Search

Close

Article Published in the Author Account of

Amy D Proal

Inflammatory Disease and the Human Microbiome

Abstract: The human body is a superorganism in which thousands of microbial genomes continually interact with the human genome. A range of physical and neurological inflammatory diseases are now associated with shifts in microbiome composition. Seemingly disparate inflammatory conditions may arise from similar disruption of microbiome homeostasis. Intracellular pathogens long associated with inflammatory disease are able to slow the innate immune response by dysregulating activity of the VDR nuclear receptor. This facilitates the ability of other species to gradually accumulate in tissue and blood, where they generate proteins and metabolites that significantly interfere with the body’s metabolic processes. The microbes that contribute to this dysfunction are often inherited from family members. Immunosuppressive therapies for inflammatory disease allow pathogens driving these processes to spread with greater ease. In contrast to immunosuppression, treatments that stimulate the immune system seem to allow for reversal of this pathogen-induced genomic dysregulation.



Introduction

Over just the past decade, molecular tools have revolutionized the field of microbiology. These tools — which include pyrosequencing, single cell sampling, and shotgun sequencing — directly extract and clone DNA from communities of microorganisms in the human body. We can subsequently characterize microbes based on their genomic signatures rather than their appearance in a Petri dish.

Indeed, it is now understood that the vast majority of microbes capable of persisting in Homo sapiens cannot be cultured in a laboratory. In contrast, studies using these new molecular tools have revealed the presence of thousands of previously unknown microbes in human tissue and blood (Pagani et al., 2012). These microbes persist both in and on the human body, and are collectively referred to as the human microbiome.

Many of these newly characterized microbes have the capacity to contribute to human disease processes. For example, the amniotic fluid is widely regarded as being sterile. However, in 2010, DiGiulio and Relman and their colleagues at Stanford University published a seminal study that used molecular methods to identify eighteen different bacterial taxa in the amniotic fluid of women during preterm labor (DiGiulio et al., 2008). A number of bacterial species identified were previously uncultivated and uncharacterized. The positive predictive value of PCR (polymerase chain reaction) for preterm delivery was 100%.

Discoveries like Relman’s allow us to study chronic inflammatory disease from a new perspective. The theory of autoimmunity — in which the immune system is thought to lose tolerance and generate antibodies against self — gained traction before molecular-based technologies existed. The human body was logically assumed to be largely sterile. It followed that inflammatory conditions associated with autoantibodies could simply not be tied to infectious processes.

Today however, armed with a growing understanding of the diversity and extent of the human microbiome, we can re-evaluate many of these processes. Much of the inflammation and systemic dysfunction observed among patients with autoimmune and inflammatory disease can now be traced to components of the microbiome, and to the genetic pathways that pathogens within the microbiome dysregulate in order to survive.

The Human Superorganism

Two recent large-scale collaborations spearheaded the use of these metagenomic technologies. One was the Human Microbiome Project (2008-2012), a U.S.-based initiative funded by the U.S. National Institutes of Health (Turnbaugh et al., 2007). The Project generated over 3.5 terabases of metagenomic sequences (Methé et al., 2012). Another initiative was MetaHIT, a Europe-based initiative (2008-2012), which focused primarily on better characterizing the gut microbiome (Ehrlich, 2011).

In conjunction with data generated by other private initiatives, these projects detected and characterized so many novel microbes in Homo sapiens that at least 90% of cells in the human body are now understood to be bacterial, fungal, or otherwise non-human in origin. As of March 2014, the Genomes Online database lists 2,723 completed and published bacterial genomes detected in the human body with at least 14,867 in progress (Pagani et al., 2012).

Our knowledge of the chronic viruses that persist in Homo sapiens is also rapidly evolving. Gordon and colleagues analyzed the fecal virome of monozygotic twins and their mothers (Reyes et al., 2010). Eighty one percent of the reads generated from this virome did not match those of any known viruses. In 2011, Pride et al. (2012) found that hundreds of previously uncharacterized bacteriophage species dominate the oral cavity, some of them serving as reservoirs for pathogenic gene function.

The over nine million non-human genes represented by the microbiome (Yang et al., 2009) dwarf the meager 20,500 that comprise the human genome. This knowledge implies a redefinition of the human/microbe relationship. The human body is best understood as a superorganism whose metabolism represents a combination of microbial and human interaction.

While pathogens clearly persist on mucosal surfaces, they are also present in blood and tissue. A microbiome is now known to persist in the lungs (Erb-Downward et al., 2011). Microbial RNA can be easily detected in healthy human blood (McLaughlin et al., 2002), with pathogens such as Helicobacter pylori identified by routine testing (Al-Jobori et al., 2011). Polybacterial and chronic pathogens have been detected in atherosclerotic plaque. These include active species such as Porphyromonas gingivalis (Kozarov et al., 2005).

The Interactome

The millions of proteins and metabolites expressed by these and other microbes continually interact with those expressed by our own human genomes. The resulting interactions between these foreign and host proteins — the interactome — significantly impact all of the body’s metabolic pathways. For example, the HIV genome transcribes 19 proteins, yet there are 1,443 direct interactions between just these few proteins and the human genome (Fu et al., 2009).

The expression of our human genes is continually modified by the microbes and their metabolic products. For example, the gene PTPN22 has been connected to rheumatoid arthritis, lupus, and diabetes mellitus (Goh et al., 2007). However, PTPN22 expression is also altered by the bacterial metagenome — it is upregulated as part of the innate immune response to mycobacteria (Lykouras et al., 2008).

Molecular Mimicry Further Complicates the Interactome

These genomic interactions are complicated by the fact that the structures of many nucleic acids and microbial proteins are identical or very similar to those expressed by those of their human hosts. For example, humans and E. coli metabolize glucose in nearly the same fashion, so the human superorganism may have difficulty distinguishing proteins and metabolites created by the microbes from those recognized as “self.”

Tens of thousands of protein-protein interactions have been documented between just the genomes of Salmonella, Escherichia coli, Yersinia, and the human genome (Krishnadev and Srinivasan, 2011).

Composition of the Body’s Microbial Communities Often Changes in Disease

Microbiome composition is often altered in patients with chronic inflammatory conditions. This dysbiosis, or microbial imbalance, has been associated with a growing number of chronic diagnoses including Crohn’s disease, ulcerative colitis (Morgan et al., 2012), irritable bowel syndrome (Franceschi et al., 2009), psoriasis, both type 1 (Giongo et al., 2010) and type 2 diabetes (Larsen et al., 2010), and cardiovascular disease (Rajendhran et al., 2013).

Some studies demonstrate direct relationships between microbial composition and disease onset. Amar et al. (2011) found that in 3,280 subjects without diabetes or obesity at baseline, pathogens in the microbiome led to 16S rDNA blood serum concentrations significantly elevated in those who went on to develop diabetes.

It is becoming increasingly clear that inflammatory disease processes are not due to acquisition of any single pathogen. Instead, they appear to result from alterations in the complex microbial communities. It follows that Koch’s postulates, which dictate that one microbe must be proven causative of a single disease state, can no longer be supported in the era of the metagenome.

Pathogens Capable of Intracellular Persistence Can Cause Significant Dysregulation

Many of the pathogens that contribute to chronic inflammatory disease persist inside the nucleated cells. When macrophages internalize Salmonella, persister cells can form that are resistant to most antibiotic therapies (Helaine et al., 2014). When E. coli encounters a macrophage, it can evolve in a manner that allows it to remain alive within the phagosome (Miskinyte et al., 2013).

These intracellular pathogens directly interfere with transcription, translation, and DNA repair at the cellular level. For example, Xu et al. (2003) demonstrated that upon infecting a cell, Mycobacterium tuberculosis alters the expression of 463 human genes. This kind of interference results in severe dysregulation of the interactome.

Dysregulation of the VDR Nuclear Receptor Leads to Dysbiosis and Chronic Disease

The ability of several prominent intracellular pathogens to dysregulate the Vitamin D Nuclear Receptor (VDR) points to a pathway in the molecular biology by which they can drive the systemic dysregulation associated with inflammatory disease.

The VDR expresses at least 913 genes, many connected to autoimmune and inflammatory processes (Wang et al., 2005). In addition, it lies at the heart of the innate immune response. The receptor expresses TLR2, which recognizes bacterial polysaccharides. It also regulates expression of the cathelicidin and beta-defensin antimicrobial peptides, which play vital roles in targeting intracellular pathogens (Auvynet and Rosenstein, 2009). For example, vitamin D-mediated human antimicrobial activity against M. tuberculosis is dependent on the induction of cathelicidin (Liu et al., 2007).

Any microbe capable of dysregulating VDR activity significantly impairs the innate immune response, allowing the pathogen to persist with greater ease. Infection of human B lymphocytes with Epstein-Barr virus downregulates VDR activity by a factor of at least fifteen — particularly in younger, longer-lasting lymphoblastoid cells (Yenamandra et al., 2009). Persistent M. tuberculosis slows VDR activity (Xu et al., 2003). Borrelia burgdorferi (Salazar et al., 2009), Cytomegalovirus (Chan et al., 2008), and Mycobacterium leprae (Liu et al., 2012) also slow VDR activity to varying degrees. The fungus Aspergillus fumigatus secretes a gliotoxin which significantly downregulates VDR expression (Coughlan et al., 2012). Disabling the innate immune system via the VDR pathway is an extremely logical pathogen survival mechanism. Thus, other undetected or uncharacterized microbes likely survive in the same or similar fashion.

Flow on Effects of VDR Dysregulation Further Slow Immune Activity

VDR dysregulation is characterized by rising levels of the active vitamin D metabolite 1,25-dihydroxyvitamin D (1,25-D) and lower levels of the inactive 25-hydroxyvitamin D (Blaney et al., 2009). Our in silico data suggests that, when elevated, 1,25-D can interfere with expression of other nuclear receptors, including the androgen receptor, the glucocorticoid receptor, and the thyroid receptor (Proal et al., 2009).

Each of these other nuclear receptors express additional families of antimicrobial peptides (Brahmachary et al., 2006), which can also be compromised by this same VDR dysregulation. This leads to profound immunosuppression. Indeed, decreased expression of cathelicidin over time has been demonstrated in both Crohn’s disease (Nuding et al., 2007) and sarcoidosis (Barna et al., 2012).

Successive Infection

Under these conditions, an individual’s microbiome may shift towards a composition that promotes disease. We refer to this process, in which the host microbiome shifts gradually away from a homeostatic state, as successive infection (Proal et al., 2010). Infected cells struggle to correctly produce human metabolites in the presence of the pathogenic proteins, enzymes, and metabolites. Any pathogen that decreases antimicrobial peptide expression facilitates the ability of yet other pathogens to persist and cumulatively slow innate immune activity. This creates a snowball effect, where it becomes progressively easier for the host to acquire pathogens as the strength of the innate immune response decreases. Since the immune system strives to target the persistent microbes but never fully succeeds, a stalemate results, and low-grade inflammation accumulates.

Eventually, a person undergoing successive infection may present with symptoms sufficient for an inflammatory diagnosis. The unique symptoms any one person develops vary depending on the location, species, and virulence of the pathogens they have acquired over time, along with the myriad ways in which the proteins and metabolites created by these microbes cause dysfunction by interacting with those of the host.

Microbes are notoriously competitive. Some pathogens successfully outcompete larger populations of less aggressive microbes. Serious dysfunction occurs if keystone species are lost (Cho and Blaser, 2012). Further, the dominance of certain microbial species may increase or decrease depending on the state of the interactome. For example, the guts of infants with atopic eczema are characterized by decreased microbial diversity (Abrahamsson et al., 2012). However, in bacterial vaginosis, vaginal microbiome composition becomes much more diverse and taxon-rich than that of healthy individuals (Oakley et al., 2008).

Comorbidities

Figure 1.

Figure 1. Comorbidities among common inflammatory diseases. Each ′spoke′ of this wheel represents a published study appearing in MEDLINE, which shows a significant statistical relationship between one disease and another.

Patients with one inflammatory diagnosis frequently develop another. For example, composition of the lung microbiome can predict the onset of rheumatoid arthritis (Demoruelle et al., 2014). Figure 1 demonstrates the overlap in disease presentation among patients with a broad range of inflammatory conditions.

Comorbidities directly reflect the successive infectious process, in which different inflammatory conditions may be driven by the same underlying mechanisms. Consequently, seemingly disparate conditions are best studied concurrently. The line between cause and effect is frequently unclear. For example, obesity is often believed to cause diabetes. Yet both conditions have now been tied to changes in microbiome composition (Larsen et al., 2010; Turnbaugh et al., 2006) and obesity itself has been declared a disease.

The microbiome is also involved in neurological conditions including anxiety, depression, and obsessive compulsive disorder (Gonzalez et al., 2011). Thus, successive infection may also contribute to the development of neurological disease. For example, the PRIME study found that in healthy, European, middle-aged men, baseline depressive symptoms were associated with an increased risk of coronary heart disease in the short-term, and with stroke over the long-term (Majed et al., 2012).

Familial Aggregation

An understanding of the comorbid conditions makes it easier to recognize familial aggregation. For instance, Benros et al. (2014) found that patients with schizophrenia are more likely to develop an autoimmune disease.

Familial aggregation likely results when elements of the microbiome are inherited. For example, infected siblings, mothers, and fathers are all major sources for H. pylori acquisition among young children, with the infected mother serving as the main source for childhood (Weyermann et al., 2009). Relman and colleagues have demonstrated that within just weeks of birth, infants develop a microbiome that reflects components of not just their parents’ microbiomes but those of their extended families as well (Costello et al., 2013).

Evidence that pathogens are passed down the maternal line is particularly strong. Breast milk delivers a microbiome that varies between women depending on a host of factors (Cabrera-Rubio et al., 2012). The health of the mother also appears to directly impact composition of her milk microbiome. These authors also found that milk from obese mothers tended to contain a different and less diverse bacterial community than that obtained from normal weight mothers.

Autoantibodies

Autoantibodies can be generated in response to microbes. High titers of rheumatoid factor (RF) have been detected not only in patients with rheumatoid arthritis, but also in patients with a number of bacterial, viral, and parasitic infections (Russell et al., 1992).

Autoantibodies are notoriously polyspecific — antibodies created to target pathogens may additionally target human proteins. This results in “collateral damage” and inflammation. For example, Sutjita et al. (1988) found that when normal individuals were injected with tetanus toxoid, at least seven antibodies were produced. One was an autoantibody to cardiolipin. B cells infected with EBV secrete antibodies capable of reacting with dozens of self and non-self antigens including albumin, renin, and thyroglobulin (Seigneurin et al., 1988).

This means that the “autoantibodies” often detected in patients with inflammatory disease may simply be antibodies generated from the accumulation of pathogens into the microbiome. “Autoantibodies” are frequently detected in patients months or years before the onset of clinical disease symptoms. This reflects the gradual increase in microbiome dysbiosis characteristic of successive infection.

Immunosuppression Contributes to Prolongation of Disease by Delaying Resolution

Immunosuppresive therapies represent the standard of care for most inflammatory conditions, particularly those considered to be autoimmune. Corticosteroids, TNF-alpha antagonists, and rituximab are among the many treatments routinely used to slow immune activity. These treatments often provide short-term symptom palliation, but have poor long-term track records when it comes to relapse and stability. No definitive studies have demonstrated that corticosteroids improve long-term prognosis or reduce mortality. Indeed, the opposite is true (Gottlieb et al., 1997).

This pattern is already recognized in the context of acute infection. For example, Earn et al. (2014) recently concluded that using antipyretic medications to suppress fever (and subsequently the immune response) in patients with influenza allowed viral particles to spread more easily between people. Thus, while subjects taking the antipyretic medications felt fewer symptoms, they were actually more contagious.

As the concept of “autoimmunity” is re-evaluated in light of chronic infection, the utility of immunosuppression must be reconsidered. Temporary symptom relief among patients taking immunosuppressive medications may result because the immune system cannot adequately respond to pathogenic insult. Cytokine and chemokine levels may decrease, yet this allows pathogens to proliferate more easily.

The secosteroid vitamin D has potent immunosuppressive properties (Kimball et al., 2011). Benefits observed among patients ingesting the substance may subsequently result from the short-term palliative effect described above. Indeed, a number of recent randomized controlled trials have studied the effect of vitamin D supplementation in patients with chronic inflammatory conditions. Most have failed to support a link between supplementation and improved health. In some cases, harm has been reported (Albert et al., 2009; Autier et al., 2014).

Immunostimulation May Better Target the Inflammatory Disease Process

If slowing the immune response in patients with inflammatory disease allows infectious agents to proliferate more easily, then approaches that seek to stimulate immune defenses should result in the opposite outcome. A reinvigorated immune system could more effectively kill pathogens at the heart of the inflammatory disease process.

Figure 2.

Figure 2. Immunopathology is a cascade of reactions including inflammation, cytokine release, and endotoxin release that occur as part of the immune response against microbes (From Ruslan Medzhitov, La Jolla Immunology Conference, October 2011).

Yet if the immune system successfully targets pathogens, some elements of the response — the cytokine storm, for instance — may cause the host to experience discomfort or an increase in disease symptoms. This is because as chronic pathogens and the cells they once inhabited are killed, toxins, cytokines, chemokines, and debris are released into the bloodstream. This leads to temporary increases in signs and symptoms of disease, and in many cases, temporary fluctuations in the levels of inflammatory disease markers. This phenomenon is called immunopathology (Figure 2).

Over the past decade, in concert with our clinical collaborators, we have developed an immunostimulative therapy that has been used to treat patients with a wide range of chronic inflammatory conditions. The treatment centers on use of a putative VDR agonist, with the goal of reactivating the antimicrobial peptides and other components of innate immunity under VDR control. Although the treatment is in early stages, physicians have started to publish case histories that seem to demonstrate both objective and subjective improvement amongst their patients (Proal et al., 2011; 2013).

Patients using the therapy report high levels of immunopathology, especially during earlier periods of treatment. Yet over time, this immunopathology tends to wane at the same time that symptom improvement is frequently noted (Proal et al., 2011). This pattern strongly suggests that patients on the therapy are indeed targeting pathogens associated with their disease states.

Discussion

The human body is a superorganism in which the human genome continually interacts with the millions of microbial genes from the microbiome. Pathogens have now been identified in tissue and blood previously considered sterile. Those capable of persisting inside the nucleated cells can directly alter cellular transcription, translation, and DNA repair mechanisms. Dysregulation of VDR nuclear receptor activity by a number of prominent pathogens can slow innate immune activity to the point where microbiome composition shifts away from a state of homeostasis. Under these conditions, a person may develop an inflammatory disease, the nature of which varies depending upon many factors. These include the location and virulence of the pathogens they acquire and the semi-infinite number of ways in which the proteins and metabolites created by these pathogens can cause dysfunction by dysregulating the body’s metabolic pathways.

The comorbidities so frequently observed among patients with a wide range of both physical and neurological conditions support the possibility that different inflammatory conditions may develop from common underlying mechanisms. Babies begin to harbor a microbiome just weeks after birth, the composition of which reflects the microbiome of their parents and even those of extended relatives. Thus, the familial aggregation characteristic of inflammatory disease may well result when components of the microbiome are inherited.

The theory of autoimmunity was developed at a time when the human body was believed to be largely sterile. However, as the thousands of species within the microbiome are increasingly characterized, it is more likely that the autoantibodies detected in patients with autoimmune disease are generated in response to pathogens rather than “self.”

Immunosuppressive therapies for inflammatory disease may provide short-term relief by slowing the cytokine and chemokine release associated with a healthy immune response towards acquired pathogens. However, pathogens are able to spread with much greater ease over the long term, leading to relapse and instability. Indeed, during the period that immunosuppressive therapies have become the standard of care in the United States, the incidence of nearly every chronic disease has increased. The secosteroid vitamin D has immunosuppressive properties, and should subsequently be evaluated in this context.

It is urgent that we re-evaluate the long-term efficacy of immunosuppressive therapies. In lieu of slowing the innate immune response in patients with inflammatory disease, it seems we should seek to activate it, so that chronic pathogens might be successfully targeted. However, patients on an immunostimulative therapy will inevitably experience immunopathology as toxins and debris generated from microbial death enter the bloodstream. While the resulting symptoms may be difficult to manage, the root cause of the disease is being addressed.

There is a pressing need for researchers to focus on developing tests that might better characterize and measure immunopathology in a clinical setting. Additionally, the current standard of care prioritizes symptom palliation. This means that physicians have few guidelines with which to evaluate the symptom and metabolite fluctuations characteristic of immunopathology. Development of techniques that might help patients better manage the reaction must also become a priority.

Disclosure

The authors report no conflicts of interest.

Corresponding Author

Amy D. Proal, Ph.D., Autoimmunity Research Foundation, 3423 Hill Canyon Ave., Thousand Oaks, CA 91360, USA.

References

Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol 129(2):434-440, 2012.

Al-Jobori MB, Al-Ouqaili MT, Abdullah EM. Detection of 16S rRNA gene of Helicobacter pylori in patients with peptic ulcer and gastric carcinoma: molecular and bacteriological study. Egypt Acad J Biolog Sci 3:95-104, 2011.

Albert PJ, Proal AD, Marshall TG. Vitamin D: the alternative hypothesis. Autoimmun Rev 8(8):639-644, 2009.

Amar J, Serino M, Lange C, Chabo C, Iacovoni J, Mondot S, Lepage P, Klopp C, Mariette J, Bouchez O, Perez L, Courtney M, Marre M, Klopp P, Lantieri O, Dore J, Charles MA, Balkau B, Burcelin R. Involvement of tissue bacteria in the onset of diabetes in humans: evidence for a concept. Diabetologia 54(12):3055-3061, 2011.

Autier P, Boniol M, Pizot C, Mullie P. Vitamin D status and ill health: a systematic review. Lancet Diabetes Endocrinol 2(1):76-89, 2014.

Auvynet C, Rosenstein Y. Multifunctional host defense peptides: antimicrobial peptides, the small yet big players in innate and adaptive immunity. FEBS J 276(22):6497-6508, 2009.

Barna BP, Culver DA, Kanchwala A, Singh RJ, Huizar I, Abraham S, Malur A, Marshall I, Kavuru MS, Thomassen MJ. Alveolar macrophage cathelicidin deficiency in severe sarcoidosis. J Innate Immun 4(5-6):569-578, 2012.

Benros ME, Pedersen MG, Rasmussen H, Eaton WW, Nordentoft M, Mortensen PB. A nationwide study on the risk of autoimmune diseases in individuals with a personal or a family history of schizophrenia and related psychosis. Am J Psychiatry 171(2):218-226, 2014.

Blaney GP, Albert PJ, Proal AD. Vitamin D metabolites as clinical markers in autoimmune and chronic disease. Ann N Y Acad Sci 1173:384-390, 2009.

Brahmachary M, Schonbach C, Yang L, Huang E, Tan SL, Chowdhary R, Krishnan SP, Lin CY, Hume DA, Kai C, Kawai J, Carninci P, Hayashizaki Y, Bajic VB. Computational promoter analysis of mouse, rat and human antimicrobial peptide-coding genes. BMC Bioinformatics 7(Suppl 5):S8, 2006.

Cabrera-Rubio R, Collado MC, Laitinen K, Salminen S, Isolauri E, Mira A. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr 96(3):544-551, 2012.

Chan G, Bivins-Smith ER, Smith MS, Smith PM, Yurochko AD. Transcriptome analysis reveals human cytomegalovirus reprograms monocyte differentiation toward an M1 macrophage. J Immunol 181(1):698-711, 2008.

Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet 13(4):260-270, 2012.

Costello EK, Carlisle EM, Bik EM, Morowitz MJ, Relman DA. Microbiome assembly across multiple body sites in low-birthweight infants. MBio 4(6):e00782-e00713, 2013.

Coughlan CA, Chotirmall SH, Renwick J, Hassan T, Low TB, Bergsson G, Eshwika A, Bennett K, Dunne K, Greene CM, Gunaratnam C, Kavanagh K, Logan PM, Murphy P, Reeves EP, Mcelvaney NG. The effect of Aspergillus fumigatus infection on vitamin D receptor expression in cystic fibrosis. Am J Respir Crit Care Med 186(10):999-1007, 2012.

Demoruelle M, Norris J, Holers V, Harris J, Deane K. The lung microbiome differs in asymptomatic subjects at elevated risk of future rheumatoid arthritis compared with healthy control subjects. Ann Am Thorac Soc 11:S74, 2014.

Digiulio DB, Romero R, Amogan HP, Kusanovic JP, Bik EM, Gotsch F, Kim CJ, Erez O, Edwin S, Relman DA. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One 3(8):e3056, 2008.

Earn DJ, Andrews PW, Bolker BM. Population-level effects of suppressing fever. Proc Biol Sci 281(1778):20132570, 2014.

Ehrlich SD. MetaHIT: The European Union Project on metagenomics of the human intestinal tract. In: Metagenomics of the Human Body. KE Nelson (Ed.). pp307-316. Springer Science+Business Media, Berlin, Germany, 2011.

Erb-Downward JR, Thompson DL, Han MK, Freeman CM, Mccloskey L, Schmidt LA, Young VB, Toews GB, Curtis JL, Sundaram B, Martinez FJ, Huffnagle GB. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS One 6(2):e16384-e16384, 2011.

Franceschi F, Niccoli G, Ferrante G, Gasbarrini A, Baldi A, Candelli M, Feroce F, Saulnier N, Conte M, Roccarina D, Lanza GA, Gasbarrini G, Gentiloni SN, Crea F. CagA antigen of Helicobacter pylori and coronary instability: insight from a clinico-pathological study and a meta-analysis of 4241 cases. Atherosclerosis 202(2):535-542, 2009.

Fu W, Sanders-Beer BE, Katz KS, Maglott DR, Pruitt KD, Ptak RG. Human immunodeficiency virus type 1, human protein interaction database at NCBI. Nucleic Acids Res 37(Database issue):D417-D422, 2009.

Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyoty H, Veijola R, Simell T, Simell O, Neu J, Wasserfall CH, Schatz D, Atkinson MA, Triplett EW. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J 5(1):82-91, 2010.

Goh KI, Cusick ME, Valle D, Childs B, Vidal M, Barabasi AL. The human disease network. Proc Natl Acad Sci U S A 104(21):8685-8690, 2007.

Gonzalez A, Stombaugh J, Lozupone C, Turnbaugh PJ, Gordon JI, Knight R. The mind-body-microbial continuum. Dialogues Clin Neurosci 13(1):55-62, 2011.

Gottlieb JE, Israel HL, Steiner RM, Triolo J, Patrick H. Outcome in sarcoidosis. The relationship of relapse to corticosteroid therapy. Chest 111(3):623-631, 1997.

Helaine S, Cheverton AM, Watson KG, Faure LM, Matthews SA, Holden DW. Internalization of Salmonella by macrophages induces formation of nonreplicating persisters. Science 343(6167):204-208, 2014.

Kimball S, Vieth R, Dosch HM, Bar-Or A, Cheung R, Gagne D, O’connor P, D’souza C, Ursell M, Burton JM. Cholecalciferol plus calcium suppresses abnormal PBMC reactivity in patients with multiple sclerosis. J Clin Endocrinol Metab 96(9):2826-2834, 2011.

Kozarov EV, Dorn BR, Shelburne CE, Dunn WA, Progulske-Fox A. Human atherosclerotic plaque contains viable invasive Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Arterioscler Thromb Vasc Biol 25(3):e17-e18, 2005.

Krishnadev O, Srinivasan N. Prediction of protein-protein interactions between human host and a pathogen and its application to three pathogenic bacteria. Int J Biol Macromol 48(4):613-619, 2011.

Larsen N, Vogensen FK, Van Den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, Al-Soud WA, Sorensen SJ, Hansen LH, Jakobsen M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 5(2):e9085, 2010.

Liu PT, Stenger S, Tang DH, Modlin RL. Cutting edge: vitamin D-mediated human antimicrobial activity against Mycobacterium tuberculosis is dependent on the induction of cathelicidin. J Immunol 179(4):2060-2063, 2007.

Liu PT, Wheelwright M, Teles R, Komisopoulou E, Edfeldt K, Ferguson B, Mehta MD, Vazirnia A, Rea TH, Sarno EN, Graeber TG, Modlin RL. MicroRNA-21 targets the vitamin D-dependent antimicrobial pathway in leprosy. Nat Med 18(2):267-273, 2012.

Lykouras D, Sampsonas F, Kaparianos A, Karkoulias K, Tsoukalas G, Spiropoulos K. Human genes in TB infection: their role in immune response. Monaldi Arch Chest Dis 69(1):24-31, 2008.

Majed B, Arveiler D, Bingham A, Ferrieres J, Ruidavets J-B, Montaye M, Appleton K, Haas B, Kee F, Amouyel P, Ducimetiere P, Empana J-P. Depressive symptoms, a time-dependent risk factor for coronary heart disease and stroke in middle-aged men: the PRIME Study. Stroke 43(7):1761-1767, 2012.

Mclaughlin RW, Vali H, Lau PCK, Palfree RGE, De Ciccio A, Sirois M, Ahmad D, Villemur R, Desrosiers M, Chan ECS. Are there naturally occurring pleomorphic bacteria in the blood of healthy humans? J Clin Microbiol 40(12):4771-4775, 2002.

Methé BA, Nelson KE, Pop M, Creasy HH, Giglio MG, Huttenhower C, Gevers D, Petrosino JF, Abubucker S, Badger JH, Chinwalla AT, Earl AM, Fitzgerald MG. A framework for human microbiome research. Nature 486(7402):215-221, 2012.

Miskinyte M, Sousa A, Ramiro RS, De Sousa JA, Kotlinowski J, Caramalho I, Magalhaes S, Soares MP, Gordo I. The genetic basis of Escherichia coli pathoadaptation to macrophages. PLoS Pathog 9(12):e1003802, 2013.

Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, Leleiko N, Snapper SB, Bousvaros A, Korzenik J, Sands BE, Xavier RJ, Huttenhower C. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 13(9):R79, 2012.

Nuding S, Fellermann K, Wehkamp J, Stange EF. Reduced mucosal antimicrobial activity in Crohn’s disease of the colon. Gut 56(9):1240-1247, 2007.

Oakley BB, Fiedler TL, Marrazzo JM, Fredricks DN. Diversity of human vaginal bacterial communities and associations with clinically defined bacterial vaginosis. Appl Environ Microbiol 74(15):4898-4909, 2008.

Pagani I, Liolios K, Jansson J, Chen IM, Smirnova T, Nosrat B, Markowitz VM, Kyrpides NC. The Genomes OnLine Database (GOLD) v.4: status of genomic and metagenomic projects and their associated metadata. Nucleic Acids Res 40(Database issue):D571-D579, 2012.

Pride DT, Salzman J, Haynes M, Rohwer F, Davis-Long C, White RA, 3rd, Loomer P, Armitage GC, Relman DA. Evidence of a robust resident bacteriophage population revealed through analysis of the human salivary virome. ISME J 6(5):915-926, 2012.

Proal AD, Albert PJ, Blaney GP, Lindseth IA, Benediktsson C, Marshall TG. Immunostimulation in the era of the metagenome. Cell Mol Immunol 8(3):213-225, 2011.

Proal AD, Albert PJ, Marshall TG. Dysregulation of the vitamin D nuclear receptor may contribute to the higher prevalence of some autoimmune diseases in women. Ann N Y Acad Sci 1173:252-259, 2009.

Proal AD, Albert PJ, Marshall TG. Autoimmune disease and the human metagenome. In: Metagenomics of the Human Body. KE Nelson (ed.). pp231-275. Springer Science+Business Media, Berlin, Germany, 2010.

Proal AD, Albert PJ, Marshall TG, Blaney GP, Lindseth IA. Immunostimulation in the treatment for chronic fatigue syndrome/myalgic encephalomyelitis. Immunol Res 56(2-3):398-412, 2013.

Rajendhran J, Shankar M, Dinakaran V, Rathinavel A, Gunasekaran P. Contrasting circulating microbiome in cardiovascular disease patients and healthy individuals. Int J Cardiol 168(5):5118-5120, 2013.

Reyes A, Haynes M, Hanson N, Angly FE, Heath AC, Rohwer F, Gordon JI. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 466(7304):334-338, 2010.

Russell MW, Wu HY, White PL, Kilian M, Henrichsen J. Serum antibody responses to Streptococcus mutans antigens in humans systemically infected with oral streptococci. Oral Microbiol Immunol 7(6):321-325, 1992.

Salazar JC, Duhnam-Ems S, La Vake C, Cruz AR, Moore MW, Caimano MJ, Velez-Climent L, Shupe J, Krueger W, Radolf JD. Activation of human monocytes by live Borrelia burgdorferi generates TLR2-dependent and -independent responses which include induction of IFN-beta. PLoS Pathog 5(5):e1000444, 2009.

Seigneurin JM, Guilbert B, Bourgeat MJ, Avrameas S. Polyspecific natural antibodies and autoantibodies secreted by human lymphocytes immortalized with Epstein-Barr virus. Blood 71(3):581-585, 1988.

Sutjita M, Hohmann A, Comacchio R, Bradley J. Polyspecific human and murine antibodies to diphtheria and tetanus toxoids and phospholipids. Clin Exp Immunol 73(2):191-197, 1988.

Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature 449(7164):804-810, 2007.

Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444(7122):1027-1031, 2006.

Wang TT, Tavera-Mendoza LE, Laperriere D, Libby E, Macleod NB, Nagai Y, Bourdeau V, Konstorum A, Lallemant B, Zhang R, Mader S, White JH. Large-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genes. Mol Endocrinol 19(11):2685-2695, 2005.

Weyermann M, Rothenbacher D, Brenner H. Acquisition of Helicobacter pylori infection in early childhood: independent contributions of infected mothers, fathers, and siblings. Am J Gastroenterol 104(1):182-189, 2009.

Xu Y, Xie J, Li Y, Yue J, Chen J, Chunyu L, Wang H. Using a cDNA microarray to study cellular gene expression altered by Mycobacterium tuberculosis. Chin Med J (Engl) 116(7):1070-1073, 2003.

Yang X, Xie L, Li Y, Wei C. More than 9,000,000 unique genes in human gut bacterial community: estimating gene numbers inside a human body. PLoS One 4(6):e6074, 2009.

Yenamandra SP, Lundin A, Arulampalam V, Yurchenko M, Pettersson S, Klein G, Kashuba E. Expression profile of nuclear receptors upon Epstein — Barr virus induced B cell transformation. Exp Oncol 31(2):92-96, 2009.

[Discovery Medicine; ISSN: 1539-6509; Discov Med (17):257-265, Number 95, May 2014. Copyright © Discovery Medicine. All rights reserved.]

Access This PDF as a Subscriber |
Close
Close
E-mail It
Close