clostridium difficile and MRSA (methicillin-resistant Staphylococcus aureus ), 17 , 18 while other causes of HCAI have emerged, including new epidemic strains of Escherichia coli, Pseudomonas spp. and viruses.

Governments, looking at prevention as a means to reduce health spending, have introduced shorter hospital stays and increased homecare. This requires new policies to prevent HCAIs in community settings 19 where there is no evidence of a decline. Until recently, most episodes of C. difficile infection were believed to result from acquisition in health-care settings. There is now increasing evidence of multiple other potential sources, including asymptomatic patients, and sources in the wider environment, such as water, farm animals or pets, and food. 20 The contribution of cases acquired from these sources to the overall burden of disease is unclear, particularly with concerns about increased community-associated C. difficile infection. 21

Societal changes mean that people with greater susceptibility to infectious disease make up an increasing proportion of the population, up to 20% or more. 10 The largest proportion comprises the elderly who have reduced immunity, often exacerbated by other illnesses. It also includes the very young and family members with invasive devices such as catheters and people whose immuno-competence is impaired as a result of chronic and degenerative illness (including HIV/AIDS) or drug therapies such as cancer chemotherapy.

Emerging pathogens and new strains are a significant concern. It is remarkable that norovirus, Campylobacter and Legionella were largely unknown as human pathogens before the 1970s, with others such as E. coli O157 and O104 emerging in subsequent decades. It is now thought likely that we shall identify many more, the latest being Zika virus. 22 Agencies worldwide recognise that for threats such as new influenza strains, SARS (severe acute respiratory syndrome) and Ebola, hygiene is a first line of defence during the early critical period before mass measures such as vaccination become available. 23 The low infectious dose observed for several of the emerging pathogens, such as E. coli O157:H7 and norovirus, is an additional concern that emphasises the role that hygiene can play in prevention. 24 , 25

Antibiotic resistance is a global priority. 26 Hygiene addresses this problem by reducing the need for antibiotic prescribing and reducing ‘silent’ spread of antibiotic resistant strains in the community and hospitals. 27 As persistent nasal or bowel carriage of these strains spreads in the healthy population, this increases the risk of infection with resistant strains in both hospitals and the community. 27

Infections can act as co-factors in diseases, such as cancer and chronic degenerative diseases. Syndromes such as Guillain–Barré 28 and triggering of allergy by viral infections 29 add to the burden of hygiene-related infection.

The rise of allergies in the 20 th century

While infectious disease and hygiene have been key public health issues for centuries, 30 allergic diseases have only relatively recently been regarded as a significant health burden. The marked increase in prevalence of allergic diseases, such as eczema, 31 allergic rhinitis 32 and food allergy, 32 has been a prominent trend over the past century in all regions of the world, but most characterised in Western countries. 33 While this is frequently presented as an ‘epidemic’, epidemiological data indicate the situation is more complex. As highlighted by Platts-Mills 34 ( Figure 1 ), the ‘spikes’ in prevalence of allergic rhinitis, asthma and food allergy have occurred at different times in the past 120 years, and thus different atopic diseases may have different contributing factors. Indeed, there are emerging data that in some areas (mostly in ‘Western’ countries) these increases may have plateaued and even begun to subside. 34 A further issue is that at least for food allergy, prevalence may have been overestimated, depending on the methodology used. Venter et al. 35 assessed the rate of challenge-positive food allergy in three birth cohorts on the Isle of Wight (UK) between 1989 and 2002. A major finding of this study, confirmed in other reports, is that rates of parent-reported allergy were significantly higher (33%) than those confirmed by placebo-controlled food challenge (6%) (the accepted gold standard for diagnosis). For peanut allergy, the same study reported a rate of 1 in 200 children aged 3–4 years in 1989, increasing to 1 in 70 in the mid-1990s, but plateauing thereafter. A 2016 UK intervention study, in children breast-fed to at least 6 months of age, reported a rate of 1 in 40. 36 Of note, the development of an inappropriate immune response to foods (‘sensitisation’), which occurs before onset of clinical disease, is an early event often occurring in the first few months of life. 36

An external file that holds a picture, illustration, etc.
Object name is 10.1177_1757913916650225-fig1.jpg

Trends in allergic disease

Reprinted from Platts-Mills, 34 Copyright (2015), with permission from American Academy of Allergy, Asthma & Immunology and Elsevier

Perhaps the relatively late appearance of food allergy over the past few decades is a consequence of a progression from allergic airways disease (hay fever, asthma) in parents to a more severe clinical phenotype (food allergy) in their offspring. 37 However, a compelling alternative is the interaction between genetic predisposition and environmental influences, particularly for food allergy, where immune sensitisation to foods may originate with exposure to food allergens in the environment through the skin, a situation exacerbated by eczema and reduced skin barrier function. 36 At the same time, there have been changes in how foods are consumed (e.g. roasted peanut, as consumed in Europe and North America, is more allergenic than raw or other forms of processed peanut).

From Hygiene Hypothesis to Old Friends Mechanism

Building on the significant amount of research published since 1989, a number of refinements to the original hygiene hypothesis now seem to offer more plausible explanations. The Old Friends (OF) Mechanism was proposed by Rook in 2003 and argues that the vital microbial exposures are not colds, measles and other childhood infections (the crowd infections), but rather microbes already present during primate evolution and in hunter-gatherer times when the human immune system was evolving. 38 – 40 OF microbes include environmental species which inhabit indoor and outdoor environments, and the largely non-harmful commensal microbes acquired from the skin, gut and respiratory tract of other humans. In evolving humans, before the advent of modern medicine, the OF also included organisms such as helminths, Helicobacter pylori , and hepatitis A virus that could persist for life in hunter-gatherer groups and that needed to be tolerated. They all therefore activated immunoregulatory mechanisms, 38 but few experts believe that they need to be replaced or even that there is any feasible way of doing so.

Whereas the hygiene hypothesis implicated childhood virus infections as the vital exposures, from an evolutionary point of view this was never likely. Crowd infections were not part of human evolutionary experience because they either kill or induce solid immunity, so could not persist in small hunter-gatherer groups. 41 Epidemiological studies carried out in Finland, Denmark and the United Kingdom now confirm that childhood infections do not protect against allergic disorders. 42 – 44

Studies show how OF exposures are vital because they interact with the regulatory systems that keep the immune system in balance and prevent overreaction, which is an underlying cause of allergies. Diversity of microbial exposure is key. First, a large experience of harmless bacteria and archaea during infancy, when immunoregulatory systems are being established, increases the repertoire of organisms that can be tolerated. Second, since all life-forms are ultimately constructed from similar building blocks, exposed individuals acquire some memory lymphocytes that recognise novel pathogens or even novel viruses. 45

What are the likely causes of reduced or altered microbial exposures

In order to look for strategies which might restore the necessary microbial exposures, it is first necessary to understand the underlying causes of the loss of exposure. Since allergic diseases are largely conditions of the last 100 years, an obvious assumption is that the sanitary revolution is a root cause. The latter part of the 19 th century saw radical improvements in sanitation, cleaner food and water, clean-up of cities, and rapid decline in infectious diseases. 46 However, it is likely that these changes also inadvertently reduced exposure to OF microbes which occupy the same habitats. Since the major changes in water, sanitation and hygiene had occurred by 1920, it is difficult to ascribe the massive changes in the asthma prevalence from 1960 onwards to these changes. 34

It is now clear that the most important times for OF exposure are early in development, during pregnancy, delivery, and the first few days or months of infancy. 47 , 48 A 2008 review of epidemiological studies show that Caesarean section is linked to increased risk of allergy. 49 C-sections have become increasingly common since 1950 and now account for 25% of UK births. 50 Furthermore, transfer of microbiota occurs via the mother’s milk, which is not sterile. 51 Breast versus bottle feeding has a large influence on gut microbiome, 52 , 53 but further studies are needed to confirm any association with allergic disease. In high-income settings, there is likely to be a trans-generational effect where each generation receives a more impoverished microbiota, and essential microbiota are lost from the community. 54

Continuing early-life exposure from the mother and siblings is also important. 55 , 56 Studies show that children from large families are at lower risk of developing allergies. 52 , 57 Exposure to pets protects against allergies, 58 , 59 although domestic animals in the home have increased rather than decreased. 60 People seem to share their microbiota via dogs, 61 which greatly increase the microbial biodiversity of the home. 62 , 63

There is good evidence that contact with microbial diversity from the natural environment is crucial. Numerous studies now show that exposure to farm environments during the first 2–3 years of life protects against allergic disorders 64 – 66 and correlates with microbial biodiversity in the air 67 and the home. 68 Animal models show candidate organisms from these environments protect against allergic disorders. 69 Studies in Finland show that living close to green space and agriculture rather than close to a town increases biodiversity of the skin microbiota and correlates with reduced allergic sensitisation. 70 Urbanisation has accelerated loss of exposure to the natural environment. In the United Kingdom, 82% of the population now live in urban areas, 71 with up to 90% of our time spent indoors. 72

Although research has tended to focus on the gut microbiome, it seems likely that the microbiome of skin and airways is also involved. 73 – 75 Much of the exposure obtained from outdoor environments is likely to be via the airways. The air contains bacteria, archaea, viruses, fungi, spores, pollen, plant biomass and dust. Depending on the environment and on degree of exertion, the number of bacteria/archaea breathed in could vary between about 10 6 and 10 10 in 24 h. A proportion of these will be retained in the airways, and recent work reveals that exposure to bacterial components causes increased expression of a protein that inhibits inflammation. 73 , 74 Gut exposure is also mediated via the airways where ciliary action brings about transfer to the gut. The likelihood that skin microbiota are OF microbes is indicated by studies showing that Acinetobacter species in skin protect against allergy. 75

Factors that maintain the gut microbiota

Once the microbiome has been acquired and evolved during childhood, 48 the critical question becomes what factors maintain optimum composition and biodiversity, because loss of biodiversity is strongly associated with disease states, inflammation and decline. 76 – 78

Increasingly, the answer appears to be that the optimal composition of the microbiota is maintained by diet, 79 which needs to be diverse, and contain fibre (polysaccharides digested by the microbiota rather than the human host), 80 and polyphenols found in plant products. 81 – 83 A diet deficient in fibre can lead to progressive extinctions of important groups of organisms, 54 which are cumulative and increasingly difficult to reverse in subsequent generations. 42 Polyphenols and also fish oils also appear to modulate the composition of the microbiota. 84 , 85

Citizens of high-income countries have less diverse microbiota than do hunter-gatherers. 77 – 79 Other studies show that the elderly living in the community with healthy diets 78 have higher gut microbiota diversity than those in long-stay residential care who have a less diverse diet. Studies in Sweden and Denmark show that reduced gut microbiota diversity in infants is associated with increased risk of allergic disease in childhood. 86 – 88

Introduction of antibiotics in the 1950s and subsequent prescribing trends, show a compelling temporal fit with rising allergies since the 1970s. A 2014 review of evidence from over 50 epidemiological studies shows a reasonably consistent relationship between excessive antibiotic use, particularly in early childhood, and increased risk of allergic disease. 89 Evidence showing that exposure to antibiotics during pregnancy increases the risk of allergic disorders in infants 90 , 91 has been further confirmed in recent studies. 92 , 93 Antibiotics, particularly macrolides, have lasting effects on the microbiota of young children and increase risks of asthma. 92 This mirrors effects documented in animal models, where early disruption of gut microbiota causes long-term damage to metabolic regulation. 94

Disruptions of maternal microbiota diversity by antibiotics or inadequate diet are found to be transmitted to future generations. 54

Domestic and personal hygiene

Of all the trends that might explain declining OF exposure, one of the weakest is the popular notion of ‘being too clean in our own homes’. If this factor contributes, its role is likely to be small relative to other factors. An explosion of data, obtained using high-throughput RNA sequencing of samples from US homes, suggests that modern homes are ‘teeming with microbes’. It also suggests that the bacterial communities found in the home relate to the people and domestic animals living there and the food they eat, together with input from the local outdoor environment. 63 , 95

Microbiological studies in westernised homes indicate that routine daily or weekly cleaning habits (even involving use of antibacterial cleaners) have no sustained effect on levels of microbes in our homes. 96 – 98 The idea that we could create ‘sterile’ homes through excessive cleanliness is implausible; as fast as microbes are removed, they are replaced, via dust and air from the outdoor environment, and commensal microbes shed from the human body and our pets, and contaminated foods brought into the homes. Strachan’s 1 1989 proposition that ‘ higher standards of personal cleanliness ’ could also contribute to reduced exposure to essential microbes may be compatible with increased bathing/showering/shampooing since around 1950s, 46 but although bathing and so on removes large numbers of microbes from the skin, these are rapidly replaced.

Although data from westernised homes suggest that more diverse communities can be found on less-cleaned surfaces (TV screen, door trims, floors) than regularly cleaned surfaces (cutting board, kitchen surface, toilet seat), 63 , 95 to date, there is no confirmed evidence of a link between personal or home cleanliness and increased risk of allergic disease. In a German birth cohort study of 399 families, personal cleanliness (e.g. handwashing and showering) was associated with lower levels of endotoxin and muramic acid (bacterial markers) in bedding and floor dust. In comparison, household cleanliness (e.g. cleaning floors and bathrooms, dusting, and changing towels) was associated with less dust but not with lower microbial marker levels. Endotoxin in infancy was associated with less allergic sensitisation and less asthma when these children reached school age, whereas muramic acid exposure at school age, but not infancy, was associated with less school-age asthma and eczema. 99 It might seem surprising that neither personal nor home cleanliness activities were directly associated with allergy outcomes, but Liu 100 suggests that this may reflect the importance of early-life timing of microbial exposures and not cleanliness behaviours, with the influence of endotoxin exposure being in infancy. A 2002 data analysis of UK children born in 1991/1992 found association between parent-reported frequency of hand and face washing, showering and bathing at 15 months and wheezing and atopic eczema at 30–42 months, but this association was not reported in other studies. 101 , 102

The key point may be that the microbial content of modern urban homes has altered relative to earlier generations, not because of home and personal cleanliness but because, prior to the 1800s, people lived in predominantly rural surroundings. Also, although human gut and skin microbiota are constantly shed from family members, it is likely that exposure has altered reflecting the reduced diversity of the human microbiota due to factors described above. This means we now interact with an altogether different and less diverse mix of microbes.

Other factors also argue against the role of hygiene. Hygiene is irrelevant to microbiome disruption through altered diet and antibiotics. Also, if contact with the natural environment and microbial components of house dust occurs mostly via the airways, hygiene and cleanliness is unlikely to be responsible for reduced inputs from this key source.

Communicating Microbiome Science to Society – Prelude to Reversing Immunoallergic Disorders

Although evidence suggests that strategies such as promoting natural childbirth and breast feeding, increased social exposure through sport, other outdoor activities, less time spent indoors, diet and appropriate antibiotic use could help restore the microbiome and perhaps reduce risks of allergic disease, clinical and other evaluations are required to establish whether and to what extent this might occur and when intervention is most beneficial.

There is a window of time when the developing microbiome is critical for the education of the maturing immune system. Disruption or delay in acquisition of the microbiome in the first few years of life may predispose to later immune dysfunction. It follows that preventive efforts against immuno-allergic disorders must be focussed on early life events. Attempts to correct abnormal host–microbe interactions, once immunological events which lead to allergic disease are established, may be too late. These issues are further discussed by Shanahan and colleagues. 103 – 105 Gaps in understanding host–microbe interactions will be addressed as research continues, and one can anticipate a time, when optimal conditions for colonisation of the newborn are understood and can be controlled by strategies ensuring neonates begin life with a robust and diverse microbiota. In the interim, there is much that can be achieved by education and behaviour change, based on current information.

Several factors seem to conspire to limit effective communication of microbiome science to society ( Table 1 ). Some elements within the popular media do disservice to their readership. Examples include mis-representation of the role of hygiene and cleanliness, failure to clarify that probiotics are not all the same, and failure to probe unsubstantiated health claims or address seemingly complex concepts in detail. Fault also lies elsewhere ( Table 1 ). In contrast to policy makers and public health officials, clinicians deal with individual patients, not populations. Unless concerns about antibiotic usage are brought to an individual level, with emphasis on the consumer rather than the prescriber, reform initiatives will have limited impact. Patients are less likely to demand antibiotics if provided with information on the impact of such agents on the microbiota and the risk of immune disorders in later life. 106

Science and society – communication barriers

The stakeholdersChallenge
MediaPreoccupation with sensationalism rather than truth
Over-simplification and mis-portrayal of concepts such as hygiene, probiotics, and microbiota
Assumption that the readership cannot understand complex concepts
Medical/clinical professionalsLagging behind the science
Medical curricula focus on threat of infection rather than benefits of indigenous microbiota
Inadequately equipped to address patients’ questions on information acquired from the media
Inadequate response by the medical establishment to inaccurate and misleading material presented in the media
ScientistsExcessive use of hyperbole
Poor language
Failure to standardise terminology and methodology
Policy makers and Public health officialsIneffective and mixed messages to the public
Excessive focus on antibiotic resistance rather than risk of collateral damage to microbiota in promoting judicious use of antibiotics
Poor communication of influence of diet on microbiota
Lay publicPoor conceptualisation of risk versus benefit
Inadequately served by media for appraisal of medical and scientific claims

Promotion of breast feeding is lacking in precise rationale for modern women. Breast-feeding mothers need to know they are promoting a lifelong healthy microbiota for their offspring. Since the neonate acquires its microbiome primarily from its mother, greater attention needs to be paid to the mother’s diet, faecal and vaginal microbiome. Increasing awareness of the importance of the microbiome and the factors which sustain or disrupt it should be part of antenatal education.

Microbiome science already provides a glimpse of how the microbiota may be preserved or restored, including development of smart antibiotics, 107 non-antibiotic anti-microbials, microbial transplants, microbial consortia or single strains, and use of personalised biomarkers of disease risk prediction. 108 , 109 Restoration of the microbiome by vaginal microbiota transplants in C-section infants has been demonstrated, 110 albeit of unproven long-term benefit and controversial. 111 In addition, the molecular basis by which bifidobacteria engage with the host immune system is emerging; 112 , 113 this is important because such organisms are a predominant component of the microbiota in neonates.

Because of the multiplicity of factors involved, strategies to preserve or manipulate the microbiota will probably require a personalised approach tailored to individual genetics and lifestyle factors. 109

Developing and Promoting A Targeted Approach to Hygiene in Home and Everyday Life

Over the last 20 years or so, for reasons outlined above, there has not only been a revival of concern about infection and the role of hygiene 10 , 114 but also a realisation that the ‘scrupulous cleanliness’ approach advocated by Florence Nightingale 115 is no longer appropriate. If, as this review suggests, allergic diseases are not the price we have to pay for protection against infection, this is good news for hygiene. However, if we are to maximise protection against infection while at the same time sustaining exposure to essential microbes, we need a revised approach to hygiene based on current scientific evidence.

The International Scientific Forum on Home Hygiene (IFH) ( http://www.ifh-homehygiene.org ) was established in 1997 with the aim of developing and promoting a more effective approach to hygiene, based on scientific principles and the growing database of evidence about pathogen transmission. 116 To achieve this, IFH adopted the principle of targeted hygiene. 117 Targeted Hygiene is based on a four-step risk assessment requiring identification of the sources and reservoirs of pathogens, the routes of transmission, the critical control points, and appropriate hygiene interventions.

Targeted hygiene is based on the chain of infection transmission ( Figure 2 ) which shows that pathogenic organisms are continually shed into the environment from sources such as human occupants, pets and raw foods. 118

An external file that holds a picture, illustration, etc.
Object name is 10.1177_1757913916650225-fig2.jpg

The chain of infection transmission in the home

To get from an infected source to another individual, pathogens use well defined routes. Sampling studies record the presence of non-pathogenic bacteria and bacteria and viruses of medical interest on environmental surfaces in home and community settings, and laboratory and field studies have evaluated the rates of transfer of viral and bacterial pathogens via hands and common touch surfaces. 116 These demonstrate that the critical control points for transmission of infection are the hands, hand contact surfaces, food contact surfaces, and cleaning utensils and that these present the highest risk of transmission ( Figure 3 ).

An external file that holds a picture, illustration, etc.
Object name is 10.1177_1757913916650225-fig3.jpg

Ranking of sites and surfaces based on risk of transmission of infection

Equally important considerations are the interventions used to eliminate pathogens from critical control points before they spread further. This is important since inadequate procedures can increase transmission. 119 – 123 Hygienic (as opposed to visible) cleaning of hands, surfaces, fabrics and so on can be achieved by the following:

  • Physical removal of pathogens from inanimate or skin surfaces using soap or detergent-based cleaning. To be effective as a hygiene measure, this should be accompanied with thorough rinsing under running water, such that pathogens are not further disseminated.
  • Using an antimicrobial product (disinfectants or alcohol hand sanitisers) or processes (heat) that inactivate pathogens in situ . Antimicrobials are required where adequate removal is not possible by wiping/cleaning and/or rinsing alone, or in situations of higher risk. 124
  • Combined action, for example, laundering, where physical removal is combined with inactivation by heat together with an oxygen bleach–based laundry product.

While it is difficult to quantify the impact, evidence suggests that targeted hygiene reduces spread of infection. A review of evidence published between 1980 and 2001 concluded that the strength of the association between hygiene in the community and infections, as measured by the relative reduction in risk of illness by one or more hygiene measures (including handwashing), was generally greater than 20%. 125 A meta-analysis of community studies showed that improvements in hand hygiene alone resulted in reductions in gastrointestinal and respiratory illness of 31% and 21%, respectively. 126

Changing hygiene behaviour, however, requires changing public perceptions about hygiene, most particularly that hygiene is different from cleanliness, that is, more than just absence of dirt. Hygiene is what we do in the places and at the times that matter (hand, food, toilet and respiratory hygiene, health care, etc.) to protect against infection.

Communication and social marketing campaigns are now being evaluated and used as a means to achieve behaviour change mainly (but not exclusively) in relation to food and respiratory hygiene. These campaigns, however, focus on changing behaviours rather than changing understanding and dispelling misconceptions. 13 , 127 – 130 The e-bug project is a Europe-wide initiative aimed at ensuring all children leave school with an understanding of targeted hygiene. 131 An important feature of this teaching resource is that it is based on understanding infection and how it is transmitted.

The evidence reviewed in this study reflects the significant shift in thinking in the last 25 years. It shows that the interaction of the OF microbes which inhabit the natural environment and human microbiome with our immune system plays an essential role in immune regulation, promoting a tolerising milieu for the immune system which may impact against the development of allergic disease. Changes in lifestyle and environment, along with rapid urbanisation, have all contributed to changes in our exposure to essential microbes. 132 In addition, altered diet and excessive antibiotic use have also sustained detrimental effects on the content and diversity of the human microbiome. Together, these factors have had profound effects on the immune system, which are likely to have contributed to the onset of allergic disease.

By contrast, the public idea that obsessive hygiene and cleanliness is the root cause of the rise in allergies is no longer supported. Data show that relevant microbial exposures are almost entirely unrelated to hygiene as the public understands it. This is partly because sustaining the human microbiome through diet and avoiding excessive antibiotic usage are factors entirely unrelated to hygiene.

As far as understanding strategies which may reduce the risk of allergic disease, work is progressing fast, but there is still a long way to go. The multiple factors involved (including those not directly associated with microbiome interactions (allergen exposure, genetic, pollution, etc.)) make it impossible to assess the contribution of each factor. It is likely that success will only be achieved through combined effects of lifestyle changes, together with improved diet and reduced antibiotic prescribing. Nevertheless, data are now strong enough to encourage changes, such as encouraging natural childbirth, physical interaction between siblings and non-siblings, more sport and other outdoor activities (including babies in prams), and less time spent indoors, and reduced antibiotic consumption.

This review further supports the view that the term ‘hygiene hypothesis’ is a misleading and dangerous misnomer which needs to be abandoned in favour of a more appropriate term such as the OF Mechanism. However, in order to tackle both allergy and infection issues we also need to develop a smarter approach to hygiene. Although targeted hygiene was developed to optimise protection against infection, it provides a framework for maximising protection against pathogen exposure but, at the same time, minimising disturbance of the indoor microbiome and spread of essential microbes between family members.

As summarised in Table 1 , if we want to take advantage of these new findings, we first have to change public, public health and professional perceptions about the microbiome and about hygiene. Unstructured and conflicting advice and vague health warnings in the consumer and professional media must be replaced with simple clear mechanistic explanations and consistent messages using consistent terminology which avoids the use of the term ‘hygiene hypothesis’ to define the concept of a link between microbial exposure and allergies. Recent media articles which promote unsubstantiated suggestions that reduced handwashing could be a means to build and sustain a diverse gut microbiome are in direct conflict with public health agency advice on handwashing which is identified as probably the most important ‘critical control point’ for preventing spread of infection in all settings. 133 , 134

An underlying problem that needs addressing is that, both nationally and internationally there are no lead agencies which take ownership of hygiene promotion, looking at it from the point of view of the public at large and what they need to understand and know. Campaigns targeting food or respiratory, pet or health-care hygiene are developed by different agencies, often with conflicting messages. They also do little to address public misunderstandings about how infections are transmitted, the difference between hygiene, cleanliness and dirt, the widespread misuse of the term ‘germs’, and the hygiene hypothesis misnomer. 135

The imperative to understand and reverse the epidemiologic trends in allergic and immune-mediated disorders relates not solely to the personal suffering and health-care burden in the developed world. Without urgent effective intervention, such trends will be replicated around the globe as societies undergo socio-economic development. 105

Funding: P.J.T. holds a Clinician Scientist Award from the UK Medical Research Council (reference MR/K010468/1) and is supported through the National Institute for Health Research (NIHR)/Imperial Biomedical Research Centre. F.S. is a founder shareholder in Atlantia Food Clinical Trials, Tucana Health, and Alimentary Health Ltd. He is director of the APC Microbiome Institute, a research centre funded in part by Science Foundation Ireland (APC/SFI/12/RC/2273) and which has recently been in receipt of research grants from the following companies: Abbvie, Alimentary Health Ltd, Cremo, Danone, General Mills, Friesland Campina, Janssen, Kerry, MeadJohnson, Nutricia, 4D Pharma plc, Second Genome, and Sigmoid pharma. The authors received an honorarium from the International Scientific Forum on Home Hygiene for their time in preparation of this manuscript.

Contributor Information

Sally F Bloomfield, London School of Hygiene & Tropical Medicine and International Scientific Forum on Home Hygiene, The Old Dairy Cottage, Montacute, Somerset TA15 6XL, UK.

Graham AW Rook, Centre for Clinical Microbiology, Department of Infection, University College London (UCL), London, UK.

Elizabeth A Scott, Center for Hygiene and Health, Department of Biology, Simmons College, Boston, MA, USA.

Fergus Shanahan, APC Microbiome Institute, University College Cork – National University of Ireland, Cork, Ireland.

Rosalind Stanwell-Smith, London School of Hygiene & Tropical Medicine, London, UK.

Paul Turner, Section of Paediatrics (Allergy & Infectious Diseases) and MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK; Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, NSW, Australia.

IMAGES

  1. (PDF) The coming-of-age of the hygiene hypothesis

    hygiene hypothesis original paper

  2. (PDF) Is the Hygiene Hypothesis an Example of Hormesis?

    hygiene hypothesis original paper

  3. The Hygiene Hypothesis Essay Example

    hygiene hypothesis original paper

  4. (PDF) The Hygiene Hypothesis and New Perspectives—Current Challenges

    hygiene hypothesis original paper

  5. PPT

    hygiene hypothesis original paper

  6. Hygiene Hypothesis

    hygiene hypothesis original paper

COMMENTS

  1. Hygiene hypothesis

    The idea of a link between parasite infection and immune disorders was first suggested in 1968 [13] before the advent of large scale DNA sequencing techniques.The original formulation of the hygiene hypothesis dates from 1989, when David Strachan proposed that lower incidence of infection in early childhood could be an explanation for the rise in allergic diseases such as asthma and hay fever ...

  2. The hygiene hypothesis: current perspectives and future therapies

    In 1989, Strachan proposed the hygiene hypothesis of allergic disease after observing that hay fever was less common among children with older siblings. 8 He reasoned that children growing up in larger families may experience increased exposure to microbes in early childhood due to inevitable unhygienic contact with older siblings or prenatal exposure from the mother infected by similar ...

  3. Too clean, or not too clean: the Hygiene Hypothesis and home hygiene

    Discussion of the hygiene hypothesis and the implications for hygiene practice. The link between atopy, and microbial exposure and infection. In the first part of this paper the evidence for a causal link between the sharp rise in atopy over the past 30 years and the possibility of a reduction in our level of exposure to microbes was reviewed.

  4. The Hygiene Hypothesis and New Perspectives—Current Challenges Meeting

    Introduction. Throughout its history, the Hygiene Hypothesis has shown itself to be adaptable and flexible whenever it has been challenged by innovation in science ().A number of new findings need to be considered in this ongoing revisiting process: The originally proposed Th1/Th2 paradigm is challenged by currently elucidated new classes of effector and regulating immune cells pointing out to ...

  5. The hygiene hypothesis at a glance: Early exposures, immune mechanism

    The hygiene hypothesis was proposed almost three decades ago. Nevertheless, its mechanism still remains with relevant controversies. Some studies defend that early exposures during childhood to microbes and parasites are key determinants to prevent allergies and autoimmune diseases; however, other studies demonstrated that these early exposures can even potentiate the clinical scenario of the ...

  6. The hygiene hypothesis: current perspectives and future therapies

    The old friends hypothesis, proposed by Rook et al, notes the co-evolution of microorganisms and macroorganisms, such as parasitic helminths, with the development of the human immune system. Citation 22 Similar to the hygiene hypothesis, it suggests that these organisms are required for normal immune system development.

  7. Hygiene Hypothesis

    The hygiene hypothesis proposes that as living environment is improved, children become less exposed to infectious agents, which leads to inadequate maturation of their immune systems. This hypothesis suggests that early exposure to pathogens may enhance the immune responses of those children, thereby suppressing autoimmune reactions involved ...

  8. The hygiene hypothesis: Current perspectives and future therapies

    The "h ygiene h ypothesis", proposed by Strachan in 1 989, aimed to explain this peculiar genera. tional rise in immune dysre gulation. Ho wev er, research o ver the past 1 0 years provides ...

  9. The hygiene hypothesis in autoimmunity: the role of pathogens and

    The hygiene hypothesis postulates that an increased frequency of infections contributes to a decrease in autoimmune and allergic diseases. ... the protective effect of the original environment ...

  10. The Hygiene Hypothesis

    The Hygiene Hypothesis comprise immunological, microbiological and evolutionary aspects. Thus, the original postulate developed into a holistic model that explains the impact of post-modern life-style on humans, who initially evolved in close proximity to a more natural environment. Focusing on diet and the microbiome as the most prominent ...

  11. Frontiers

    The Hygiene Hypothesis comprise immunological, microbiological and evolutionary aspects. Thus, the original postulate developed into a holistic model that explains the impact of post-modern life-style on humans, who initially evolved in close proximity to a more natural environment.

  12. Cleaning up the hygiene hypothesis

    They decided the name has to go ( 15 ). "The trouble is, as soon as you use the words 'hygiene hypothesis,' the word hygiene prejudges what the cause is," says Bloomfield. To the public, "hygiene" is interpreted as personal cleanliness: washing hands, keeping food clean and fresh, sanitizing the home.

  13. The Hygiene Hypothesis

    The Hygiene Hypothesis comprise immunological, microbiological and evolutionary aspects. Thus, the original postulate developed into a holistic model that explains the impact of post-modern life-style on humans, who initially evolved in close proximity to a more natural environment. Focusing on diet and the microbiome as the most prominent ...

  14. The hygiene hypothesis, the COVID pandemic, and consequences for the

    The model in Fig. 1 outlines the process by which microbial diversity is lost. Gut microbial richness results from a balance of the acquisition and the loss of microbial species. The original hygiene hypothesis, first framed by David Strachan (), has evolved into new, more complex and explicit hypotheses that capture many of the processes that influence gut microbial establishment and ...

  15. The Hygiene Hypothesis

    As mentioned previously, the first description of the hygiene hypothesis was made on the observation that siblings, with increased exposure to infectious disease through contact with other children, suffer less from allergies (Strachan 1989).Since then, these results were supported by a large number of studies (Strachan et al. 1997a, b; Ball et al. 2000; Bodner et al. 1998).

  16. Is the Hygiene Hypothesis True?

    The hygiene hypothesis is the idea that kids need to be exposed to germs in order to develop healthy immune systems. We know that many common viruses did not circulate as widely during the pandemic, thanks to social distancing, masking, and other COVID mitigation measures. Are there downsides to those missed infections? In this Q&A, Caitlin ...

  17. The Ecoimmunology of Health and Disease: The Hygiene Hypothesis and

    The original hygiene hypothesis proposed that certain diseases derive from low levels of early-life microbial exposure. Since then, the hypothesis has been applied to numerous inflammatory, autoimmune, and allergic conditions. The changes in hygiene linked to these diseases include numerous changes in biotic exposure and lifestyle. To this end, some scholars have called for abandonment of the ...

  18. The hygiene hypothesis for allergy

    The "hygiene hypothesis" In 1989 Strachan published a paper entitled "Hay fever, hygiene and household size", in which he observed that increasing family size was associated with a reduced risk of developing hay fever ().Using data from a British 1958 birth cohort (the 1958 National Child Development Study) he found that the prevalence of hay fever at both 11 (parent-reported) and 23 ...

  19. The hygiene hypothesis, the COVID pandemic, and consequences for the

    Abstract. The COVID-19 pandemic has the potential to affect the human microbiome in infected and uninfected individuals, having a substantial impact on human health over the long term. This pandemic intersects with a decades-long decline in microbial diversity and ancestral microbes due to hygiene, antibiotics, and urban living (the hygiene ...

  20. Hay fever, hygiene, and household size.

    Hay fever, hygiene, and household size. - PMC. As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health. BMJ. 1989 Nov 18; 299 (6710): 1259-1260.

  21. Hay fever, hygiene, and household size.

    Please note: your email address is provided to the journal, which may use this information for marketing purposes.

  22. The hygiene hypothesis: current perspectives and future therapies

    The old friends hypothesis: parasitic helminths The old friends hypothesis, proposed by Rook et al, notes the co-evolution of microorganisms and macroorganisms, such as parasitic helminths, with the development of the human immune system.22 Similar to the hygiene hypothesis, it sug-gests that these organisms are required for normal immune

  23. Time to abandon the hygiene hypothesis: new perspectives on allergic

    These were analysed and key themes were integrated into a paper which was made available online to all authors for review. This included further questions soliciting the author's views. After further rounds of questions and revision, a consensus position was obtained. ... a number of refinements to the original hygiene hypothesis now seem to ...