201
|
Peñalver Bernabé B, Maki PM, Dowty SM, Salas M, Cralle L, Shah Z, Gilbert JA. Precision medicine in perinatal depression in light of the human microbiome. Psychopharmacology (Berl) 2020; 237:915-941. [PMID: 32065252 DOI: 10.1007/s00213-019-05436-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
Perinatal depression is the most common complication of pregnancy and affects the mother, fetus, and infant. Recent preclinical studies and a limited number of clinical studies have suggested an influence of the gut microbiome on the onset and course of mental health disorders. In this review, we examine the current state of knowledge regarding genetics, epigenetics, heritability, and neuro-immuno-endocrine systems biology in perinatal mood disorders, with a particular focus on the interaction between these factors and the gut microbiome, which is mediated via the gut-brain axis. We also provide an overview of experimental and analytical methods that are currently available to researchers interested in elucidating the influence of the gut microbiome on mental health disorders during pregnancy and postpartum.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States.
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shannon M Dowty
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Mariana Salas
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Lauren Cralle
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Zainab Shah
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Scripts Oceanographic Institute, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
202
|
Abstract
PURPOSE OF REVIEW This review highlights current knowledge on the dichotomous role played by T helper 17 cells (Th17)-polarized CD4 T cells in maintaining mucosal immunity homeostasis versus fueling HIV/simian immunodeficiency virus (SIV) replication/persistence during antiretroviral therapy (ART), with a focus on molecular mechanisms underlying these processes. RECENT FINDING Th17 cells bridge innate and adaptive immunity against pathogens at mucosal barrier surfaces. Th17 cells are located at portal sites of HIV/SIV entry, express a unique transcriptional/metabolic status compatible with viral replication, and represent the first targets of infection. The paucity of Th17 cells during HIV/SIV infection is caused by infection itself, but also by an altered Th17 differentiation, survival, and trafficking into mucosal sites. This causes major alterations of mucosal barrier integrity, microbial translocation, and disease progression. Unless initiated during the early acute infection phases, ART fails to restore the frequency/functionality of mucosal Th17 cells. A fraction of Th17 cells is long-lived and carry HIV reservoir during ART. Recent studies identified Th17-specific host factors controlling HIV transcription, a step untargeted by current ART. SUMMARY The identification of molecular mechanisms contributing to HIV replication/persistence in mucosal Th17 cells paves the way toward the design of new Th17-specific therapeutic strategies aimed at improving mucosal immunity in HIV-infected individuals.
Collapse
|
203
|
Vujkovic-Cvijin I, Somsouk M. HIV and the Gut Microbiota: Composition, Consequences, and Avenues for Amelioration. Curr HIV/AIDS Rep 2020; 16:204-213. [PMID: 31037552 DOI: 10.1007/s11904-019-00441-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We discuss recent advances in understanding of gut bacterial microbiota composition in HIV-infected subjects and comment on controversies. We discuss the putative effects of microbiota shifts on systemic inflammation and HIV disease progression and potential mechanisms, as well as ongoing strategies being developed to modulate the gut microbiota in humans for amelioration of infectious and inflammatory diseases. RECENT FINDINGS Lifestyle and behavioral factors relevant to HIV infection studies have independent effects on the microbiota. Microbial metabolism of immunomodulatory compounds and direct immune stimulation by translocation of microbes are putative mechanisms contributing to HIV disease. Fecal microbiota transplantation, microbial enzyme inhibition, phage therapy, and rationally selected probiotic cocktails have emerged as promising strategies for microbiota modulation. Numerous surveys of the HIV gut microbiota matched for lifestyle factors suggest consistent shifts in gut microbiota composition among HIV-infected subjects. Evidence exists for a complex pathogenic role of the gut microbiota in HIV disease progression, warranting further study.
Collapse
Affiliation(s)
- Ivan Vujkovic-Cvijin
- Metaorganism Immunity Section, National Institute of Allergy & Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Ma Somsouk
- Division of Gastroenterology, University of California, San Francisco, CA, USA.
| |
Collapse
|
204
|
Abstract
PURPOSE OF REVIEW Aging and HIV share features of intestinal damage and alterations in the communities of enteric bacteria, termed dysbiosis. The purpose of this review is to highlight the various features of the gut microbiome in aging and in people with HIV (PWH) and to discuss how aging and HIV converge to impact the gut microbiome. The term microbiome reflects the combined genetic material of micro-organisms present including bacteria, viruses, bacteriophages, and fungi. To date, the majority of studies investigating the impact of aging and HIV on the gut microbiome have focused on bacteria, and therefore, for the purposes of this review, the term 'microbiome' is used to reflect enteric bacterial communities. RECENT FINDINGS Aging is associated with alterations in the gut bacterial microbiome. Although changes vary by the age of the population, lifestyle (diet, physical activity) and geographic location, the age-associated dysbiosis is typically characterized by an increase in facultative anaerobes with inflammatory properties and a decrease in obligate anaerobes that play critical roles in maintaining intestinal homeostasis and in regulating host immunity. PWH also have dysbiotic gut microbiomes, many features of which reflect those observed in elderly persons. In one study, the age effect on the gut microbiome differed based on HIV serostatus in older adults. SUMMARY HIV and age may interact to shape the gut microbiome. Future studies should investigate relationships between the gut microbiome and age-associated comorbidities in older PWH populations. Identifying these links will provide new avenues for treatments and interventions to improve the healthspan and lifespan of older PWH.
Collapse
|
205
|
Abstract
Recent studies have raised interest in the possibility that dysbiosis of the gut microbiome (i.e., the communities of bacteria residing in the intestine) in HIV-infected patients could contribute to chronic immune activation, and, thus, to elevated mortality and increased risk of inflammation-related clinical diseases (e.g., stroke, cardiovascular disease, cancer, long-bone fractures, and renal dysfunction) found even in those on effective antiretroviral therapy. Yet, to date, a consistent pattern of HIV-associated dysbiosis has not been identified. What is becoming clear, however, is that status as a man who has sex with men (MSM) may profoundly impact the structure of the gut microbiota, and that this factor likely confounded many HIV-related intestinal microbiome studies. However, what factor associated with MSM status drives these gut microbiota-related changes is unclear, and what impact, if any, these changes may have on the health of MSM is unknown. In this review, we outline available data on changes in the structure of the gut microbiome in HIV, based on studies that controlled for MSM status. We then examine what is known regarding the gut microbiota in MSM, and consider possible implications for research and the health of this population. Lastly, we discuss knowledge gaps and needed future studies.
Collapse
Affiliation(s)
- Susan Tuddenham
- Division of Infectious Diseases, Johns Hopkins School of
Medicine, Baltimore, MD
| | - Wei Li Koay
- Department of Infectious Disease, Children’s
National Hospital, Washington, D.C.;,School of Medicine and Health Sciences, George Washington
University, Washington, D.C
| | - Cynthia Sears
- Division of Infectious Diseases, Johns Hopkins School of
Medicine, Baltimore, MD
| |
Collapse
|
206
|
Luján JA, Rugeles MT, Taborda NA. Contribution of the Microbiota to Intestinal Homeostasis and its Role in the Pathogenesis of HIV-1 Infection. Curr HIV Res 2020; 17:13-25. [PMID: 30854974 DOI: 10.2174/1570162x17666190311114808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
During HIV infection, massive destruction of CD4+ T cells ensues, preferentially depleting the Th17 subset at the gut-associated lymphoid tissue (GALT), leading to a loss of mucosal integrity and an increase in cell permeability. This process favors microbial translocation between the intestinal lumen and the circulatory system, contributing to persistent immune activation and chronic inflammation characteristic of HIV infection. Thus, the gut microbiota plays an integral role in maintaining the structure and function of the mucosal barrier, a critical factor for immune homeostasis. However, in the context of HIV infection, changes in the gut microbiota have been reported and have been linked to disease progression. Here, we review evidence for the role of the gut microbiota in intestinal homeostasis, its contribution to HIV pathogenesis, as well as its use in the development of therapeutic strategies.
Collapse
Affiliation(s)
- Jorge A Luján
- Grupo Inmunovirologia, Facultad de Medicina. Universidad de Antioquia, Medellin, Colombia
| | - Maria T Rugeles
- Grupo Inmunovirologia, Facultad de Medicina. Universidad de Antioquia, Medellin, Colombia
| | - Natalia A Taborda
- Grupo Inmunovirologia, Facultad de Medicina. Universidad de Antioquia, Medellin, Colombia.,Grupo de Investigaciones Biomédicas, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| |
Collapse
|
207
|
Dominick L, Midgley N, Swart LM, Sprake D, Deshpande G, Laher I, Joseph D, Teer E, Essop MF. HIV-related cardiovascular diseases: the search for a unifying hypothesis. Am J Physiol Heart Circ Physiol 2020; 318:H731-H746. [PMID: 32083970 DOI: 10.1152/ajpheart.00549.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although the extensive rollout of antiretroviral (ARV) therapy resulted in a longer life expectancy for people living with human immunodeficiency virus (PLHIV), such individuals display a relatively increased occurrence of cardiovascular diseases (CVD). This health challenge stimulated significant research interests in the field, leading to an improved understanding of both lifestyle-related risk factors and the underlying mechanisms of CVD onset in PLHIV. However, despite such progress, the precise role of various risk factors and mechanisms underlying the development of HIV-mediated CVD still remains relatively poorly understood. Therefore, we review CVD onset in PLHIV and focus on 1) the spectrum of cardiovascular complications that typically manifest in such persons and 2) underlying mechanisms that are implicated in this process. Here, the contributions of such factors and modulators and underlying mechanisms are considered in a holistic and integrative manner to generate a unifying hypothesis that includes identification of the core pathways mediating CVD onset. The review focuses on the sub-Saharan African context, as there are relatively high numbers of PLHIV residing within this region, indicating that the greater CVD risk will increasingly threaten the well-being and health of its citizens. It is our opinion that such an approach helps point the way for future research efforts to improve treatment strategies and/or lifestyle-related modifications for PLHIV.
Collapse
Affiliation(s)
- Leanne Dominick
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Natasha Midgley
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Lisa-Mari Swart
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Devon Sprake
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Gaurang Deshpande
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Ismail Laher
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.,Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Danzil Joseph
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eman Teer
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M Faadiel Essop
- Centre for Cardio-metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
208
|
Lu J, Ma SS, Zhang WY, Duan JP. Changes in peripheral blood inflammatory factors (TNF-α and IL-6) and intestinal flora in AIDS and HIV-positive individuals. J Zhejiang Univ Sci B 2020; 20:793-802. [PMID: 31489799 DOI: 10.1631/jzus.b1900075] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE In this study, we investigated the changes in peripheral blood inflammatory factors and intestinal flora in acquired immune deficiency syndrome (AIDS) and human immunodeficiency virus (HIV)-positive individuals (AIDS/HIV patients), and explored the relationships among intestinal flora, peripheral blood inflammatory factors, and CD4+ T lymphocytes. METHODS Thirty blood and stool samples from an AIDS group and a control group were collected. The levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were determined by enzyme-linked immunosorbent assay (ELISA), and the number of CD4+ T lymphocytes by a FACSCount automated instrument. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the messenger RNA (mRNA) levels of Bifidobacterium, Lactobacillus, Escherichia coli, Enterococcus faecalis, and Enterococcus faecium. Correlations among intestinal flora, inflammatory factor levels, and CD4+ T lymphocyte values were evaluated using the Spearman correlation coefficient. RESULTS The levels of TNF-α and IL-6 in the AIDS group were higher than those in the control group, while the number of CD4+ T lymphocytes was lower. The amounts of Bifidobacterium and Lactobacillus in the AIDS group were significantly lower than those in control group, while the amounts of E. coli, E. faecalis, and E. faecium were much higher. The amounts of Bifidobacterium and Lactobacillus were negatively correlated with the content of TNF-α and IL-6 and the CD4+ T lymphocyte count, while those correlations were reversed for E. coli, E. faecalis, and E. faecium. CONCLUSIONS The intestinal microbiota of AIDS/HIV patients were disordered, and there was a correlation between the amount of intestinal flora and the number of CD4+ T lymphocytes and the levels of TNF-α and IL-6.
Collapse
Affiliation(s)
- Jing Lu
- Department of Infectious Disease, Qingdao Sixth People's Hospital, Qingdao 266033, China
| | - Sai-Sai Ma
- Department of Infectious Disease, Qingdao Sixth People's Hospital, Qingdao 266033, China
| | - Wei-Ying Zhang
- Department of Infectious Disease, Qingdao Sixth People's Hospital, Qingdao 266033, China
| | - Jian-Ping Duan
- Department of Infectious Disease, Qingdao Sixth People's Hospital, Qingdao 266033, China
| |
Collapse
|
209
|
Abstract
Alterations in the gut microbiome during HIV infection have been implicated in chronic inflammation, but the role of the oral microbiome in this process is less clear. The article by M. K. Annavajhala, S. D. Khan, S. B. Sullivan, J. Alterations in the gut microbiome during HIV infection have been implicated in chronic inflammation, but the role of the oral microbiome in this process is less clear. The article by M. K. Annavajhala, S. D. Khan, S. B. Sullivan, J. Shah, et al. (mSphere 5:e00798-19, 2020, https://doi.org/10.1128/mSphere.00798-19) investigated the relationship between oral and gut microbiome diversity and immune activation in patients with HIV on antiretroviral therapy. In this study, oral microbiome diversity was inversely associated with inflammatory markers such as soluble CD14 (sCD14), but surprisingly similar associations were not seen with gut microbiome diversity. Oral microbiome diversity was also associated with periodontitis in these patients. This study highlights the importance of continuing multisite examinations in studying the gastrointestinal tract microbiome and also stimulates important directions for future research defining the role of the oral-gut axis in HIV-associated inflammation.
Collapse
|
210
|
Oral and Gut Microbial Diversity and Immune Regulation in Patients with HIV on Antiretroviral Therapy. mSphere 2020; 5:5/1/e00798-19. [PMID: 32024712 PMCID: PMC7002309 DOI: 10.1128/msphere.00798-19] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A feedback loop between dysbiotic gut microbiota, increased translocation of microbial products such as lipopolysaccharide, and inflammation has been hypothesized to cause immune system dysfunction in early HIV infection. However, despite evidence of a chronic inflammatory phenotype in patients on antiretroviral therapy (ART), the role of oral microbiota in systemic immune activation and the relationship between oral and gut bacterial and fungal diversity have not been explored. Our study suggests a crucial role for oral bacterial and fungal communities in long-term systemic immune activation in patients on ART, expanding the current paradigm focused on gut bacteria. Our results indicate that interventions targeting both inflammation and microbial diversity are needed to mitigate oral inflammation-related comorbidities, particularly in HIV-positive patients. More broadly, these findings can bolster general models of microbiome-mediated chronic systemic immune activation and aid the development of precise microbiota-targeted interventions to reverse chronic inflammation. Despite evidence of a chronic inflammatory phenotype in people living with HIV (PLWH) on antiretroviral therapy (ART), the role of oral microbiota in chronic immune activation has not been fully explored. We aimed to determine the relationship between oral and gut microbiome diversity and chronic systemic inflammation in ART-treated PLWH with prevalent severe periodontitis, an inflammatory condition commonly associated with HIV infection. We assessed bacterial and fungal communities at oral and gastrointestinal sites in a cohort (n = 52) of primarily postmenopausal women on ART using 16S rRNA and internal transcribed spacer (ITS) sequencing and measured cellular and soluble markers of inflammation and immune dysfunction. Linear mixed-effect regression and differential abundance analyses were used to associate clinical characteristics and immunological markers with bacterial and fungal diversity and community composition. Bacterial α-diversity in plaque, saliva, and gut was associated with different immunological markers, while mycobial diversity was not associated with soluble or cellular biomarkers of immune stimulation or T cell dysfunction. Furthermore, lipopolysaccharide-positive (LPS+) bacteria previously linked to inflammatory outcomes were enriched at oral sites in patients with severe periodontitis. Fungal α-diversity was reduced in plaque from teeth with higher clinical attachment loss, a marker of periodontitis, and in saliva and plaque from patients with a history of AIDS. Our results show that both bacterial and fungal oral microbiome communities likely play a role in chronic systemic immune activation in PLWH. Thus, interventions targeting both inflammation and the microbiome, particularly in the oral cavity, may be necessary to reduce chronic immune dysregulation in patients with HIV. IMPORTANCE A feedback loop between dysbiotic gut microbiota, increased translocation of microbial products such as lipopolysaccharide, and inflammation has been hypothesized to cause immune system dysfunction in early HIV infection. However, despite evidence of a chronic inflammatory phenotype in patients on antiretroviral therapy (ART), the role of oral microbiota in systemic immune activation and the relationship between oral and gut bacterial and fungal diversity have not been explored. Our study suggests a crucial role for oral bacterial and fungal communities in long-term systemic immune activation in patients on ART, expanding the current paradigm focused on gut bacteria. Our results indicate that interventions targeting both inflammation and microbial diversity are needed to mitigate oral inflammation-related comorbidities, particularly in HIV-positive patients. More broadly, these findings can bolster general models of microbiome-mediated chronic systemic immune activation and aid the development of precise microbiota-targeted interventions to reverse chronic inflammation.
Collapse
|
211
|
Tuddenham SA, Koay WLA, Zhao N, White JR, Ghanem KG, Sears CL. The Impact of Human Immunodeficiency Virus Infection on Gut Microbiota α-Diversity: An Individual-level Meta-analysis. Clin Infect Dis 2020; 70:615-627. [PMID: 30921452 PMCID: PMC7319268 DOI: 10.1093/cid/ciz258] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Whether human immunodeficiency virus (HIV) infection impacts gut microbial α-diversity is controversial. We reanalyzed raw 16S ribosomal RNA (rRNA) gene sequences and metadata from published studies to examine α-diversity measures between HIV-uninfected (HIV-) and HIV-infected (HIV+) individuals. METHODS We conducted a systematic review and individual level meta-analysis by searching Embase, Medline, and Scopus for original research studies (inception to 31 December 2017). Included studies reported 16S rRNA gene sequences of fecal samples from HIV+ patients. Raw sequence reads and metadata were obtained from public databases or from study authors. Raw reads were processed through standardized pipelines with use of a high-resolution taxonomic classifier. The χ2 test, paired t tests, and generalized linear mixed models were used to relate α-diversity measures and clinical metadata. RESULTS Twenty-two studies were identified with 17 datasets available for analysis, yielding 1032 samples (311 HIV-, 721 HIV+). HIV status was associated with a decrease in measures of α-diversity (P < .001). However, in stratified analysis, HIV status was associated with decreased α-diversity only in women and in men who have sex with women (MSW) but not in men who have sex with men (MSM). In analyses limited to women and MSW, controlling for HIV status, women displayed increased α-diversity compared with MSW. CONCLUSIONS Our study suggests that HIV status, sexual risk category, and gender impact gut microbial community α-diversity. Future studies should consider MSM status in gut microbiome analyses.
Collapse
Affiliation(s)
| | - Wei Li A Koay
- Children’s National Medical Center, Baltimore, Maryland
- George Washington University, Washington, District of Columbia, Baltimore, Maryland
| | - Ni Zhao
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Khalil G Ghanem
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cynthia L Sears
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
212
|
Yang X, Su B, Zhang X, Liu Y, Wu H, Zhang T. Incomplete immune reconstitution in HIV/AIDS patients on antiretroviral therapy: Challenges of immunological non-responders. J Leukoc Biol 2020; 107:597-612. [PMID: 31965635 PMCID: PMC7187275 DOI: 10.1002/jlb.4mr1019-189r] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
The morbidity and mortality of HIV type-1 (HIV-1)-related diseases were dramatically diminished by the grounds of the introduction of potent antiretroviral therapy, which induces persistent suppression of HIV-1 replication and gradual recovery of CD4+ T-cell counts. However, ∼10-40% of HIV-1-infected individuals fail to achieve normalization of CD4+ T-cell counts despite persistent virological suppression. These patients are referred to as "inadequate immunological responders," "immunodiscordant responders," or "immunological non-responders (INRs)" who show severe immunological dysfunction. Indeed, INRs are at an increased risk of clinical progression to AIDS and non-AIDS events and present higher rates of mortality than HIV-1-infected individuals with adequate immune reconstitution. To date, the underlying mechanism of incomplete immune reconstitution in HIV-1-infected patients has not been fully elucidated. In light of this limitation, it is of substantial practical significance to deeply understand the mechanism of immune reconstitution and design effective individualized treatment strategies. Therefore, in this review, we aim to highlight the mechanism and risk factors of incomplete immune reconstitution and strategies to intervene.
Collapse
Affiliation(s)
- Xiaodong Yang
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Bin Su
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Xin Zhang
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Yan Liu
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Hao Wu
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| | - Tong Zhang
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for HIV/AIDS Research, Beijing, China
| |
Collapse
|
213
|
Flygel TT, Sovershaeva E, Claassen-Weitz S, Hjerde E, Mwaikono KS, Odland JØ, Ferrand RA, Mchugh G, Gutteberg TJ, Nicol MP, Cavanagh JP, Flægstad T. Composition of Gut Microbiota of Children and Adolescents With Perinatal Human Immunodeficiency Virus Infection Taking Antiretroviral Therapy in Zimbabwe. J Infect Dis 2020; 221:483-492. [PMID: 31549151 PMCID: PMC7457326 DOI: 10.1093/infdis/jiz473] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) infection causes impairment of the gastrointestinal barrier, with substantial depletion of CD4+ T cells in the gut. Antiretroviral therapy (ART) restores CD4+ counts and may have beneficial effects on gut microbiota in adults. Little is known about effect of long-term ART on gut microbiome in HIV-infected children. We investigated composition of gut microbiota in HIV-infected and -uninfected children and assessed associations between gut microbiota and patient characteristics. METHODS In a cross-sectional study, rectal swabs were collected from 177 HIV-infected and 103 HIV-uninfected controls. Gut microbial composition was explored using 16S ribosomal ribonucleic acid sequencing. RESULTS Human immunodeficiency virus-infected children had significantly lower alpha-diversity and higher beta-diversity compared to HIV-uninfected. No association was observed between microbiome diversity and CD4+ T-cell count, HIV viral load, or HIV-associated chronic lung disease. We found enriched levels of Corynebacterium (P < .01), Finegoldia (P < .01), and Anaerococcus (P < .01) in HIV-infected participants and enrichment of Enterobacteriaceae (P = .02) in participants with low CD4+ counts (<400 cells/mm3). Prolonged ART-treatment (≥10 years) was significantly associated with a richer gut microbiota by alpha diversity. CONCLUSIONS Human immunodeficiency virus-infected children have altered gut microbiota. Prolonged ART may restore the richness of the microbiota closer to that of HIV-uninfected children.
Collapse
Affiliation(s)
- Trym T Flygel
- Paediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Evgeniya Sovershaeva
- Paediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Community Medicine, Faculty of Health Sciences, UiT- The Arctic University of Norway, Tromsø, Norway
| | - Shantelle Claassen-Weitz
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Erik Hjerde
- Department of Chemistry, Norstruct, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Kilaza S Mwaikono
- Computational Biology Division, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jon Ø Odland
- Department of Community Medicine, Faculty of Health Sciences, UiT- The Arctic University of Norway, Tromsø, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rashida A Ferrand
- Biomedial Research and Training Institute, Harare, Zimbabwe.,Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Grace Mchugh
- Biomedial Research and Training Institute, Harare, Zimbabwe
| | - Tore J Gutteberg
- Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Mark P Nicol
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Division of Infection and Immunity, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Jorunn P Cavanagh
- Paediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Trond Flægstad
- Paediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | | |
Collapse
|
214
|
Epeldegui M, Hussain SK. The Role of Microbial Translocation and Immune Activation in AIDS-Associated Non-Hodgkin Lymphoma Pathogenesis: What Have We Learned? Crit Rev Immunol 2020; 40:41-51. [PMID: 32421978 PMCID: PMC7241309 DOI: 10.1615/critrevimmunol.2020033319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human immunodeficiency virus (HIV) infection is associated with greatly increased risk for development of non-Hodgkin lymphoma (NHL). Nearly all acquired immunodeficiency syndrome (AIDS)-associated NHL (AIDS-NHL) is of B-cell origin. Two major mechanisms are believed to contribute to the genesis of AIDS-NHL: (1) loss of immunoregulation of Epstein-Barr virus (EBV)+ B cells, resulting from impaired T-cell function late in the course of HIV disease and (2) chronic B-cell activation, leading to DNA-modifying events that contribute to oncogene mutations/ translocations. HIV infection has long been known to be associated with chronic inflammation and polyclonal B-cell activation, and more recently, microbial translocation. Microbial translocation is bacterial product leakage from gut lumen into the peripheral circulation, resulting in high levels of lipopolysaccharide (LPS) in the peripheral circulation, leading to chronic immune activation and inflammation. We review recent literature linking microbial translocation to lymphom-agenesis. This includes epidemiological studies of biomarkers of microbial translocation with risk of AIDS-NHL and emerging data on the mechanisms by which microbial translocation may lead to AIDS-NHL development.
Collapse
Affiliation(s)
- Marta Epeldegui
- Department of Obstetrics and Gynecology, UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles
| | - Shehnaz K. Hussain
- Cedars-Sinai Cancer and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles
| |
Collapse
|
215
|
Sialylation and fucosylation modulate inflammasome-activating eIF2 Signaling and microbial translocation during HIV infection. Mucosal Immunol 2020; 13:753-766. [PMID: 32152415 PMCID: PMC7434596 DOI: 10.1038/s41385-020-0279-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/04/2023]
Abstract
An emerging paradigm suggests that gut glycosylation is a key force in maintaining the homeostatic relationship between the gut and its microbiota. Nevertheless, it is unclear how gut glycosylation contributes to the HIV-associated microbial translocation and inflammation that persist despite viral suppression and contribute to the development of several comorbidities. We examined terminal ileum, right colon, and sigmoid colon biopsies from HIV-infected virally-suppressed individuals and found that gut glycomic patterns are associated with distinct microbial compositions and differential levels of chronic inflammation and HIV persistence. In particular, high levels of the pro-inflammatory hypo-sialylated T-antigen glycans and low levels of the anti-inflammatory fucosylated glycans were associated with higher abundance of glycan-degrading microbial species (in particular, Bacteroides vulgatus), a less diverse microbiome, higher levels of inflammation, and higher levels of ileum-associated HIV DNA. These findings are linked to the activation of the inflammasome-mediating eIF2 signaling pathway. Our study thus provides the first proof-of-concept evidence that a previously unappreciated factor, gut glycosylation, is a force that may impact the vicious cycle between HIV infection, microbial translocation, and chronic inflammation.
Collapse
|
216
|
Dong L, Xie J, Wang Y, Zuo D. Gut Microbiota and Immune Responses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1238:165-193. [PMID: 32323185 DOI: 10.1007/978-981-15-2385-4_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gut microbiota consists of a dynamic multispecies community living within a particular niche in a mutual synergy with the host organism. Recent findings have revealed roles for the gut microbiota in the modulation of host immunity and the development and progression of immune-mediated diseases. Besides, growing evidence supports the concept that some metabolites mainly originated from gut microbiota are linked to the immune regulation implicated in systemic inflammatory and autoimmune disorders. In this chapter, we describe the recent advances in our understanding of how host-microbiota interactions shape the immune system, how they affect the pathogenesis of immune-associated diseases and the impact of these mechanisms in the efficacy of disease therapy.
Collapse
Affiliation(s)
- Lijun Dong
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingwen Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Youyi Wang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- School of Laboratory Medicine and Biotechnology, Institute of Molecular Immunology, Southern Medical University, Guangzhou, 510515, China
| | - Daming Zuo
- School of Laboratory Medicine and Biotechnology, Institute of Molecular Immunology, Southern Medical University, Guangzhou, 510515, China.
- Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
217
|
Naderpoor N, Mousa A, Fernanda Gomez Arango L, Barrett HL, Dekker Nitert M, de Courten B. Effect of Vitamin D Supplementation on Faecal Microbiota: A Randomised Clinical Trial. Nutrients 2019; 11:nu11122888. [PMID: 31783602 PMCID: PMC6950585 DOI: 10.3390/nu11122888] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
In animal studies, vitamin D supplementation has been shown to improve gut microbiota and intestinal inflammation. However, limited evidence exists on the effect of vitamin D supplementation on the human gut microbiota. We examined the effect of vitamin D supplementation on faecal microbiota in 26 vitamin D-deficient (25-hydroxyvitamin D (25(OH)D) ≤50 nmol/L), overweight or obese (BMI ≥25 kg/m2) otherwise healthy adults. Our study was ancillary to a community based double-blind randomised clinical trial, conducted between 2014 and 2016. The participants provided stool samples at baseline and after 100,000 international units (IU) loading dose of cholecalciferol followed by 4000 IU daily or matching placebo for 16 weeks. Faecal microbiota was analysed using 16S rRNA sequencing; V6-8 region. There was no significant difference in microbiome α-diversity between vitamin D and placebo groups at baseline and follow-up (all p > 0.05). In addition, no clustering was found based on vitamin D supplementation at follow-up (p = 0.3). However, there was a significant association between community composition and vitamin D supplementation at the genus level (p = 0.04). The vitamin D group had a higher abundance of genus Lachnospira, and lower abundance of genus Blautia (linear discriminate analysis >3.0). Moreover, individuals with 25(OH)D >75 nmol/L had a higher abundance of genus Coprococcus and lower abundance of genus Ruminococcus compared to those with 25(OH)D <50 nmol/L. Our findings suggest that vitamin D supplementation has some distinct effects on faecal microbiota. Future studies need to explore whether these effects would translate into improved clinical outcomes.
Collapse
Affiliation(s)
- Negar Naderpoor
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, VIC 3168 Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Clayton, VIC 3168, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, VIC 3168 Australia
| | | | - Helen L. Barrett
- Mater Research Institute, The University of Queensland, South Brisbane, QLD 4101, Australia
- Department of Endocrinology, Mater Health, South Brisbane, QLD 4101, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4101, Australia
- Mater Research Institute, The University of Queensland, South Brisbane, QLD 4101, Australia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Clayton, VIC 3168 Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-3-857-22651; Fax: +61-3-9594-7554
| |
Collapse
|
218
|
Tett A, Huang KD, Asnicar F, Fehlner-Peach H, Pasolli E, Karcher N, Armanini F, Manghi P, Bonham K, Zolfo M, De Filippis F, Magnabosco C, Bonneau R, Lusingu J, Amuasi J, Reinhard K, Rattei T, Boulund F, Engstrand L, Zink A, Collado MC, Littman DR, Eibach D, Ercolini D, Rota-Stabelli O, Huttenhower C, Maixner F, Segata N. The Prevotella copri Complex Comprises Four Distinct Clades Underrepresented in Westernized Populations. Cell Host Microbe 2019; 26:666-679.e7. [PMID: 31607556 PMCID: PMC6854460 DOI: 10.1016/j.chom.2019.08.018] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/05/2019] [Accepted: 08/28/2019] [Indexed: 12/29/2022]
Abstract
Prevotella copri is a common human gut microbe that has been both positively and negatively associated with host health. In a cross-continent meta-analysis exploiting >6,500 metagenomes, we obtained >1,000 genomes and explored the genetic and population structure of P. copri. P. copri encompasses four distinct clades (>10% inter-clade genetic divergence) that we propose constitute the P. copri complex, and all clades were confirmed by isolate sequencing. These clades are nearly ubiquitous and co-present in non-Westernized populations. Genomic analysis showed substantial functional diversity in the complex with notable differences in carbohydrate metabolism, suggesting that multi-generational dietary modifications may be driving reduced prevalence in Westernized populations. Analysis of ancient metagenomes highlighted patterns of P. copri presence consistent with modern non-Westernized populations and a clade delineation time pre-dating human migratory waves out of Africa. These findings reveal that P. copri exhibits a high diversity that is underrepresented in Western-lifestyle populations.
Collapse
Affiliation(s)
- Adrian Tett
- CIBIO Department, University of Trento, 38123 Trento, Italy.
| | - Kun D Huang
- CIBIO Department, University of Trento, 38123 Trento, Italy; Department of Sustainable Agro-Ecosystems and Bioresources, Fondazione Edmund Mach, 1 38010 S, San Michele all'Adige, Italy
| | | | - Hannah Fehlner-Peach
- Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | - Paolo Manghi
- CIBIO Department, University of Trento, 38123 Trento, Italy
| | - Kevin Bonham
- The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA; Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy
| | - Moreno Zolfo
- CIBIO Department, University of Trento, 38123 Trento, Italy
| | - Francesca De Filippis
- Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy; Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Cara Magnabosco
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA; Departments of Biology and Computer Science, New York University, New York, NY 10003, USA
| | - John Lusingu
- National Institute for Medical Research, Tanga Centre, Tanzania
| | - John Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Ghana
| | - Karl Reinhard
- Hardin Hall, School of Natural Resources, University of Nebraska, Lincoln, NE 68583-0987, USA
| | - Thomas Rattei
- CUBE - Division of Computational Systems Biology, Department of Microbiology and Ecosystem Science, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Fredrik Boulund
- Centre for Translational Microbiome Research, Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 171 65 Solna, Stockholm, Sweden
| | - Lars Engstrand
- Institute for Mummy Studies, EURAC Research, Viale Druso 1, 39100 Bolzano, Italy
| | - Albert Zink
- Institute for Mummy Studies, EURAC Research, Viale Druso 1, 39100 Bolzano, Italy
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
| | - Dan R Littman
- Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Daniel Eibach
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; German Center for Infection Research, Hamburg-Borstel-Lübeck-Riems, 20359 Hamburg, Germany
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy; Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Omar Rota-Stabelli
- Department of Sustainable Agro-Ecosystems and Bioresources, Fondazione Edmund Mach, 1 38010 S, San Michele all'Adige, Italy
| | - Curtis Huttenhower
- The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA; Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Frank Maixner
- Institute for Mummy Studies, EURAC Research, Viale Druso 1, 39100 Bolzano, Italy
| | - Nicola Segata
- CIBIO Department, University of Trento, 38123 Trento, Italy.
| |
Collapse
|
219
|
Pellicano C, Leodori G, Innocenti GP, Gigante A, Rosato E. Microbiome, Autoimmune Diseases and HIV Infection: Friends or Foes? Nutrients 2019; 11:E2629. [PMID: 31684052 PMCID: PMC6893726 DOI: 10.3390/nu11112629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
Several studies highlighted the importance of the interaction between microbiota and the immune system in the development and maintenance of the homeostasis of the human organism. Dysbiosis is associated with proinflammatory and pathological state-like metabolic diseases, autoimmune diseases and HIV infection. In this review, we discuss the current understanding of the possible role of dysbiosis in triggering and/or exacerbating symptoms of autoimmune diseases and HIV infection. There are no data about the influence of the microbiome on the development of autoimmune diseases during HIV infection. We can hypothesize that untreated patients may be more susceptible to the development of autoimmune diseases, due to the presence of dysbiosis. Eubiosis, re-established by probiotic administration, can be used to reduce triggers for autoimmune diseases in untreated HIV patients, although clinical studies are needed to evaluate the role of the microbiome in autoimmune diseases in HIV patients.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Giorgia Leodori
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Antonietta Gigante
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Edoardo Rosato
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
220
|
AKO SE, AKUM EA, NKENFOU CN, ASSOB JCN, POKAM TB. In-vitro susceptibility of gut pathobiont associated with microbial translocation to cotrimoxazole and antiretroviral. SCIENTIFIC AFRICAN 2019. [DOI: 10.1016/j.sciaf.2019.e00192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
221
|
Samuelson DR, Siggins RW, Ruan S, Amedee AM, Sun J, Zhu QK, Marasco WA, Taylor CM, Luo M, Welsh DA, Shellito JE. Alcohol consumption increases susceptibility to pneumococcal pneumonia in a humanized murine HIV model mediated by intestinal dysbiosis. Alcohol 2019; 80:33-43. [PMID: 30213614 PMCID: PMC6449221 DOI: 10.1016/j.alcohol.2018.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023]
Abstract
Alcohol use in persons living with HIV (PLWH) worsens the severity of bacterial pneumonia. However, the exact mechanism(s) by which this occurs remain ill-defined. We hypothesized that alcohol in the setting of HIV infection decreases Streptococcus pneumoniae clearance from the lung through mechanisms mediated by the gut microbiota. Humanized BLT (bone marrow, liver, thymus) mice were infected with 1 × 104 TCID50 of HIV (BAL and JRCSF strains) via intraperitoneal (i.p.) injection. One week post-HIV infection, animals were switched to a Lieber-DeCarli 5% ethanol diet or an isocaloric control diet for 10 days. Alcohol-fed animals were also given two binges of 2 g/kg ethanol on days 5 and 10. Feces were also collected, banked, and the community structures were analyzed. Mice were then infected with 1 × 105 CFU (colony-forming units) of S. pneumoniae and were sacrificed 48 h later. HIV-infected mice had viral loads of ∼2 × 104 copies/mL of blood 1 week post-infection, and exhibited an ∼57% decrease in the number of circulating CD4+ T cells at the time of sacrifice. Fecal microbial community structure was significantly different in each of the feeding groups, as well as with HIV infection. Alcohol-fed mice had a significantly higher burden of S. pneumoniae 48 h post-infection, regardless of HIV status. In follow-up experiments, female C57BL/6 mice were treated with a cocktail of antibiotics daily for 2 weeks and recolonized by gavage with intestinal microbiota from HIV+ ethanol-fed, HIV+ pair-fed, HIV- ethanol-fed, or HIV- pair-fed mice. Recolonized mice were then infected with S. pneumoniae and were sacrificed 48 h later. The intestinal microbiota from alcohol-fed mice (regardless of HIV status) significantly impaired clearance of S. pneumoniae. Collectively, these data indicate that alcohol feeding, as well as alcohol-associated intestinal dysbiosis, compromise pulmonary host defenses against pneumococcal pneumonia. Determining whether HIV infection acts synergistically with alcohol use in impairing pulmonary host defenses will require additional study.
Collapse
Affiliation(s)
- Derrick R Samuelson
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Robert W Siggins
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Sanbao Ruan
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Angela M Amedee
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jiusong Sun
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, United States
| | - Quan Karen Zhu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, United States
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School Boston, MA, United States
| | - Christopher M Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - David A Welsh
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Sciences Center, New Orleans, LA, United States; Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Judd E Shellito
- Department of Internal Medicine, Section of Pulmonary/Critical Care & Allergy/Immunology, Louisiana State University Health Sciences Center, New Orleans, LA, United States; Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States.
| |
Collapse
|
222
|
Fulcher JA, Li F, Cook RR, Zabih S, Louie A, Okochi H, Tobin NH, Gandhi M, Shoptaw S, Gorbach PM, Aldrovandi GM. Rectal Microbiome Alterations Associated With Oral Human Immunodeficiency Virus Pre-Exposure Prophylaxis. Open Forum Infect Dis 2019; 6:ofz463. [PMID: 32258202 PMCID: PMC7105055 DOI: 10.1093/ofid/ofz463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Oral daily tenofovir (TFV) disoproxil fumarate/emtricitabine (TDF/FTC) for human immunodeficiency virus (HIV) pre-exposure prophylaxis (PrEP) is highly effective for HIVprevention, yet long-term effects are not fully understood. We investigated the effects of PrEP on the rectal microbiome in a cohort of men who have sex with men (MSM). METHODS This cross-sectional analysis included HIV-negative MSM either on PrEP (n = 37) or not (n = 37) selected from an ongoing cohort using propensity score matching. Rectal swabs were used to examine microbiome composition using 16S ribosomal ribonucleic acid gene sequencing, and associations between PrEP use and microbiota abundance were examined. Hair specimens were used to quantify TFV and FTC exposure over the past 6 weeks on a subset of participants (n = 15). RESULTS Pre-exposure prophylaxis use was associated with a significant increase in Streptococcus abundance (adjusted P = .015). Similar associations were identified using least absolute shrinkage and selection operator (LASSO) regression, confirming the increase in Streptococcus and also showing increased Mitsuokella, Fusobacterium, and decreased Escherichia/Shigella. Increased Fusobacterium was significantly associated with increasing TFV exposure. CONCLUSIONS Oral TDF/FTC for PrEP is associated with rectal microbiome changes compared to well matched controls, specifically increased Streptococcus and Fusobacterium abundance. This study highlights the need for future investigations of the role of microbiome changes on HIV susceptibility and effectiveness of PrEP.
Collapse
Affiliation(s)
- Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Fan Li
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ryan R Cook
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Sara Zabih
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Alexander Louie
- Division of HIV, Infectious Diseases, and Global Medicine (Hair Analytical Laboratory), Department of Medicine, University of California, San Francisco, California, USA
| | - Hideaki Okochi
- Division of HIV, Infectious Diseases, and Global Medicine (Hair Analytical Laboratory), Department of Medicine, University of California, San Francisco, California, USA
| | - Nicole H Tobin
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Monica Gandhi
- Division of HIV, Infectious Diseases, and Global Medicine (Hair Analytical Laboratory), Department of Medicine, University of California, San Francisco, California, USA
| | - Steven Shoptaw
- Department of Family Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Pamina M Gorbach
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Grace M Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
223
|
Effect of Lactobacillus rhamnosus GG Supplementation on Intestinal Inflammation Assessed by PET/MRI Scans and Gut Microbiota Composition in HIV-Infected Individuals. J Acquir Immune Defic Syndr 2019; 78:450-457. [PMID: 29874201 DOI: 10.1097/qai.0000000000001693] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alterations in the gut microbiome have been associated with inflammation and increased cardiovascular risk in HIV-infected individuals. The aim of this study was to investigate the effects of the probiotic strain Lactobacillus rhamnosus GG (LGG) on intestinal inflammation, gut microbiota composition, and systemic markers of microbial translocation and inflammation in HIV-infected individuals. METHODS This prospective, clinical interventional trial included 45 individuals [15 combination antiretroviral treatment (cART) naive and 30 cART treated] who ingested LGG twice daily at a dosage of 6 × 109 colony-forming units per capsule for a period of 8 weeks. Intestinal inflammation was assessed using F-2-fluoro-2-deoxy-D-glucose positron emission tomography/magnetic resonance imaging (F-FDG PET/MRI) scans in 15 individuals. Gut microbiota composition (V3-V4 region of the 16s rRNA gene) and markers of microbial translocation and inflammation (lipopolysaccharide, sCD14, sCD163, sCD25, high-sensitive CRP, IL-6, and tumor necrosis factor-alpha) were analyzed at baseline and after intervention. RESULTS At baseline, evidence of intestinal inflammation was found in 75% of the participants, with no significant differences between cART-naive and cART-treated individuals. After LGG supplementation, a decrease in intestinal inflammation was detected on PET/MRI (-0.3 mean difference in the combined activity grade score from 6 regions, P = 0.006), along with a reduction of Enterobacteriaceae (P = 0.018) and Erysipelotrichaceae (P = 0.037) in the gut microbiome, with reduced Enterobacteriaceae among individuals with decreased F-FDG uptake on PET/MRI (P = 0.048). No changes were observed for soluble markers of microbial translocation and inflammation. CONCLUSIONS A decrease in intestinal inflammation was found in HIV-infected individuals after ingestion of LGG along with a reduced abundance of Enterobacteriaceae, which may explain the local anti-inflammatory effect in the gut.
Collapse
|
224
|
Olalla J, García de Lomas JM, Chueca N, Pérez-Stachowski X, De Salazar A, Del Arco A, Plaza-Díaz J, De la Torre J, Prada JL, García-Alegría J, Fernández-Sánchez F, García F. Effect of daily consumption of extra virgin olive oil on the lipid profile and microbiota of HIV-infected patients over 50 years of age. Medicine (Baltimore) 2019; 98:e17528. [PMID: 31626113 PMCID: PMC6824693 DOI: 10.1097/md.0000000000017528] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Extra virgin olive oil (EVOO) has shown beneficial effects on the lipid profile and inflammatory parameters in general population. Our goal is to analyze these changes together with those of intestinal microbiota in human immunodeficiency virus (HIV)-infected patients over 50 years of age. METHODS Experimental single arm open study. HIV patients over the age of 50 with undetectable viral load were selected. EVOO was distributed among the patients so that each one consumed 50 g daily for 12 weeks. Lipid profile, C-reactive protein (CRP), and intestinal microbiota composition were analyzed at the beginning and at the end of the intervention. RESULTS Total cholesterol decreased significantly (5 mg/dL), and a nonsignificant decrease in low-density lipoprotein cholesterol (12 mg/dL), triglycerides (21 mg/dL), and CRP (1.25 mg/dL) was observed. There was a significant increase in alpha diversity after the intervention in men and a decrease in proinflammatory genera such as Dethiosulfovibrionaceae was observed. Differences were also observed in the microbiota of men and women and according to the type of antiretroviral treatment. CONCLUSION Sustained consumption of 50 g of EVOO in elderly HIV-infected patients might be associated with an improvement in lipid profile and alfa diversity of intestinal microbiota.
Collapse
Affiliation(s)
- Julián Olalla
- Servicio de Medicina Interna, Hospital Costa del Sol, Marbella
| | | | | | | | | | | | - Julio Plaza-Díaz
- Instituto de Investigación Biosanitaria IBS
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Granada
| | | | - José Luis Prada
- Servicio de Medicina Interna, Hospital Costa del Sol, Marbella
| | | | | | | |
Collapse
|
225
|
Williams BB, Green SJ, Bosch RJ, Chan ES, Jacobson JM, Margolis DM, Engen P, Landay AL, Wilson CC, A5286 protocol team. Four Weeks of Treatment With Rifaximin Fails to Significantly Alter Microbial Diversity in Rectal Samples of HIV-Infected Immune Non-Responders (ACTG A5286) Which May be Attributed to Rectal Swab Use. Pathog Immun 2019; 4:235-250. [PMID: 31583331 PMCID: PMC6768152 DOI: 10.20411/pai.v4i2.290] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/20/2019] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION HIV-infected individuals have evidence of intestinal microbial translocation which is associated with immune activation and unfavorable clinical outcomes. Rifaximin, a non-absorbable antibiotic which reduces microbial translocation in other disease states, was shown to have a marginal beneficial effect on microbial translocation, T-cell activation, and inflammation in a multisite randomized trial (ACTG A5286; NCT01466595) of HIV-infected persons with poor immunologic recovery receiving ART. Here, we report analysis of the rectal microbiome changes associated with that trial. METHODS HIV-1-infected individuals receiving ART with CD4-T cell count < 350cells/mm3 and viral suppression were randomized 2:1 to rifaximin or no therapy for 4 weeks. Rectal swabs were collected at baseline (pre-treatment) and at week 4 of rifaximin therapy. Genomic DNA extracted from rectal swab samples was analyzed using high throughput sequencing and quantitative PCR of bacterial 16S ribosomal RNA (rRNA) genes. RESULTS Forty-eight HIV-infected participants (31 received rifaximin, 17 no treatment) were included. There was broad variability in the recovery of bacterial rRNA from the specimens at baseline. No major significant (FDR P < 0.05) effects of rifaximin treatment on alpha- or beta-diversity or individual taxa were observed between or within the treatment arms, with analyses conducted at taxonomic levels from phylum to genus. CONCLUSIONS Rifaximin did not meaningfully alter the diversity or composition of the rectal microbiome of HIV-infected individuals after 4 weeks of therapy, although rectal swab specimens varied widely in their microbial load.
Collapse
Affiliation(s)
- Brett B. Williams
- Division of Infectious Disease; Rush University Medical Center; Chicago, Illinois
| | - Stefan J. Green
- Sequencing Core; University of Illinois at Chicago; Chicago, Illinois
| | - Ronald J. Bosch
- Center for Biostatistics in AIDS Research; Harvard School of Public Health; Boston, Massachusetts
| | - Ellen S. Chan
- Center for Biostatistics in AIDS Research; Harvard School of Public Health; Boston, Massachusetts
| | - Jeffrey M. Jacobson
- Division of Infectious Diseases and HIV; Drexel University; Philadelphia, Pennsylvania
| | - David M. Margolis
- Department of Medicine; University of North Carolina; Chapel Hill, North Carolina
| | - Phillip Engen
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Rush University Medical Center; Chicago, Illinois
| | - Alan L. Landay
- Department of Immunology and Microbiology; Rush University Medical Center; Chicago, Illinois
| | - Cara C. Wilson
- Department of Medicine; University of Colorado at Denver; Aurora, Colorado
| | - A5286 protocol team
- Division of Infectious Disease; Rush University Medical Center; Chicago, Illinois
- Sequencing Core; University of Illinois at Chicago; Chicago, Illinois
- Center for Biostatistics in AIDS Research; Harvard School of Public Health; Boston, Massachusetts
- Division of Infectious Diseases and HIV; Drexel University; Philadelphia, Pennsylvania
- Department of Medicine; University of North Carolina; Chapel Hill, North Carolina
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Rush University Medical Center; Chicago, Illinois
- Department of Immunology and Microbiology; Rush University Medical Center; Chicago, Illinois
- Department of Medicine; University of Colorado at Denver; Aurora, Colorado
| |
Collapse
|
226
|
Ortiz AM, Flynn JK, DiNapoli SR, Sortino O, Vujkovic-Cvijin I, Belkaid Y, Sereti I, Brenchley JM. Antiretroviral Therapy Administration in Healthy Rhesus Macaques Is Associated with Transient Shifts in Intestinal Bacterial Diversity and Modest Immunological Perturbations. J Virol 2019; 93:e00472-19. [PMID: 31270225 PMCID: PMC6714794 DOI: 10.1128/jvi.00472-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/16/2019] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal (GI) immune system competency is dependent upon interactions with commensal microbiota, which can be influenced by wide-ranging pharmacologic interventions. In simian immunodeficiency virus (SIV)-infected Asian macaque models of human immunodeficiency virus (HIV) infection, we previously noted that initiation of antiretroviral therapy (ART) is associated with a specific imbalance (dysbiosis) of the composition of the intestinal bacteriome. To determine if ART itself might contribute to dysbiosis or immune dysfunction, we treated healthy rhesus macaques with protease, integrase, or reverse transcriptase inhibitors for 1 to 2 or for 5 to 6 weeks and evaluated intestinal immune function and the composition of the fecal bacterial microbiome. We observed that individual antiretrovirals (ARVs) modestly altered intestinal T-cell proinflammatory responses without disturbing total or activated T-cell frequencies. Moreover, we observed transient disruptions in bacterial diversity coupled with perturbations in the relative frequencies of bacterial communities. Shifts in specific bacterial frequencies were not persistent posttreatment, however, with individual taxa showing only isolated associations with T-cell proinflammatory responses. Our findings suggest that intestinal bacterial instability and modest immunological alterations can result from ART itself. These data could lead to therapeutic interventions which stabilize the microbiome in individuals prescribed ART.IMPORTANCE Dysbiosis of the fecal microbiome is a common feature observed in ARV-treated people living with HIV. The degree to which HIV infection itself causes this dysbiosis remains unclear. Here, we demonstrate that medications used to treat HIV infection can influence the composition of the GI tract immune responses and its microbiome in the nonhuman primate SIV model.
Collapse
Affiliation(s)
- Alexandra M Ortiz
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Jacob K Flynn
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Sarah R DiNapoli
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Ornella Sortino
- HIV Pathogenesis Section, Laboratory of Immunoregulation, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Ivan Vujkovic-Cvijin
- Metaorganism Immunity Section, Laboratory of Immune Systems Biology, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune Systems Biology, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| |
Collapse
|
227
|
Younas M, Psomas C, Reynes C, Cezar R, Kundura L, Portales P, Merle C, Atoui N, Fernandez C, Le Moing V, Barbuat C, Moranne O, Sotto A, Sabatier R, Fabbro P, Vincent T, Dunyach-Remy C, Winter A, Reynes J, Lavigne JP, Corbeau P. Microbial Translocation Is Linked to a Specific Immune Activation Profile in HIV-1-Infected Adults With Suppressed Viremia. Front Immunol 2019; 10:2185. [PMID: 31572392 PMCID: PMC6753629 DOI: 10.3389/fimmu.2019.02185] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
Persistent immune activation in virologically suppressed HIV-1 patients, which may be the consequence of various factors including microbial translocation, is a major cause of comorbidities. We have previously shown that different profiles of immune activation may be distinguished in virological responders. Here, we tested the hypothesis that a particular profile might be the consequence of microbial translocation. To this aim, we measured 64 soluble and cell surface markers of inflammation and CD4+ and CD8+ T-cell, B cell, monocyte, NK cell, and endothelial activation in 140 adults under efficient antiretroviral therapy, and classified patients and markers using a double hierarchical clustering analysis. We also measured the plasma levels of the microbial translocation markers bacterial DNA, lipopolysaccharide binding protein (LBP), intestinal-fatty acid binding protein, and soluble CD14. We identified five different immune activation profiles. Patients with an immune activation profile characterized by a high percentage of CD38+CD8+ T-cells and a high level of the endothelial activation marker soluble Thrombomodulin, presented with higher LBP mean (± SEM) concentrations (33.3 ± 1.7 vs. 28.7 ± 0.9 μg/mL, p = 0.025) than patients with other profiles. Our data are consistent with the hypothesis that the immune activation profiles we described are the result of different etiological factors. We propose a model, where particular causes of immune activation, as microbial translocation, drive particular immune activation profiles responsible for particular comorbidities.
Collapse
Affiliation(s)
- Mehwish Younas
- Institute of Human Genetics, CNRS-Montpellier University, UMR9002, Montpellier, France
| | - Christina Psomas
- Institute of Human Genetics, CNRS-Montpellier University, UMR9002, Montpellier, France.,Infectious Diseases Department, University Hospital, Montpellier, France
| | - Christelle Reynes
- Institute for Functional Genomics, Montpellier University, UMR5203, Montpellier, France
| | - Renaud Cezar
- Immunology Department, University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, CNRS-Montpellier University, UMR9002, Montpellier, France
| | - Pierre Portales
- Immunology Department, University Hospital, Montpellier, France
| | - Corinne Merle
- Infectious Diseases Department, University Hospital, Montpellier, France
| | - Nadine Atoui
- Infectious Diseases Department, University Hospital, Montpellier, France
| | - Céline Fernandez
- Infectious Diseases Department, University Hospital, Montpellier, France
| | - Vincent Le Moing
- Infectious Diseases Department, University Hospital, Montpellier, France.,IRD UMI 233, INSERM U1175, Montpellier University, Montpellier, France.,Montpellier University, Montpellier, France
| | - Claudine Barbuat
- Infectious Diseases Department, University Hospital, Nîmes, France
| | | | - Albert Sotto
- Montpellier University, Montpellier, France.,Infectious Diseases Department, University Hospital, Nîmes, France
| | - Robert Sabatier
- Institute for Functional Genomics, Montpellier University, UMR5203, Montpellier, France
| | - Pascale Fabbro
- Medical Informatics Department, University Hospital, Nîmes, France
| | - Thierry Vincent
- Immunology Department, University Hospital, Montpellier, France.,Montpellier University, Montpellier, France
| | - Catherine Dunyach-Remy
- U1047, INSERM, Microbiology University Hospital Nîmes, Montpellier University, Nîmes, France
| | - Audrey Winter
- Institute of Human Genetics, CNRS-Montpellier University, UMR9002, Montpellier, France
| | - Jacques Reynes
- Infectious Diseases Department, University Hospital, Montpellier, France.,IRD UMI 233, INSERM U1175, Montpellier University, Montpellier, France.,Montpellier University, Montpellier, France
| | - Jean-Philippe Lavigne
- U1047, INSERM, Microbiology University Hospital Nîmes, Montpellier University, Nîmes, France
| | - Pierre Corbeau
- Institute of Human Genetics, CNRS-Montpellier University, UMR9002, Montpellier, France.,Immunology Department, University Hospital, Nîmes, France.,Montpellier University, Montpellier, France
| |
Collapse
|
228
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
229
|
Moran LM, McLaurin KA, Booze RM, Mactutus CF. Neurorestoration of Sustained Attention in a Model of HIV-1 Associated Neurocognitive Disorders. Front Behav Neurosci 2019; 13:169. [PMID: 31447657 PMCID: PMC6691343 DOI: 10.3389/fnbeh.2019.00169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Due to the sustained prevalence of human immunodeficiency virus (HIV)-1 associated neurocognitive disorders (HAND) in the post-combination antiretroviral therapy (cART) era, as well as the increased prevalence of older HIV-1 seropositive individuals, there is a critical need to develop adjunctive therapeutics targeted at preserving and/or restoring neurocognitive function. To address this knowledge gap, the present study examined the utility of S-Equol (SE), a phytoestrogen produced by gut microbiota, as an innovative therapeutic strategy. A signal detection operant task with varying signal durations (1,000, 500, 100 ms) was utilized to assess sustained attention in HIV-1 transgenic (Tg) and control animals. During the signal detection pretest assessment, HIV-1 Tg animals displayed profound deficits in stimulus-response learning and sustained attention relative to control animals. Subsequently, between 6 and 8 months of age, HIV-1 Tg and control animals were treated with a daily oral dose of either placebo or SE (0.05, 0.1, 0.2 mg) and a posttest assessment was conducted in the signal detection operant task with varying signal durations. In HIV-1 Tg animals, a linear decrease in the number of misses at 100 ms was observed as SE dose increased, suggesting a dose response with the most effective dose at 0.2 mg SE, approximating controls. Comparison of the number of misses across signal durations at the pretest and posttest revealed a preservation of neurocognitive function in HIV-1 Tg animals treated with 0.2 mg SE; an effect that was in sharp contrast to the neurocognitive decline observed in HIV-1 Tg animals treated with placebo. The results support the utility of 0.2 mg SE as a potential efficacious neuroprotective and/or neurorestorative therapeutic for sustained attention, in the absence of any adverse peripheral effects, in the HIV-1 Tg rat. Thus, the present study highlights the critical need for further in vivo studies to elucidate the full potential and generalizability of phytoestrogen treatment for HAND.
Collapse
Affiliation(s)
- Landhing M Moran
- Program in Behavioral Neuroscience, Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Kristen A McLaurin
- Program in Behavioral Neuroscience, Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Rosemarie M Booze
- Program in Behavioral Neuroscience, Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Charles F Mactutus
- Program in Behavioral Neuroscience, Department of Psychology, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
230
|
Alzahrani J, Hussain T, Simar D, Palchaudhuri R, Abdel-Mohsen M, Crowe SM, Mbogo GW, Palmer CS. Inflammatory and immunometabolic consequences of gut dysfunction in HIV: Parallels with IBD and implications for reservoir persistence and non-AIDS comorbidities. EBioMedicine 2019; 46:522-531. [PMID: 31327693 PMCID: PMC6710907 DOI: 10.1016/j.ebiom.2019.07.027] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal mucosa is critical for maintaining the integrity and functions of the gut. Disruption of this barrier is a hallmark and a risk factor for many intestinal and chronic inflammatory diseases. Inflammatory bowel disease (IBD) and HIV infection are characterized by microbial translocation and systemic inflammation. Despite the clinical overlaps between HIV and IBD, significant differences exist such as the severity of gut damage and mechanisms of immune cell homeostasis. Studies have supported the role of metabolic activation of immune cells in promoting chronic inflammation in HIV and IBD. This inflammatory response persists in HIV+ persons even after long-term virologic suppression by antiretroviral therapy (ART). Here, we review gut dysfunction and microbiota changes during HIV infection and IBD, and discuss how this may induce metabolic reprogramming of monocytes, macrophages and T cells to impact disease outcomes. Drawing from parallels with IBD, we highlight how factors such as lipopolysaccharides, residual viral replication, and extracellular vesicles activate biochemical pathways that regulate immunometabolic processes essential for HIV persistence and non-AIDS metabolic comorbidities. This review highlights new mechanisms and support for the use of immunometabolic-based therapeutics towards HIV remission/cure, and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jehad Alzahrani
- Life Sciences, Burnet Institute, Melbourne, Australia; School of Medical Science, RMIT University, Melbourne, Australia
| | - Tabinda Hussain
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David Simar
- School of Medical Sciences, UNSW, Sydney, Australia
| | | | | | - Suzanne M Crowe
- Life Sciences, Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, Monash University, Melbourne, Australia
| | | | - Clovis S Palmer
- Life Sciences, Burnet Institute, Melbourne, Australia; School of Medical Science, RMIT University, Melbourne, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
231
|
Nijmeijer BM, Geijtenbeek TBH. Negative and Positive Selection Pressure During Sexual Transmission of Transmitted Founder HIV-1. Front Immunol 2019; 10:1599. [PMID: 31354736 PMCID: PMC6635476 DOI: 10.3389/fimmu.2019.01599] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
Sexual transmission of HIV-1 consists of processes that exert either positive or negative selection pressure on the virus. The sum of these selection pressures lead to the transmission of only one specific HIV-1 strain, termed the transmitted founder virus. Different dendritic cell subsets are abundantly present at mucosal sites and, interestingly, these DC subsets exert opposite pressure on viral selection during sexual transmission. In this review we describe receptors and cellular compartments in DCs that are involved in HIV-1 communication leading to either viral restriction by the host or further dissemination to establish a long-lived reservoir. We discuss the current understanding of host antiretroviral restriction factors against HIV-1 and specifically against the HIV-1 transmitted founder virus. We will also discuss potential clinical implications for exploiting these intrinsic restriction factors in developing novel therapeutic targets. A better understanding of these processes might help in developing strategies against HIV-1 infections by targeting dendritic cells.
Collapse
Affiliation(s)
- Bernadien M Nijmeijer
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
232
|
Herrera S, Martínez-Sanz J, Serrano-Villar S. HIV, Cancer, and the Microbiota: Common Pathways Influencing Different Diseases. Front Immunol 2019; 10:1466. [PMID: 31316514 PMCID: PMC6610485 DOI: 10.3389/fimmu.2019.01466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
HIV infection exerts profound and perhaps irreversible damage to the gut mucosal-associated lymphoid tissues, resulting in long-lasting changes in the signals required for the coordination of commensal colonization and in perturbations at the compositional and functional level of the gut microbiota. These abnormalities in gut microbial communities appear to affect clinical outcomes, including T-cell recovery, vaccine responses, HIV transmission, cardiovascular disease, and cancer pathogenesis. For example, the microbial signature associated with HIV infection has been shown to induce tryptophan catabolism, affect the butyrate synthesis pathway, impair anti-tumoral immunity and affect oxidative stress, which have also been linked to the pathogenesis of cancer. Furthermore, some of the taxa that are depleted in subjects with HIV have proved to modulate the anti-tumor efficacy of various chemotherapies and immunotherapeutic agents. The aim of this work is to provide a broad overview of recent advances in our knowledge of how HIV might affect the microbiota, with a focus on the pathways shared with cancer pathogenesis.
Collapse
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Javier Martínez-Sanz
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| |
Collapse
|
233
|
Abstract
PURPOSE OF REVIEW We aim to provide an in-depth review of recent literature highlighting the role of inflammation involving the adipose tissue, liver, skeletal muscles, and gastrointestinal tract in the development of metabolic complications among persons living with HIV (PLWH). RECENT FINDINGS Recent studies in PLWH have demonstrated a significant association between circulating inflammatory markers and development of insulin resistance and metabolic complications. In adipose tissue, pro-inflammatory cytokine expression inhibits adipocyte insulin signaling, which alters lipid and glucose homeostasis. Increased lipolysis and lipogenesis elevate levels of circulating free fatty acids and promote ectopic fat deposition in liver and skeletal muscles. This leads to lipotoxicity characterized by a pro-inflammatory response with worsening insulin resistance. Finally, HIV is associated with gastrointestinal tract inflammation and changes in the gut microbiome resulting in reduced diversity, which is an additional risk factor for diabetes. Metabolic complications in PLWH are in part due to chronic, multisite tissue inflammation resulting in dysregulation of glucose and lipid trafficking, utilization, and storage.
Collapse
|
234
|
Sindberg GM, Callen SE, Banerjee S, Meng J, Hale VL, Hegde R, Cheney PD, Villinger F, Roy S, Buch S. Morphine Potentiates Dysbiotic Microbial and Metabolic Shifts in Acute SIV Infection. J Neuroimmune Pharmacol 2019; 14:200-214. [PMID: 30242614 PMCID: PMC6917429 DOI: 10.1007/s11481-018-9805-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022]
Abstract
Human Immunodeficiency Virus (HIV) pathogenesis has been closely linked with microbial translocation, which is believed to drive inflammation and HIV replication. Opioid drugs have been shown to worsen this symptom, leading to a faster progression of HIV infection to Acquired Immunodeficiency Syndrome (AIDS). The interaction of HIV and opioid drugs has not been studied at early stages of HIV, particularly in the gut microbiome where changes may precede translocation events. This study modeled early HIV infection by examining Simian Immunodeficiency Virus (SIV)-infected primates at 21 days or less both independently and in the context of opioid use. Fecal samples were analyzed both for 16S analysis of microbial populations as well as metabolite profiles via mass spectrometry. Our results indicate that changes are minor in SIV treated animals in the time points examined, however animals treated with morphine and SIV had significant changes in their microbial communities and metabolic profiles. This occurred in a time-independent fashion with morphine regardless of how long the animal had morphine in its system. Globally, the observed changes support that microbial dysbiosis is occurring in these animals at an early time, which likely contributes to the translocation events observed later in SIV/HIV pathogenesis. Additionally, metabolic changes were predictive of specific treatment groups, which could be further developed as a diagnostic tool or future intervention target to overcome and slow the progression of HIV infection to AIDS.
Collapse
Affiliation(s)
- Gregory M Sindberg
- Department of Veterinary Biosciences, University of Minnesota, Saint Paul, MN, USA
| | - Shannon E Callen
- Department of Pharmacology, University of Nebraska, Omaha, NE, USA
| | - Santanu Banerjee
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Surgery, University of Miami, Miami, Florida, USA
| | - Jingjing Meng
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Surgery, University of Miami, Miami, Florida, USA
| | - Vanessa L Hale
- Department of Veterinary Preventative Medicine, College of Veterinary Medicine, Ohio State University, Columbus, OH, USA
| | - Ramakrishna Hegde
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Paul D Cheney
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Sabita Roy
- Department of Veterinary Biosciences, University of Minnesota, Saint Paul, MN, USA.
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
- Department of Surgery, University of Miami, Miami, Florida, USA.
| | - Shilpa Buch
- Department of Pharmacology, University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
235
|
Crakes KR, Jiang G. Gut Microbiome Alterations During HIV/SIV Infection: Implications for HIV Cure. Front Microbiol 2019; 10:1104. [PMID: 31191468 PMCID: PMC6539195 DOI: 10.3389/fmicb.2019.01104] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Gut mucosal damage, associated with Human Immunodeficiency Virus-1 (HIV) infection, is characterized by depletion in CD4+ T cells and persistent immune activation as a result of early epithelial barrier disruption and systemic translocation of microbial products. Unique approaches in studying both HIV infection in human patients and Simian Immunodeficiency Virus (SIV) infection in rhesus macaques have provided critical evidence for the pathogenesis and treatment of HIV/AIDS. While there is vast resemblance between SIV and HIV infection, the development of gut dysbiosis attributed to HIV infection in chronically infected patients has not been consistently reported in SIV infection in the non-human primate model of AIDS, raising concerns for the translatability of gut microbiome studies in rhesus macaques. This review outlines our current understanding of gut microbial signatures across various stages of HIV versus SIV infection, with an emphasis on the impact of microbiome-based therapies in restoring gut mucosal immunity as well as their translational potential to supplement current HIV cure efforts.
Collapse
Affiliation(s)
- Katti R. Crakes
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
| | - Guochun Jiang
- Department of Biochemistry and Biophysics, Institute for Global Health & Infectious Diseases, UNC HIV Cure Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
236
|
Rocafort M, Noguera-Julian M, Rivera J, Pastor L, Guillén Y, Langhorst J, Parera M, Mandomando I, Carrillo J, Urrea V, Rodríguez C, Casadellà M, Calle ML, Clotet B, Blanco J, Naniche D, Paredes R. Evolution of the gut microbiome following acute HIV-1 infection. MICROBIOME 2019; 7:73. [PMID: 31078141 PMCID: PMC6511141 DOI: 10.1186/s40168-019-0687-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/22/2019] [Indexed: 05/12/2023]
Abstract
BACKGROUND In rhesus macaques, simian immunodeficiency virus infection is followed by expansion of enteric viruses but has a limited impact on the gut bacteriome. To understand the longitudinal effects of HIV-1 infection on the human gut microbiota, we prospectively followed 49 Mozambican subjects diagnosed with recent HIV-1 infection (RHI) and 54 HIV-1-negative controls for 9-18 months and compared them with 98 chronically HIV-1-infected subjects treated with antiretrovirals (n = 27) or not (n = 71). RESULTS We show that RHI is followed by increased fecal adenovirus shedding, which persists during chronic HIV-1 infection and does not resolve with ART. Recent HIV-1 infection is also followed by transient non-HIV-specific changes in the gut bacterial richness and composition. Despite early resilience to change, an HIV-1-specific signature in the gut bacteriome-featuring depletion of Akkermansia, Anaerovibrio, Bifidobacterium, and Clostridium-previously associated with chronic inflammation, CD8+ T cell anergy, and metabolic disorders, can be eventually identified in chronically HIV-1-infected subjects. CONCLUSIONS Recent HIV-1 infection is associated with increased fecal shedding of eukaryotic viruses, transient loss of bacterial taxonomic richness, and long-term reductions in microbial gene richness. An HIV-1-associated microbiome signature only becomes evident in chronically HIV-1-infected subjects.
Collapse
Affiliation(s)
- Muntsa Rocafort
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain
| | - Javier Rivera
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain
| | - Lucía Pastor
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Institut Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autonoma de Barcelona, 08916, Badalona, Catalonia, Spain
- Centro de Investigação em Saúde da Manhiça (CISM), 1929, Maputo, Mozambique
| | - Yolanda Guillén
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Jost Langhorst
- Integrative Gastroenterology, Kliniken Essen-Mitte, University of Duisburg-Essen, Essen, Germany
- Chair for Integrative Medicine and translationale Gastroenterology, Klinikum Bamberg, University of Duisburg-Essen, Essen, Bavaria, Germany
| | - Mariona Parera
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Inacio Mandomando
- Centro de Investigação em Saúde da Manhiça (CISM), 1929, Maputo, Mozambique
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Víctor Urrea
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Cristina Rodríguez
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Maria Casadellà
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
| | - Maria Luz Calle
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain
- Infectious Diseases Service, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain
- Institut Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autonoma de Barcelona, 08916, Badalona, Catalonia, Spain
| | - Denise Naniche
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigação em Saúde da Manhiça (CISM), 1929, Maputo, Mozambique
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain.
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Catalonia, Spain.
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, 08500, Vic, Catalonia, Spain.
- Infectious Diseases Service, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, 08916, Badalona, Catalonia, Spain.
| |
Collapse
|
237
|
Distinct gut microbiota profile in antiretroviral therapy-treated perinatally HIV-infected patients associated with cardiac and inflammatory biomarkers. AIDS 2019; 33:1001-1011. [PMID: 30946154 DOI: 10.1097/qad.0000000000002131] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Persistent inflammation and higher risk to develop cardiovascular diseases still represent a major complication for HIV-infected patients despite effective antiretroviral therapy (ART). We investigated the correlation between the gut microbiota profile, markers of inflammation, vascular endothelial activation (VEA) and microbial translocation (MT) in perinatally HIV-infected patients (PHIV) under ART. DESIGN Cross-sectional study including 61 ART-treated PHIV (age range 3-30 years old) and 71 age-matched healthy controls. Blood and stool sample were collected at the same time and analyzed for gut microbiota composition and plasma biomarkers. METHODS Gut microbiota composition was determined by 16S rRNA targeted-metagenomics. Soluble markers of MT, inflammation and VEA were quantified by ELISA or Luminex assay. Markers of immune activation were analyzed by flow cytometry on CD4 and CD8T cells. RESULTS We identified two distinct gut microbiota profiles (groups A and B) among PHIV. No different clinical parameters (age, sex, ethnicity, clinical class), dietary and sexual habits were found between the groups. The group A showed a relative dominance of Akkermansia muciniphila, whereas gut microbiota of group B was characterized by a higher biodiversity. The analysis of soluble markers revealed a significantly higher level of soluble E-selectine (P = 0.0296), intercellular adhesion molecule-1 (P = 0.0028), vascular adhesion molecule-1 (P = 0.0230), IL-6 (P = 0.0247) and soluble CD14 (P = 0.0142) in group A compared with group B. CONCLUSION Distinctive gut microbiota profiles are differently associated with inflammation, microbial translocation and VEA. Future studies are needed to understand the role of A. muciniphila and risk to develop cardiovascular diseases in PHIV.
Collapse
|
238
|
Wang Z, Qi Q. Gut microbial metabolites associated with HIV infection. Future Virol 2019; 14:335-347. [PMID: 31263508 PMCID: PMC6595475 DOI: 10.2217/fvl-2019-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
HIV infection has been associated with alterations in gut microbiota and related microbial metabolite production. However, the mechanisms of how these functional microbial metabolites may affect HIV immunopathogenesis and comorbidities, such as cardiovascular disease and other metabolic diseases, remain largely unknown. Here we review the current understanding of gut microbiota and related metabolites in the context of HIV infection. We focus on several bacteria-produced metabolites, including tryptophan catabolites, short-chain fatty acids and trimethylamine-N-oxide (TMAO), and discuss their implications in HIV infection and comorbidities. We also prospect future studies using integrative multiomics approaches to better understand host-microbiota-metabolites interactions in HIV infection, and facilitate integrative medicine utilizing the microbiota in HIV infection.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qibin Qi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
239
|
Hensley-McBain T, Wu MC, Manuzak JA, Cheu RK, Gustin A, Driscoll CB, Zevin AS, Miller CJ, Coronado E, Smith E, Chang J, Gale M, Somsouk M, Burgener AD, Hunt PW, Hope TJ, Collier AC, Klatt NR. Increased mucosal neutrophil survival is associated with altered microbiota in HIV infection. PLoS Pathog 2019; 15:e1007672. [PMID: 30973942 PMCID: PMC6459500 DOI: 10.1371/journal.ppat.1007672] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/02/2019] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal (GI) mucosal dysfunction predicts and likely contributes to non-infectious comorbidities and mortality in HIV infection and persists despite antiretroviral therapy. However, the mechanisms underlying this dysfunction remain incompletely understood. Neutrophils are important for containment of pathogens but can also contribute to tissue damage due to their release of reactive oxygen species and other potentially harmful effector molecules. Here we used a flow cytometry approach to investigate increased neutrophil lifespan as a mechanism for GI neutrophil accumulation in chronic, treated HIV infection and a potential role for gastrointestinal dysbiosis. We report that increased neutrophil survival contributes to neutrophil accumulation in colorectal biopsy tissue, thus implicating neutrophil lifespan as a new therapeutic target for mucosal inflammation in HIV infection. Additionally, we characterized the intestinal microbiome of colorectal biopsies using 16S rRNA sequencing. We found that a reduced Lactobacillus: Prevotella ratio associated with neutrophil survival, suggesting that intestinal bacteria may contribute to GI neutrophil accumulation in treated HIV infection. Finally, we provide evidence that Lactobacillus species uniquely decrease neutrophil survival and neutrophil frequency in vitro, which could have important therapeutic implications for reducing neutrophil-driven inflammation in HIV and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Tiffany Hensley-McBain
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
| | - Michael C. Wu
- Biostatistics and Biomathematics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jennifer A. Manuzak
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Ryan K. Cheu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Andrew Gustin
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
| | - Connor B. Driscoll
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Alexander S. Zevin
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
| | - Charlene J. Miller
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Ernesto Coronado
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
| | - Elise Smith
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Jean Chang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Michael Gale
- Washington National Primate Research Center, Seattle, WA, United States of America
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Ma Somsouk
- Division of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Adam D. Burgener
- National HIV and Retrovirology Labs, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Departments of Obstetrics & Gynecology and Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Thomas J. Hope
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Ann C. Collier
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Nichole R. Klatt
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, Seattle, WA, United States of America
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| |
Collapse
|
240
|
Biomarkers of aging in HIV: inflammation and the microbiome. Eur Geriatr Med 2019; 10:175-182. [PMID: 34652744 DOI: 10.1007/s41999-018-0145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/24/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE HIV-infected subjects present increased levels of inflammatory cytokines and T cell activation in the peripheral blood despite suppressive combination antiretroviral therapy which renders them susceptible to premature aging. The purpose of the present work was to review existing evidence on the ways in which the anatomical and microbiological abnormalities of the gastrointestinal tract can represent a major cause of organ disease in HIV infection. METHODS We conducted a systematic review of the Pubmed database for articles published from 2014 to 2018. We included studies on inflammatory/activation biomarkers associated with cardiovascular and bone disease, neurocognitive impairment and serious non-AIDS events in HIV-infected subjects. We also included researches which linked peripheral inflammation/activation to the anatomical, immune and microbiological alterations of the gastrointestinal tract. RESULTS Recent literature data confirm the association between non-infectious comorbidities and inflammation in HIV infection which may be driven by gastrointestinal tract abnormalities, specifically microbial translocation and dysbiosis. Furthermore, there is mounting evidence on the possible role of metabolic functions of the microbiota in the pathogenesis of premature aging in the HIV-infected population. CONCLUSIONS Biomarkers need to be validated for their use in the management of HIV infection. Compounds which counteract microbial translocation, inflammation and dysbiosis have been investigated as alternative therapeutic strategies in viro-suppressed HIV-infected individuals, but appear to have limited efficacy, probably due to the multifactorial pathogenesis of non-infectious comorbidities in this setting.
Collapse
|
241
|
Li SX, Sen S, Schneider JM, Xiong KN, Nusbacher NM, Moreno-Huizar N, Shaffer M, Armstrong AJS, Severs E, Kuhn K, Neff CP, McCarter M, Campbell T, Lozupone CA, Palmer BE. Gut microbiota from high-risk men who have sex with men drive immune activation in gnotobiotic mice and in vitro HIV infection. PLoS Pathog 2019; 15:e1007611. [PMID: 30947289 PMCID: PMC6448819 DOI: 10.1371/journal.ppat.1007611] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/31/2019] [Indexed: 12/30/2022] Open
Abstract
Men who have sex with men (MSM) have differences in immune activation and gut microbiome composition compared with men who have sex with women (MSW), even in the absence of HIV infection. Gut microbiome differences associated with HIV itself when controlling for MSM, as assessed by 16S rRNA sequencing, are relatively subtle. Understanding whether gut microbiome composition impacts immune activation in HIV-negative and HIV-positive MSM has important implications since immune activation has been associated with HIV acquisition risk and disease progression. To investigate the effects of MSM and HIV-associated gut microbiota on immune activation, we transplanted feces from HIV-negative MSW, HIV-negative MSM, and HIV-positive untreated MSM to gnotobiotic mice. Following transplant, 16S rRNA gene sequencing determined that the microbiomes of MSM and MSW maintained distinct compositions in mice and that specific microbial differences between MSM and MSW were replicated. Immunologically, HIV-negative MSM donors had higher frequencies of blood CD38+ HLADR+ and CD103+ T cells and their fecal recipients had higher frequencies of gut CD69+ and CD103+ T cells, compared with HIV-negative MSW donors and recipients, respectively. Significant microbiome differences were not detected between HIV-negative and HIV-positive MSM in this small donor cohort, and immune differences between their recipients were trending but not statistically significant. A larger donor cohort may therefore be needed to detect immune-modulating microbes associated with HIV. To investigate whether our findings in mice could have implications for HIV replication, we infected primary human lamina propria cells stimulated with isolated fecal microbiota, and found that microbiota from MSM stimulated higher frequencies of HIV-infected cells than microbiota from MSW. Finally, we identified several microbes that correlated with immune readouts in both fecal recipients and donors, and with in vitro HIV infection, which suggests a role for gut microbiota in immune activation and potentially HIV acquisition in MSM.
Collapse
Affiliation(s)
- Sam X. Li
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Sharon Sen
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Jennifer M. Schneider
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Ka-Na Xiong
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Nichole M. Nusbacher
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Nancy Moreno-Huizar
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Michael Shaffer
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Abigail J. S. Armstrong
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Erin Severs
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Kristine Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Charles P. Neff
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Martin McCarter
- Division of Colorectal Surgery, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Thomas Campbell
- Division of Infectious Diseases, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Catherine A. Lozupone
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| | - Brent E. Palmer
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Colorado Anschutz, Aurora, Colorado, United States of America
| |
Collapse
|
242
|
Abstract
OBJECTIVE We employed a high-dimensional covariate adjustment method in microbiome analysis to better control for behavioural and clinical confounders, and in doing so examine the effects of HIV on the rectal microbiome. DESIGN Three hundred and eighty-three MSM were grouped into four HIV viremia categories: HIV negative (n = 200), HIV-positive undetectable (HIV RNA < 20 copies/ml; n = 66), HIV-positive suppressed (RNA 20-200 copies/ml; n = 72) and HIV-positive viremic (RNA > 200 copies/ml; n = 45). METHODS We performed 16S rRNA gene sequencing on rectal swab samples and used inverse probability of treatment-weighted marginal structural models to examine differences in microbial composition by HIV viremia category. RESULTS HIV viremia explained a significant amount of variability in microbial composition in both unadjusted and covariate-adjusted analyses (R = 0.011, P = 0.02). Alterations in bacterial taxa were more apparent with increasing viremia. Relative to the HIV-negative group, HIV-positive undetectable participants showed depletions in Brachyspira, Campylobacter and Parasutterella, while suppressed participants demonstrated depletions in Barnesiella, Brachyspira and Helicobacter. The microbial signature of viremic men was most distinct, showing enrichment in inflammatory genera Peptoniphilus, Porphyromonas and Prevotella and depletion of Bacteroides, Brachyspira and Faecalibacterium, among others. CONCLUSION Our study shows that, after accounting for the influence of multiple confounding factors, HIV is associated with dysbiosis in the gastrointestinal microbiome in a dose-dependent manner. This analytic approach may allow for better identification of true microbial associations by limiting the effects of confounding, and thus improve comparability across future studies.
Collapse
|
243
|
Castleman MJ, Dillon SM, Purba CM, Cogswell AC, Kibbie JJ, McCarter MD, Santiago ML, Barker E, Wilson CC. Commensal and Pathogenic Bacteria Indirectly Induce IL-22 but Not IFNγ Production From Human Colonic ILC3s via Multiple Mechanisms. Front Immunol 2019; 10:649. [PMID: 30984202 PMCID: PMC6450192 DOI: 10.3389/fimmu.2019.00649] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a diverse family of cells that play critical roles in mucosal immunity. One subset of the ILC family, Group 3 ILCs (ILC3s), has been shown to aid in gut homeostasis through the production of IL-22. IL-22 promotes gut homeostasis through its functional effect on the epithelial barrier. When gut epithelial barrier integrity is compromised, such as in Human Immunodeficiency Virus (HIV) infection and inflammatory bowel disease (IBD), microbes from the gut lumen translocate into the lamina propria, inducing a multitude of potentially pathogenic immune responses. In murine models of bacterial infection, there is evidence that bacteria can induce pro-inflammatory IFNγ production in ILC3s. However, the impact of diverse translocating bacteria, particularly commensal bacteria, in dictating IFNγ versus IL-22 production by human gut ILC3s remains unclear. Here, we utilized an in vitro human lamina propria mononuclear cell (LPMC) model to evaluate ILC3 cytokine production in response to a panel of enteric Gram-positive and Gram-negative commensal and pathogenic bacteria and determined potential mechanisms by which these cytokine responses were induced. The percentages of IL-22-producing ILC3s, but not IFNγ-producing ILC3s, were significantly increased after LPMC exposure to both Gram-positive and Gram-negative commensal or pathogenic bacterial stimuli. Stimulation of IL-22 production from ILC3s was not through direct recognition of bacterial antigen by ILC3s, but rather required the help of accessory cells within the LPMC population. CD11c+ myeloid dendritic cells generated IL-23 and IL-1β in response to enteric bacteria and contributed to ILC3 production of IL-22. Furthermore, ligation of the natural cytotoxicity receptor NKp44 on ILC3s in response to bacteria stimulation also significantly increased the percentage of IL-22-producing ILC3s. Overall, these data demonstrate that human gut microbiota, including commensal bacteria, indirectly modulate colonic ILC3 function to induce IL-22, but additional signals are likely required to induce IFNγ production by colonic ILC3s in the setting of inflammation and microbial translocation.
Collapse
Affiliation(s)
- Moriah J. Castleman
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Stephanie M. Dillon
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Christine M. Purba
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Andrew C. Cogswell
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Jon J. Kibbie
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Martin D. McCarter
- Department of Surgery, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Mario L. Santiago
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, United States
| | - Cara C. Wilson
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical, Aurora, CO, United States
| |
Collapse
|
244
|
Villanueva-Millán MJ, Pérez-Matute P, Recio-Fernández E, Lezana Rosales JM, Oteo JA. Characterization of gut microbiota composition in HIV-infected patients with metabolic syndrome. J Physiol Biochem 2019; 75:299-309. [PMID: 30924020 DOI: 10.1007/s13105-019-00673-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/06/2019] [Indexed: 12/26/2022]
Abstract
The presence of metabolic syndrome (MS) per se or its separated components in HIV-infected patients contributes to an accelerated aging and increased cardiovascular risk. Gut microbiota (GM) dysbiosis has been linked with chronic inflammation associated with MS in a general non-infected population. However, no studies concerning GM have been performed in HIV-infected patients with MS. The aim of this study was to analyze bacterial translocation, inflammation, and GM composition in HIV-infected patients with and without MS. A total of 51 HIV-infected patients were recruited and classified according to the presence of MS (40 patients without MS and 11 with MS). Markers of bacterial translocation, inflammation, and cardiovascular risk were measured and GM was analyzed using 16S rRNA gene deep sequencing. No differences were observed among both HIV-infected groups in the bacterial translocation markers LBP and sCD14. A tendency to increase the inflammatory markers IL-6 (p = 0.069) and MCP-1 (p = 0.067) was observed in those patients suffering from MS. An increase in the cardiovascular risk markers PAI-1 (p = 0.007) and triglycerides/HDL cholesterol ratio (p < 0.0001) was also found in the MS group. No significant changes were observed at phylum level although a decrease in the abundance of seven genera and seven bacterial species, including some anti-inflammatory bacteria, was observed in HIV-infected patients with MS. To summarize, the presence of MS was not accompanied by major changes in GM, although the reduction observed in some anti-inflammatory bacteria may be clinically useful to develop strategies to minimize inflammation and its future deleterious consequences in these HIV-infected patients.
Collapse
Affiliation(s)
- María Jesús Villanueva-Millán
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - Patricia Pérez-Matute
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain.
| | - Emma Recio-Fernández
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - José-Miguel Lezana Rosales
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain
| | - José-Antonio Oteo
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 3rd floor, 26006, Logroño, La Rioja, Spain.,Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, La Rioja, Spain
| |
Collapse
|
245
|
Shenoy MK, Fadrosh DW, Lin DL, Worodria W, Byanyima P, Musisi E, Kaswabuli S, Zawedde J, Sanyu I, Chang E, Fong S, McCauley K, Davis JL, Huang L, Lynch SV. Gut microbiota in HIV-pneumonia patients is related to peripheral CD4 counts, lung microbiota, and in vitro macrophage dysfunction. MICROBIOME 2019; 7:37. [PMID: 30857553 PMCID: PMC6413461 DOI: 10.1186/s40168-019-0651-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 02/22/2019] [Indexed: 05/23/2023]
Abstract
Pneumonia is common and frequently fatal in HIV-infected patients, due to rampant, systemic inflammation and failure to control microbial infection. While airway microbiota composition is related to local inflammatory response, gut microbiota has been shown to correlate with the degree of peripheral immune activation (IL6 and IP10 expression) in HIV-infected patients. We thus hypothesized that both airway and gut microbiota are perturbed in HIV-infected pneumonia patients, that the gut microbiota is related to peripheral CD4+ cell counts, and that its associated products differentially program immune cell populations necessary for controlling microbial infection in CD4-high and CD4-low patients. To assess these relationships, paired bronchoalveolar lavage and stool microbiota (bacterial and fungal) from a large cohort of Ugandan, HIV-infected patients with pneumonia were examined, and in vitro tests of the effect of gut microbiome products on macrophage effector phenotypes performed. While lower airway microbiota stratified into three compositionally distinct microbiota as previously described, these were not related to peripheral CD4 cell count. In contrast, variation in gut microbiota composition significantly related to CD4 cell count, lung microbiota composition, and patient mortality. Compared with patients with high CD4+ cell counts, those with low counts possessed more compositionally similar airway and gut microbiota, evidence of microbial translocation, and their associated gut microbiome products reduced macrophage activation and IL-10 expression and increased IL-1β expression in vitro. These findings suggest that the gut microbiome is related to CD4 status and plays a key role in modulating macrophage function, critical to microbial control in HIV-infected patients with pneumonia.
Collapse
Affiliation(s)
- Meera K Shenoy
- Division of Gastroenterology, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Biomedical Sciences Graduate Program, UCSF, San Francisco, CA, USA
- Current address: Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Douglas W Fadrosh
- Division of Gastroenterology, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Din L Lin
- Division of Gastroenterology, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - William Worodria
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Patrick Byanyima
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Emmanuel Musisi
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Sylvia Kaswabuli
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Josephine Zawedde
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Ingvar Sanyu
- Infectious Diseases Research Collaboration, Mulago Hospital, Makerere University, Kampala, Uganda
| | - Emily Chang
- HIV, Infectious Diseases and Global Medicine Division, Department of Medicine, San Francisco General Hospital, UCSF, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, San Francisco General Hospital, UCSF, San Francisco, CA, USA
| | - Serena Fong
- HIV, Infectious Diseases and Global Medicine Division, Department of Medicine, San Francisco General Hospital, UCSF, San Francisco, CA, USA
| | - Kathryn McCauley
- Division of Gastroenterology, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - J Lucian Davis
- Department of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Laurence Huang
- HIV, Infectious Diseases and Global Medicine Division, Department of Medicine, San Francisco General Hospital, UCSF, San Francisco, CA, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, San Francisco General Hospital, UCSF, San Francisco, CA, USA.
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
246
|
Kang Y, Cai Y. Altered Gut Microbiota in HIV Infection: Future Perspective of Fecal Microbiota Transplantation Therapy. AIDS Res Hum Retroviruses 2019; 35:229-235. [PMID: 29877092 DOI: 10.1089/aid.2017.0268] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
HIV infection progressively destroys CD4+ mononuclear cells, leading to profound cellular immune deficiency that manifests as life-threatening opportunistic infections and malignancies (i.e., AIDS). Gut microbiota plays key roles in the modulation of host metabolism and gene expression, maintenance of epithelial integrity, and mediation of inflammatory and immunity. Hence, the normal intestinal microbiota plays a major role in the maintenance of health and disease prevention. In fact, a large number of studies have shown that the alteration of the gut microbiota contributes to the pathogenesis of several diseases, such as inflammatory bowel diseases, irritable bowel syndrome, metabolic diseases, anorexia nervosa, autoimmune diseases, multiple sclerosis, cancer, neuropsychiatric disorders, and cardiovascular diseases. Recently, accumulating evidence has shed light on the association of dysbiosis of gut microbiota with HIV infection. Hence, the modification of gut microbiota may be a potential therapeutic tool. Fecal microbiota transplantation may improve the conditions of patients with HIV infection by manipulating the human intestinal bacteria. However, the relevant research is very limited, and a large amount of scientific research work needs to be done in the near future.
Collapse
Affiliation(s)
- Yongbo Kang
- 1 School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- 2 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Yue Cai
- 1 School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- 3 Genetics and Pharmacogenomics Laboratory, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
247
|
Williams B, Weber K, Chlipala G, Evans C, Morack R, French A. HIV Status Does Not Affect Rectal Microbiome Composition, Diversity, or Stability over Time: A Chicago Women's Interagency HIV Study. AIDS Res Hum Retroviruses 2019; 35:260-266. [PMID: 30618262 DOI: 10.1089/aid.2018.0250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It remains unclear whether differences in gut microbiota noted between HIV-infected and uninfected individuals are driven by HIV or sexual behavior. We evaluated rectal swab microbiota of HIV-infected and uninfected women with similar demographic, neighborhood, and diet characteristics enrolled in the Chicago Women's Interagency HIV Study (WIHS). DNA was amplified for sequencing of fragments of bacterial small subunit (SSU or 16S) ribosomal RNA (rRNA) genes. HIV-infected and uninfected women did not differ by Shannon diversity index (p = .14), non-metric multidimensional scaling (NMDS) plot of Bray-Curtis indices (p = .488, r = 0.0027), or copy number of individual taxa. Both groups demonstrated marked microbiome stability over time (p = .889).
Collapse
Affiliation(s)
- Brett Williams
- Department of Internal Medicine, Division of Infectious Disease, Rush University Medical Center, Chicago, Illinois
- Ruth M. Rothstein Core Center of Cook County, Chicago, Illinois
| | - Kathleen Weber
- Cook County Health and Hospitals System, Chicago, Illinois
| | - George Chlipala
- Center for Research Informatics, Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | - Charlesnika Evans
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
- Center for Healthcare Studies, Northwestern University, Chicago, Illinois
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare, Hines, Illinois
| | - Ralph Morack
- Cook County Health and Hospitals System, Chicago, Illinois
| | - Audrey French
- Department of Internal Medicine, Division of Infectious Disease, Rush University Medical Center, Chicago, Illinois
- Ruth M. Rothstein Core Center of Cook County, Chicago, Illinois
- Department of Medicine, Stroger Hospital of Cook County, Chicago, Illinois
| |
Collapse
|
248
|
The Acute Influence of Acid Suppression with Esomeprazole on Gastrointestinal Microbiota and Brain Gene Expression Profiles in a Murine Model of Restraint Stress. Neuroscience 2019; 398:206-217. [DOI: 10.1016/j.neuroscience.2018.11.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022]
|
249
|
Zhang F, Yang J, Ji Y, Sun M, Shen J, Sun J, Wang J, Liu L, Shen Y, Zhang R, Chen J, Lu H. Gut Microbiota Dysbiosis Is Not Independently Associated With Neurocognitive Impairment in People Living With HIV. Front Microbiol 2019; 9:3352. [PMID: 30761121 PMCID: PMC6362426 DOI: 10.3389/fmicb.2018.03352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/31/2018] [Indexed: 12/24/2022] Open
Abstract
Gut microbiota dysbiosis, which has been linked to many neurological diseases, is common in HIV infection. However, its role in the pathogenesis of neurocognitive impairment is still not established. In this study, a total of 85 HIV infected subjects, naïve to antiretroviral therapy, were classified into two groups—those with HIV-associated neurological diseases (HAND) and those without, using the Montreal Cognitive Assessment (MoCA) test. Fecal samples were collected from all subjects and microbiota were analyzed by 16S rRNA amplicon sequencing. Subjects with HAND were older (P < 0.001), with lower levels of education (P = 0.002), lower CD4 T-cell counts (P = 0.032), and greater heterosexual preference (P < 0.001), than those without HAND. Gut microbiota from subjects with HAND showed significantly lower α-diversity compared to gut microbiota from subjects without HAND (Shannon index, P = 0.003). To exclude confounding bias, 25 subjects from each group, with comparable age, gender, CD4 T-cell count, educational level and sexual preference were further analyzed. The two groups showed comparable α-diversity (for SOB index, Shannon index, Simpson index, ACE index, and Chao index, all with P-value > 0.05) and β-diversity (ANOSIM statistic = 0.010, P = 0.231). There were no significant differences in microbiota composition between the two groups after the correction for a false discovery rate. Consistently, microbiota from the two groups presented similar predictive functional profiles. Gut microbiota dysbiosis is not independently associated with neurocognitive impairment in people living with HIV.
Collapse
Affiliation(s)
- Fengdi Zhang
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Junyang Yang
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yongjia Ji
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Meiyan Sun
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jiayin Shen
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianjun Sun
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jiangrong Wang
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Li Liu
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yinzhong Shen
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Renfang Zhang
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jun Chen
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Department of Infectious Disease, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.,Department of Infectious Disease, Huashan Hospital, Fudan University, Shanghai, China.,Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
250
|
Increased influenza-specific antibody avidity in HIV-infected women compared with HIV-infected men on antiretroviral therapy. AIDS 2019; 33:33-44. [PMID: 30234599 DOI: 10.1097/qad.0000000000002022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND It is recommended that HIV-infected individuals receive annual influenza vaccination due to their high susceptibility to influenza infection, especially among women. However, there have been few studies investigating sex-related responses to influenza vaccine in antiretroviral therapy (ART)-treated HIV-infected individuals. METHOD In this study, 26 aviremic ART-treated HIV-infected individuals and 16 healthy controls were enrolled in the current study. Blood was collected prior to vaccination (D0), on days 7-10 (D7) and on days 14-21 (D14) following administration of the 2013-2014 seasonal influenza vaccine. A series of analyses evaluated the serological and cellular responses following influenza vaccination. RESULTS Female HIV-infected individuals had increased influenza-specific antibody avidity relative to male HIV-infected individuals, but similar plasma levels of influenza-specific binding antibodies and neutralizing antibodies. Increased cycling B cells and follicular helper CD4 T (Tfh) cells were observed in female HIV-infected individuals pre and postvaccination compared with male HIV-infected individuals, and cycling Tfh cells were directly correlated with influenza-specific antibody avidity. Moreover, plasma testosterone levels were inversely correlated with antibody avidity index. The magnitude of microbial translocation [plasma lipopolysaccharide (LPS)] level was directly correlated with influenza-specific antibody avidity. Circulating 16S rDNA microbiome showed that enrichment of specific species within Proteobacteria was associated with influenza-specific antibody avidity. These results, including differences based on sex and correlations, were only observed in HIV-infected individuals but not in the healthy controls. CONCLUSION This study demonstrated sex differences in influenza-specific antibody avidity in ART-treated HIV disease, and provides valuable information on vaccination strategy in the ART-treated HIV-infected population.
Collapse
|