1
|
Tobin NH, Li F, Zhu W, Ferbas KG, Sleasman JW, Raftery D, Kuhn L, Aldrovandi GM. Altered milk tryptophan and tryptophan metabolites and health of children born to women with HIV. RESEARCH SQUARE 2025:rs.3.rs-6229815. [PMID: 40166030 PMCID: PMC11957222 DOI: 10.21203/rs.3.rs-6229815/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Children born to women with HIV (WWH) suffer increased morbidity and, in low-income settings, have two to three times the mortality of infants born to women without HIV. The basis for this increase remains elusive. In low-income settings, breastfeeding is recommended because health benefits outweigh the risk of transmission, especially when maternal antiretroviral therapy is provided. We profiled the milk metabolome of 326 women sampled longitudinally for 18 months postpartum using global metabolomics. We identify perturbations in several metabolites, including tryptophan, dimethylarginine, and a recently discovered antiviral ribonucleotide, that are robustly associated with maternal HIV infection. Quantitative tryptophan and kynurenine levels in both milk and plasma reveal that these perturbations reflect systemic depletion of tryptophan and alterations in tryptophan catabolism in WWH. Our findings provide intriguing evidence that decreases in tryptophan availability and perturbations in tryptophan catabolism in children born to WWH may contribute to their increased morbidity and mortality.
Collapse
Affiliation(s)
- Nicole H Tobin
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | - Fan Li
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | | | - Kathie G Ferbas
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| | - John W Sleasman
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | | | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons; and Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Grace M Aldrovandi
- Division of InfecGous Diseases, Department of Pediatrics, David Geffen School of Medicine at the University of California, Los Angeles
| |
Collapse
|
2
|
Nguyen HD, Kim WK. Disrupted microbial cross-feeding and altered L-phenylalanine consumption in people living with HIV. Brief Bioinform 2025; 26:bbaf111. [PMID: 40072847 PMCID: PMC11899578 DOI: 10.1093/bib/bbaf111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/18/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
This work aims to (1) identify microbial and metabolic alterations and (2) reveal a shift in phenylalanine production-consumption equilibrium in individuals with HIV. We conducted extensive searches in multiple databases [MEDLINE, Web of Science (including Cell Press, Oxford, HighWire, Science Direct, IOS Press, Springer Nature, PNAS, and Wiley), Google Scholar, and Embase] and selected two case-control 16S data sets (GenBank IDs: SRP039076 and EBI ID: ERP003611) for analysis. We assessed alpha and beta diversity, performed univariate tests on genus-level relative abundances, and identified significant microbiome features using random forest. We also utilized the MICOM model to simulate growth and metabolic exchanges within the microbiome, focusing on the Metabolite Exchange Score (MES) to determine key metabolic interactions. We found that L-phenylalanine had a higher MES in HIV-uninfected individuals compared with their infected counterparts. The flux of L-phenylalanine consumption was significantly lower in HIV-infected individuals compared with healthy controls, correlating with a decreased number of consuming species in the chronic HIV stage. Prevotella, Roseburia, and Catenibacterium were demonstrated as the most important microbial species involving an increase in L-phenylalanine production in HIV patients, whereas Bacteroides, Faecalibacterium, and Blautia contributed to a decrease in L-phenylalanine consumption. We also found significant alterations in both microbial diversity and metabolic exchanges in people living with HIV. Our findings shed light on why HIV-1 patients have elevated levels of phenylalanine. The impact on essential amino acids like L-phenylalanine underscores the effect of HIV on gut microbiome dynamics. Targeting the restoration of these interactions presents a potential therapeutic avenue for managing HIV-related dysbiosis.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433, United States
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70118, United States
| |
Collapse
|
3
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 DOI: 10.1186/s13054-025-05309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
4
|
Reimer-McAtee M, Serpa J, McAtee C, Ortega E, Somasunderam A, Arduino R, Mejia R, Utay NS. Impact of intestinal parasitic infections on gut epithelial barrier and inflammation among foreign-born persons living with HIV. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.03.25321616. [PMID: 39974003 PMCID: PMC11838668 DOI: 10.1101/2025.02.03.25321616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Systemic inflammation is a major driver of comorbidities in people with HIV (PWH). Increased levels of biomarkers of enterocyte turnover, microbial translocation, and systemic inflammation have been shown to predict morbidity and mortality in PWH on antiretroviral therapy (ART). We conducted a prospective cohort study of foreign-born PWH with undetectable HIV RNA (<20 copies/mL) with and without intestinal parasitic coinfection. Biomarkers of enterocyte turnover (intestinal fatty acid binding protein [I-FABP]), microbial translocation (soluble CD14), and systemic inflammation (soluble CD163) were measured. Stool parasite quantitative PCR (qPCR) testing and Strongyloides stercoralis recombinant IgG ELISA ( Strongy IgG) were utilized to diagnose parasitic infection. Of the 52 participants, 14 (27%) tested positive for infection with Strongyloides stercoralis by Strongy IgG, and 7 (16%) of the 45 participants who provided stool samples tested positive for a parasitic infection (not including Blastocystis ) by stool qPCR. The median sCD14 level in PWH with (+) Strongy IgG was significantly higher than PWH with (-) Strongy IgG (1.69 ug/ml versus 1.48 ug/ml, p=0.03). The median sCD163 in PWH with parasitic infections by qPCR was not significantly different from sCD163 in PWH negative for parasitic infections. I-FABP levels did not differ significantly between groups. Participants with both HIV and intestinal parasite infections had increased levels of sCD14, a marker of microbial translocation that is an independent predictor of mortality in PWH, compared to PWH without parasitic infections. These findings raise concern about the long-term sequelae of intestinal parasitic infections in PWH.
Collapse
|
5
|
Ruderman SA, Hunt PW, Beck-Engeser G, Ambayec G, Willig AL, Saag MS, Napravnik S, Cachay E, Bamford L, Landay A, Drumright LN, Mixson LS, Whitney BM, Nance RM, Kitahata MM, Crane HM, Delaney JAC, Hahn AW. Biomarkers of microbial translocation and generalized inflammation are associated with frailty among people with HIV. AIDS 2025; 39:153-161. [PMID: 39453872 PMCID: PMC11717601 DOI: 10.1097/qad.0000000000004047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/13/2024] [Indexed: 10/27/2024]
Abstract
BACKGROUND Frailty occurs at higher rates and younger ages among people with HIV (PWH) compared with the general population and is often attributed to chronic inflammation and subsequent immune exhaustion. We assessed how inflammatory biomarkers are associated with frailty among PWH. METHODS The Centers for AIDS Research (CFAR) Network of Integrated Clinical Systems (CNICS) cohort is comprised of adult PWH in care at 10 sites, and harmonizes demographic, clinical, and patient-reported outcomes (PRO) data. A panel of 13 inflammatory biomarkers was collected from a subset of virally suppressed PWH once per person between 2010 and 2018. Frailty was measured with a validated PRO phenotype, scored 0-4, from biomarker collection date through July 2022. With adjusted linear mixed models, we estimated longitudinal associations between standard deviation-scaled log 2 -transformed biomarkers and frailty score. RESULTS Among 273 PWH, most were men (91%), average age at baseline was 45, 42% were non-Hispanic White whereas 35% were non-Hispanic Black, and average follow-up time was 5.5 years. Several biomarkers were associated with higher frailty, including those linked to microbial translocation (sCD14, LBP, KT ratio) and systemic inflammation (CRP, IL-6, suPAR, sTNFR1, sTNFR2). Higher IL-6 was associated with a 0.25-point higher frailty score [95% confidence interval (CI) 0.12-0.39]. Higher sTNFR1 [0.35 (0.13-0.56)], sCD14 [0.21 (0.11-0.31)], and suPAR [0.24 (0.11-0.36)] levels were also associated with higher frailty scores over follow-up. CONCLUSION Higher levels of biomarkers linked to microbial translocation and systemic inflammation are associated with higher average frailty scores over time in a cohort of virally suppressed PWH, highlighting these pathways as potential interventional targets for mitigating frailty in PWH.
Collapse
Affiliation(s)
| | - Peter W Hunt
- University of California San Francisco, San Francisco, CA
| | | | | | | | | | | | | | | | - Alan Landay
- University of Texas Medical Branch Galveston
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
El-Badry E, Chen L, Ghneim K, Li Z, Brooks K, Rhodes J, Sekaly R, Kilembe W, Allen S, Wu H, Hunter E. Heightened expression of type I interferon signaling genes in CD4 + T cells from acutely HIV-1-infected women is associated with lower viral loads. Front Immunol 2025; 15:1507530. [PMID: 39902043 PMCID: PMC11788160 DOI: 10.3389/fimmu.2024.1507530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Sex differences play a role in the pathogenesis of a number of viral diseases. In HIV-1, several studies have reported that chronically infected women have significantly lower plasma viremia than men, although the exact mechanism by which this occurs has yet to be identified. We have performed bulk RNA-seq experiments comparing gene expression between CD4+ T cells from acutely HIV-1-infected men and women in Zambia, because we observe lower viral load (VL) despite higher CD4+ T-cell activation in these women during acute/early infection. In a univariate analysis, we have identified a number of differentially expressed genes in naïve, central memory, and effector memory CD4 T cells of women with consistent elevated expression of genes linked to type 1 interferon (IFN) signaling. Moreover, after controlling for differences in VL and CD4+ T-cell count, genes within the type I IFN signaling pathway were further shown to be more highly expressed in women, whereas those genes more highly expressed in men showed no such enrichment. A subset of the genes highly expressed in women was further identified, including several involved in type I IFN signaling in response to viral infections (IRF7, DDX58, SAMHD1, OAS2, and TRIM14), that both are more highly expressed in CD4+ T cells from women and negatively correlated with VL, suggesting that they play a role in the comparative control of VL observed in women.
Collapse
Affiliation(s)
- Elina El-Badry
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Luxiao Chen
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Khader Ghneim
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| | - Ziyi Li
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Kelsie Brooks
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Jake Rhodes
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Rafick Sekaly
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| | | | - Susan Allen
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
- Center for Family Health in Zambia, Lusaka, Zambia
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Eric Hunter
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
7
|
Vanpouille C, Wells A, DeGruttola V, Lynch M, Zhang X, Fitzgerald W, Tu X, Chaillon A, Landay A, Weber K, Scully E, Karn J, Gianella S. Cytokine trajectory over time in men and women with HIV on long-term antiretroviral therapy. AIDS 2025; 39:1-10. [PMID: 39639719 PMCID: PMC11631044 DOI: 10.1097/qad.0000000000004033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Although antiretroviral therapy (ART) suppresses viral replication and reduces inflammation, it does not lead to the normalization of cytokines. The long-term effects of ART beyond viral suppression have not been studied and are mostly limited to cross-sectional research. DESIGN The impact of long-term ART on the trajectory of 40 cytokines/chemokines in 31 men and 59 women who maintained viral suppression over a median period of 6 years (317 visits ranging from 24 to 384 weeks post ART initiation) were measured by Luminex. METHODS We used a generalized additive model with a Gaussian distribution and identity link function to model concentrations over time and investigate sex and race differences. RESULTS While most cytokine/chemokine trajectories remained stable, the trajectory of nine markers of monocyte/macrophage activation (IP-10, I-TAC, MIG, sCD163, sCD14, MCP-1, MIP-3β, CXCL13, TNF-α) decreased over time (adj. P < 0.05). Despite continuous viral suppression, M-CSF, IL-15, and LBP increased over time (adj. P < 0.05). sCD14 was the only cytokine whose trajectory differed by sex (adj. P = 0.033). Overall, women had lower mean levels of IL-18 but higher levels of sCD14 than did men (adj. P < 0.05). GROα, LBP, and sCD14 showed significant differences between races (adj. P < 0.05). No association between cytokines and cellular HIV DNA/RNA was found. CONCLUSION Our study reveals a continuous decline in markers of monocyte/macrophage activation over 6 years of suppressive ART, indicating that long-term treatment may mitigate inflammaging and cardiovascular-related outcomes. The higher levels of sCD14 observed in women are consistent with them having greater innate immune activation than men do.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Alan Wells
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Victor DeGruttola
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Miranda Lynch
- Department of Biostatistics and Bioinformatics, Hauptman-Woodward Medical Research Institute, Buffalo, NY, USA
| | - Xinlian Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Wendy Fitzgerald
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Xin Tu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Antoine Chaillon
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alan Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kathleen Weber
- Hektoen Institute of Medicine/Cook County Health, Chicago, IL, USA
| | - Eileen Scully
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Sara Gianella
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
8
|
Sheets K, Baker JV. HIV and Inflamm-Aging: How Do We Reach the Summit of Healthy Aging? TOPICS IN ANTIVIRAL MEDICINE 2024; 32:589-596. [PMID: 39765238 PMCID: PMC11737810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
People with HIV (PWH) are living longer and experiencing a greater burden of morbidity from non-AIDS-defining conditions. Chronically treated HIV disease is associated with ongoing systemic inflammation that contributes to the development of chronic conditions (eg, cardiovascular disease) and geriatric syndromes (eg, frailty). Apart from HIV disease, a progressive increase in systemic inflammation is a characteristic feature of biologic aging, a process described as "inflammaging." Inflamm-aging is driven by persistent antigen stimulation and stress, leading to an immune profile characterized by elevated levels of blood inflammatory markers and cellular activation and senescence. Chronic HIV disease is hypothesized to accentuate the immune profile of inflamm-aging, in part through viral persistence in lymphatic tissues, permanent injury impairing immune recovery, the presence of copathogens, gut dysbiosis and microbial translocation, and chromosomal and genetic alterations associated with immune activation. Few strategies exist for safe and effective modulation of systemic inflammation among older PWH. The strongest current evidence supports aggressive management of modifiable risk factors such as lipids, blood pressure, and levels of physical activity. Future inflamm-aging research should be directed toward advancing the implementation of proven approaches, such as physical activity, as well as studying novel mechanisms of, and treatments for, inflamm-aging among PWH.
Collapse
Affiliation(s)
- Kerry Sheets
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| | - Jason V. Baker
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| |
Collapse
|
9
|
Islam SMS, Singh S, Keshavarzian A, Abdel-Mohsen M. Intestinal Microbiota and Aging in People with HIV-What We Know and What We Don't. Curr HIV/AIDS Rep 2024; 22:9. [PMID: 39666149 PMCID: PMC11874070 DOI: 10.1007/s11904-024-00717-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
PURPOSE OF REVIEW People with HIV (PWH) experience premature aging and an elevated risk of age-related comorbidities, even with viral suppression through antiretroviral therapy (ART). We examine gastrointestinal disruptions, specifically impaired intestinal barrier integrity and microbial dysbiosis, as contributors to these comorbidities. RECENT FINDINGS HIV infection compromises the intestinal epithelial barrier, increasing permeability and microbial translocation, which trigger inflammation and cellular stress. ART does not fully restore gut barrier integrity, leading to persistent inflammation and cellular stress. Additionally, HIV-associated microbial dysbiosis favors pro-inflammatory bacteria, intensifying inflammation and tissue damage, which may contribute to premature aging in PWH. Understanding the interactions between intestinal microbiota, chronic inflammation, cellular stress, and aging is essential to developing therapies aimed at reducing inflammation and slowing age-related diseases in PWH. In this review, we discuss critical knowledge gaps and highlight the therapeutic potential of microbiota-targeted interventions to mitigate inflammation and delay age-associated pathologies in PWH.
Collapse
Affiliation(s)
| | - Shalini Singh
- Northwestern University, 300 E Superior St, Chicago, IL, 60611, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, USA
- Departments of Internal Medicine, physiology Rush University Medical Center, Anatomy & Cell Biology, Chicago, IL, USA
| | | |
Collapse
|
10
|
Ryu A, Clagett BM, Freeman ML. Inflammation and Microbial Translocation Correlate with Reduced MAIT Cells in People with HIV. Pathog Immun 2024; 10:19-46. [PMID: 39635460 PMCID: PMC11613984 DOI: 10.20411/pai.v10i1.746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/05/2024] [Indexed: 12/07/2024] Open
Abstract
Background Optimal control of microbial infections requires mucosal-associated invariant T (MAIT) cells. People living with HIV (PWH) on antiretroviral therapy (ART) can be divided into 2 groups: immune responders (IR) who recover or retain CD4 T cell numbers, and immune non-responders (INR) who do not. Compared to IR, INR have fewer MAIT cells and increased systemic inflammation and microbial translocation, but how these factors affect MAIT cells is unknown. Methods MAIT cells from IR, INR, and from controls without HIV were enumerated and characterized by flow cytometry. To determine the links among MAIT cells, inflammation, and microbial translocation, the correlations of MAIT cell numbers to previously published soluble inflammatory markers and plasma microbial genetic sequences were assessed by Spearman analysis. In vitro assays were used to support our findings. Results MAIT cell numbers were significantly negatively correlated with levels of IL-6 and IP-10 (markers of inflammation); CD14, LPS, and FABP2 (markers of microbial translocation); and with abundance of Serratia and other Proteobacteria genetic sequences in plasma. In a separate analysis of PWH on ART receiving the IL-6 receptor antagonist tocilizumab (TCZ), we found that blocking IL-6 signaling with TCZ increased IL-7 receptor expression on MAIT cells and reduced plasma IL-7 levels, consistent with improved uptake of IL-7 in vivo. Conclusions Our findings suggest inflammation and microbial translocation in PWH on ART lead to a loss of MAIT cells via impaired IL-7 responsiveness, resulting in further increased microbial translocation and inflammation.
Collapse
Affiliation(s)
- Angela Ryu
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Brian M. Clagett
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Michael L. Freeman
- Rustbelt Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
11
|
Basting CM, Langat R, Broedlow CA, Guerrero CR, Bold TD, Bailey M, Velez A, Schroeder T, Short-Miller J, Cromarty R, Mayer ZJ, Southern PJ, Schacker TW, Safo SE, Bramante CT, Tignanelli CJ, Schifanella L, Klatt NR. SARS-CoV-2 infection is associated with intestinal permeability, systemic inflammation, and microbial dysbiosis in hospitalized patients. Microbiol Spectr 2024; 12:e0068024. [PMID: 39345212 PMCID: PMC11537016 DOI: 10.1128/spectrum.00680-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) and its associated severity have been linked to uncontrolled inflammation and may be associated with changes in the microbiome of mucosal sites including the gastrointestinal tract and oral cavity. These sites play an important role in host-microbe homeostasis, and disruption of epithelial barrier integrity during COVID-19 may potentially lead to exacerbated inflammation and immune dysfunction. Outcomes in COVID-19 are highly disparate, ranging from asymptomatic to fatal, and the impact of microbial dysbiosis on disease severity is unclear. Here, we obtained plasma, rectal swabs, oropharyngeal swabs, and nasal swabs from 86 patients hospitalized with COVID-19 and 12 healthy volunteers. We performed 16S rRNA sequencing to characterize the microbial communities in the mucosal swabs and measured concentrations of circulating cytokines, markers of gut barrier integrity, and fatty acids in the plasma samples. We compared these plasma concentrations and microbiomes between healthy volunteers and COVID-19 patients, some of whom had unfortunately died by the end of the study enrollment, and performed a correlation analysis between plasma variables and bacterial abundances. Rectal swabs of COVID-19 patients had reduced abundances of several commensal bacteria including Faecalibacterium prausnitzii and an increased abundance of the opportunistic pathogens Eggerthella lenta and Hungatella hathewayi. Furthermore, the oral pathogen Scardovia wiggsiae was more abundant in the oropharyngeal swabs of COVID-19 patients who died. The abundance of both H. hathewayi and S. wiggsiae correlated with circulating inflammatory markers including IL-6, highlighting the possible role of the microbiome in COVID-19 severity and providing potential therapeutic targets for managing COVID-19.IMPORTANCEOutcomes in coronavirus disease 2019 (COVID-19) are highly disparate and are associated with uncontrolled inflammation; however, the individual factors that lead to this uncontrolled inflammation are not fully understood. Here, we report that severe COVID-19 is associated with systemic inflammation, microbial translocation, and microbial dysbiosis. The rectal and oropharyngeal microbiomes of COVID-19 patients were characterized by a decreased abundance of commensal bacteria and an increased abundance of opportunistic pathogens, which positively correlated with markers of inflammation and microbial translocation. These microbial perturbations may, therefore, contribute to disease severity in COVID-19 and highlight the potential for microbiome-based interventions in improving COVID-19 outcomes.
Collapse
Affiliation(s)
| | - Robert Langat
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Candace R. Guerrero
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Tyler D. Bold
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Melisa Bailey
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adrian Velez
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ty Schroeder
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonah Short-Miller
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minnesota, Minneapolis, USA
| | - Zachary J. Mayer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Peter J. Southern
- Department of Microbiology and Immunology, University of Minnesota, Minnesota, Minneapolis, USA
| | - Timothy W. Schacker
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Sandra E. Safo
- Department of Biostatistics and Health Data Science, University of Minnesota, Minnesota, Minneapolis, USA
| | - Carolyn T. Bramante
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | | | - Luca Schifanella
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- National Cancer Institute, Center for Cancer Research, Vaccine Branch, Animal Models and Retroviral Vaccines Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Nichole R. Klatt
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
12
|
Tarnathummanan C, Soimanee T, Khattiya J, Sretapunya W, Phaonakrop N, Roytrakul S, Akekawatchai C. Plasma proteomic profiles of patients with HIV infection and coinfection with hepatitis B/C virus undergoing anti‑retroviral therapy. Biomed Rep 2024; 21:155. [PMID: 39268407 PMCID: PMC11391517 DOI: 10.3892/br.2024.1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/26/2024] [Indexed: 09/15/2024] Open
Abstract
Chronic liver disease is becoming a leading cause of illness and mortality in patients living with human immunodeficiency virus (HIV; PLWH) undergoing suppressive anti-retroviral therapy. Its primary etiology is coinfection with hepatitis B and C virus (HBV and HCV, respectively). Chronic liver inflammation and fibrosis can potentially lead to the development of hepatocellular carcinoma (HCC). Therefore, monitoring of the disease progression in PLWH is required. The present study aimed to explore plasma protein profiles of PLWH and those coinfected with HBV and HCV using shotgun proteomics. HIV-monoinfected, HIV/HBV-coinfected, HIV/HCV-coinfected and uninfected control individuals were recruited. Patients in the three virus-infected groups had significantly higher levels of liver fibrosis indices (fibrosis-4 score and aspartate aminotransferase to platelet ratio index) compared with the control group. Liquid chromatography-tandem mass spectrometry analysis of plasma samples identified 1,074 proteins that were differentially expressed, where subsequent partial least squares-discriminant analysis model demonstrated clear clustering of proteomes from the four sample groups; 18 proteins that were significantly differentially expressed. Heatmap analysis identified two main groups of proteins, six proteins being upregulated only in the HIV/HBV-coinfection group and 10 proteins downregulated in all three virally infected groups. STITCH 5.0 analysis predicted an interaction network containing two identified proteins in the latter group, specifically ubiquitin interaction motif-containing 1 (UIMC1) and haptoglobin (HP), which are part of the profibrogenic TGF-1β/SMAD, inflammatory TNF and tumor suppressor BRCA1 pathways. Expression levels of UIMC1 and HP were significantly lower in HIV-infected groups compared with those in uninfected controls. Altogether, these proteomics data provide protein expression profiles potentially associated with HIV infection and coinfection with HBV/HCV, which may be applied to predict progression to advanced liver disease or HCC in PLWH.
Collapse
Affiliation(s)
- Chewaporn Tarnathummanan
- Graduate Program in Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani 12121, Thailand
| | - Thanawan Soimanee
- Thammasat University Research Unit in Diagnostic Molecular Biology of Chronic Diseases Related to Cancer, Pathumthani 12121, Thailand
| | - Janya Khattiya
- Thammasat University Research Unit in Diagnostic Molecular Biology of Chronic Diseases Related to Cancer, Pathumthani 12121, Thailand
| | - Warisara Sretapunya
- Department of Medical Technology and Pathology, Nakorn Nayok Hospital, Nakorn Nayok 26000, Thailand
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Chareeporn Akekawatchai
- Thammasat University Research Unit in Diagnostic Molecular Biology of Chronic Diseases Related to Cancer, Pathumthani 12121, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Klongluang, Pathumthani 12121, Thailand
| |
Collapse
|
13
|
Díaz-García C, Moreno E, Talavera-Rodríguez A, Martín-Fernández L, González-Bodí S, Martín-Pedraza L, Pérez-Molina JA, Dronda F, Gosalbes MJ, Luna L, Vivancos MJ, Huerta-Cepas J, Moreno S, Serrano-Villar S. Fecal microbiota transplantation alters the proteomic landscape of inflammation in HIV: identifying bacterial drivers. MICROBIOME 2024; 12:214. [PMID: 39438902 PMCID: PMC11494993 DOI: 10.1186/s40168-024-01919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/26/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Despite effective antiretroviral therapy, people with HIV (PWH) experience persistent systemic inflammation and increased morbidity and mortality. Modulating the gut microbiome through fecal microbiota transplantation (FMT) represents a novel therapeutic strategy. We aimed to evaluate proteomic changes in inflammatory pathways following repeated, low-dose FMT versus placebo. METHODS This double-masked, placebo-controlled pilot study assessed the proteomic impacts of weekly FMT versus placebo treatment over 8 weeks on systemic inflammation in 29 PWH receiving stable antiretroviral therapy (ART). Three stool donors with high Faecalibacterium and butyrate profiles were selected, and their individual stools were used for FMT capsule preparation. Proteomic changes in 345 inflammatory proteins in plasma were quantified using the proximity extension assay, with samples collected at baseline and at weeks 1, 8, and 24. Concurrently, we characterized shifts in the gut microbiota composition and annotated functions through shotgun metagenomics. We fitted generalized additive models to evaluate the dynamics of protein expression. We selected the most relevant proteins to explore their correlations with microbiome composition and functionality over time using linear mixed models. RESULTS FMT significantly reduced the plasma levels of 45 inflammatory proteins, including established mortality predictors such as IL6 and TNF-α. We found notable reductions persisting up to 16 weeks after the final FMT procedure, including in the expression of proteins such as CCL20 and CD22. We identified changes in 46 proteins, including decreases in FT3LG, IL6, IL10RB, IL12B, and IL17A, which correlated with multiple bacterial species. We found that specific bacterial species within the Ruminococcaceae, Succinivibrionaceae, Prevotellaceae families, and the Clostridium genus, in addition to their associated genes and functions, were significantly correlated with changes in inflammatory markers. CONCLUSIONS Targeting the gut microbiome through FMT effectively decreased inflammatory proteins in PWH, with sustained effects. These findings suggest the potential of the microbiome as a therapeutic target to mitigate inflammation-related complications in this population, encouraging further research and development of microbiome-based interventions. Video Abstract.
Collapse
Affiliation(s)
- Claudio Díaz-García
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Elena Moreno
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Alba Talavera-Rodríguez
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Lucía Martín-Fernández
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Sara González-Bodí
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Laura Martín-Pedraza
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - José A Pérez-Molina
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Fernando Dronda
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María José Gosalbes
- Área de Genómica y Salud, Fundación Para El Fomento de La Investigación Sanitaria y Biomédica de La Comunidad Valenciana-Salud Pública, Valencia, Spain
- CIBERESP, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Laura Luna
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María Jesús Vivancos
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Jaime Huerta-Cepas
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28223, Madrid, Spain
| | - Santiago Moreno
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS and Universidad de Alcalá, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
14
|
Ebrahimi R, Masouri MM, Salehi Amniyeh Khozani AA, Ramadhan Hussein D, Nejadghaderi SA. Safety and efficacy of fecal microbiota transplantation for viral diseases: A systematic review of clinical trials. PLoS One 2024; 19:e0311731. [PMID: 39432486 PMCID: PMC11493255 DOI: 10.1371/journal.pone.0311731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/21/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Gut microbiota play important roles in several diseases like viral infections. In this systematic review, our objective was to assess the efficacy and safety of fecal microbiota transplantation (FMT) in treating various viral diseases. METHODS We conducted searches on databases including PubMed, Web of Science, Scopus, and Google Scholar until November 2023. Clinical trials reported outcomes related to safety of FMT or its efficacy in patients with viral diseases were included. We excluded other types of studies that enrolled healthy individuals or patients with other disorders and did not use FMT. The assessment of bias risk was conducted using the National Institutes of Health (NIH) study quality evaluation tool. RESULTS Eight studies with total 196 participants were included. Viral diseases were human immunodeficiency virus (HIV), hepatitis B, COVID-19 and Clostridioides difficile coinfection, and cytomegalovirus colitis. In hepatitis B cases, HBeAg clearance was significant in those received FMT (p<0.01), while it was not significant in another one (p = 0.19). A clinical response was noted in 37.5% of patients with cytomegalovirus colitis, with an equal percentage achieving clinical remission post-FMT. There was a significant reduction in Clostridioides difficile relapse rate in FMT group than controls in coinfection of Clostridioides difficile and COVID-19 (2.17% vs. 42.5%, p<0.05). In patients with HIV, partial engraftment of the donor microbiome and increases in alpha diversity were observed after FMT. No severe adverse events were reported. Most studies had fair or good qualities. CONCLUSIONS Our findings revealed FMT as a promising, safe treatment for some viral diseases. It improved viral clearance, clinical outcomes, and inflammation. However, the varying responses and small sample sizes call for more trials on FMT in viral diseases.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | - Seyed Aria Nejadghaderi
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
15
|
Augustin M, Horn C, Ercanoglu MS, Bondet V, de Silva US, Suarez I, Chon SH, Nierhoff D, Zoufaly A, Wenisch C, Knops E, Heger E, Klein F, Duffy D, Müller-Trutwin M, Lehmann C. From Gut to Blood: Redistribution of Zonulin in People Living with HIV. Biomedicines 2024; 12:2316. [PMID: 39457626 PMCID: PMC11505231 DOI: 10.3390/biomedicines12102316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Gastrointestinal mucosal damage due to human immunodeficiency virus (HIV) infection leads to microbial translocation and immune activation, contributing to the development of non-infectious comorbidities (NICM) in people living with HIV (PLWH). Additionally, persistent proviral HIV-1 in the gut-associated lymphatic tissue (GALT) can trigger immunological changes in the epithelial environment, impacting the mucosal barrier. However, the role of zonulin, a modulator of epithelial tight junctions in GALT during HIV infection, remains poorly understood. METHODS We measured zonulin in serum and intestinal tissue sections from five treatment-naive (HIV+NAIVE) and 10 cART-treated (HIV+cART) HIV+ individuals, along with 11 controls (CTRL). We compared zonulin levels with clinical characteristics, inflammatory markers (IFN-α, CXCR3, and PD-1), and the viral reservoir in peripheral blood (PB) and terminal ileum (TI). RESULTS Upon HIV infection, TI was found to harbor more HIV DNA than PB. Circulating zonulin levels were highest in HIV+NAIVE compared to HIV+cART or CTRL. Surprisingly, in the gut tissue sections, zonulin levels were higher in CTRL than in HIV+ individuals. Elevated circulating zonulin levels were found to be correlated with CD4+T-cell depletion in PB and TI, and with intestinal IFN-α. CONCLUSIONS The findings of this study indicate a shift in zonulin levels from the gut to the bloodstream in response to HIV infection. Furthermore, elevated systemic zonulin levels are associated with the depletion of intestinal CD4+ T cells and increased gut inflammation, suggesting a potential link between systemic zonulin and intestinal damage. Gaining insight into the regulation of gut tight junctions during HIV infection could offer valuable understanding for preventing NICM in PLWH.
Collapse
Affiliation(s)
- Max Augustin
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (C.H.); (U.S.d.S.); (I.S.)
- Center for Molecular Medicine Cologne (CMMC), 50937 Cologne, Germany
- German Center for Infection Research (DZIF), 50937 Cologne, Germany
- Department IV of Internal Medicine, Klinik Favoriten, Vienna Healthcare Group, 1100 Vienna, Austria; (A.Z.); (C.W.)
- Faculty of Medicine, Sigmund Freud University, 1020 Vienna, Austria
| | - Carola Horn
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (C.H.); (U.S.d.S.); (I.S.)
- Center for Molecular Medicine Cologne (CMMC), 50937 Cologne, Germany
- German Center for Infection Research (DZIF), 50937 Cologne, Germany
| | - Meryem Seda Ercanoglu
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (M.S.E.); (E.K.); (E.H.); (F.K.)
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, 75015 Paris, France; (V.B.); (D.D.)
| | - Ute Sandaradura de Silva
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (C.H.); (U.S.d.S.); (I.S.)
- Center for Molecular Medicine Cologne (CMMC), 50937 Cologne, Germany
- German Center for Infection Research (DZIF), 50937 Cologne, Germany
| | - Isabelle Suarez
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (C.H.); (U.S.d.S.); (I.S.)
- German Center for Infection Research (DZIF), 50937 Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral Surgery and Surgical Oncology, University Hospital Cologne, 50937 Cologne, Germany;
| | - Dirk Nierhoff
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, 50937 Cologne, Germany;
| | - Alexander Zoufaly
- Department IV of Internal Medicine, Klinik Favoriten, Vienna Healthcare Group, 1100 Vienna, Austria; (A.Z.); (C.W.)
- Faculty of Medicine, Sigmund Freud University, 1020 Vienna, Austria
| | - Christoph Wenisch
- Department IV of Internal Medicine, Klinik Favoriten, Vienna Healthcare Group, 1100 Vienna, Austria; (A.Z.); (C.W.)
| | - Elena Knops
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (M.S.E.); (E.K.); (E.H.); (F.K.)
| | - Eva Heger
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (M.S.E.); (E.K.); (E.H.); (F.K.)
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany; (M.S.E.); (E.K.); (E.H.); (F.K.)
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, 75015 Paris, France; (V.B.); (D.D.)
| | - Michaela Müller-Trutwin
- HIV, Inflammation and Persistence Unit, Institut Pasteur, Université Paris-Cité, 75015 Paris, France;
| | - Clara Lehmann
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (C.H.); (U.S.d.S.); (I.S.)
- Center for Molecular Medicine Cologne (CMMC), 50937 Cologne, Germany
- German Center for Infection Research (DZIF), 50937 Cologne, Germany
| |
Collapse
|
16
|
Semenova L, Wang Y, Falcinelli S, Archin N, Cooper-Volkheimer AD, Margolis DM, Goonetilleke N, Murdoch DM, Rudin CD, Browne EP. Machine learning approaches identify immunologic signatures of total and intact HIV DNA during long-term antiretroviral therapy. eLife 2024; 13:RP94899. [PMID: 39250423 PMCID: PMC11383529 DOI: 10.7554/elife.94899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
Understanding the interplay between the HIV reservoir and the host immune system may yield insights into HIV persistence during antiretroviral therapy (ART) and inform strategies for a cure. Here, we applied machine learning (ML) approaches to cross-sectional high-parameter HIV reservoir and immunology data in order to characterize host-reservoir associations and generate new hypotheses about HIV reservoir biology. High-dimensional immunophenotyping, quantification of HIV-specific T cell responses, and measurement of genetically intact and total HIV proviral DNA frequencies were performed on peripheral blood samples from 115 people with HIV (PWH) on long-term ART. Analysis demonstrated that both intact and total proviral DNA frequencies were positively correlated with T cell activation and exhaustion. Years of ART and select bifunctional HIV-specific CD4 T cell responses were negatively correlated with the percentage of intact proviruses. A leave-one-covariate-out inference approach identified specific HIV reservoir and clinical-demographic parameters, such as age and biological sex, that were particularly important in predicting immunophenotypes. Overall, immune parameters were more strongly associated with total HIV proviral frequencies than intact proviral frequencies. Uniquely, however, expression of the IL-7 receptor alpha chain (CD127) on CD4 T cells was more strongly correlated with the intact reservoir. Unsupervised dimension reduction analysis identified two main clusters of PWH with distinct immune and reservoir characteristics. Using reservoir correlates identified in these initial analyses, decision tree methods were employed to visualize relationships among multiple immune and clinical-demographic parameters and the HIV reservoir. Finally, using random splits of our data as training-test sets, ML algorithms predicted with approximately 70% accuracy whether a given participant had qualitatively high or low levels of total or intact HIV DNA . The techniques described here may be useful for assessing global patterns within the increasingly high-dimensional data used in HIV reservoir and other studies of complex biology.
Collapse
Affiliation(s)
| | - Yingfan Wang
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - Shane Falcinelli
- UNC HIV Cure Center UNC Chapel HillChapel HillUnited States
- Department of Microbiology and Immunology, UNC Chapel HillChapel HillUnited States
| | - Nancie Archin
- UNC HIV Cure Center UNC Chapel HillChapel HillUnited States
- Department of Medicine, UNC Chapel HillChapel HillUnited States
| | | | - David M Margolis
- UNC HIV Cure Center UNC Chapel HillChapel HillUnited States
- Department of Microbiology and Immunology, UNC Chapel HillChapel HillUnited States
- Department of Medicine, UNC Chapel HillChapel HillUnited States
| | - Nilu Goonetilleke
- UNC HIV Cure Center UNC Chapel HillChapel HillUnited States
- Department of Microbiology and Immunology, UNC Chapel HillChapel HillUnited States
| | | | - Cynthia D Rudin
- Department of Computer Science, Duke UniversityDurhamUnited States
| | - Edward P Browne
- UNC HIV Cure Center UNC Chapel HillChapel HillUnited States
- Department of Microbiology and Immunology, UNC Chapel HillChapel HillUnited States
- Department of Medicine, UNC Chapel HillChapel HillUnited States
| |
Collapse
|
17
|
Zeng X, Wang L, Zhang X, Zheng H, Song S, Xu T, Zhang H, Yang P. Nemo mRNA vaccination improves airway barrier function in mice with airway allergy. Cell Signal 2024; 121:111257. [PMID: 38857681 DOI: 10.1016/j.cellsig.2024.111257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Epithelial barrier dysfunction plays an important role in the pathogenesis of Th2 bias. The mechanism requires further clarification. NEMO is associated with regulating apoptotic activities in the cell. The purpose of this study is to investigate the role of insufficient Nemo signals in developing Th2 bias in the respiratory tract. Nemof/fEpcam-Cre mice (A mouse strain carrying NEMO-deficient epithelial cells. NemoKO mice, in short) was generated. An airway Th2 bias mouse model was established with the ovalbumin/alum protocol. The NemoKO mice exhibited spontaneous airway Th2 bias. Respiratory tract epithelial barrier integrity was compromised in NemoKO mice. Apoptosis was found in approximately 10% of the epithelial cells of the respiratory tract in NemoKO mice. The reconstruction of the Nemo expression restored homeostasis within the epithelial barrier of the airways. Restoration of Nemo gene expression in epithelial cells by Nemo mRNA vaccination alleviated Th2 bias in mice with airway allergy. To sum up, NEMO plays an important role in maintaining the integrity of the epithelial barrier in the respiratory tract. Administration of NEMO mRNA vaccines can restore epithelial barrier functions and alleviate Th2 bias in the airways.
Collapse
Affiliation(s)
- Xianhai Zeng
- Longgang ENT Hospital, Shenzhen ENT Institute & Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Lihuan Wang
- Department of Allergy Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiwen Zhang
- Shenzhen Clinical School of Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; Department of General Practice Medicine and Pulmonary Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Haoyue Zheng
- Shenzhen Clinical School of Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; Department of General Practice Medicine and Pulmonary Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Shuo Song
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; Department of General Practice Medicine and Pulmonary Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Tao Xu
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; Department of General Practice Medicine and Pulmonary Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Huanping Zhang
- Department of Allergy Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China.
| | - Pingchang Yang
- Longgang ENT Hospital, Shenzhen ENT Institute & Shenzhen Key Laboratory of ENT, Shenzhen, China; State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China.
| |
Collapse
|
18
|
Shorer EF, Dastgheyb RM, French AL, Daubert E, Morack R, Yohannes T, Clish C, Gustafson D, Sharma A, Rogando A, Qi Q, Burgess H, Rubin LH, Weber KM. Tryptophan-Kynurenine Pathway Activation and Cognition in Virally Suppressed Women With HIV. J Acquir Immune Defic Syndr 2024; 96:494-500. [PMID: 38985447 PMCID: PMC11236271 DOI: 10.1097/qai.0000000000003454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/22/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Immune and cognitive dysfunction persists even in virally suppressed women with HIV (VS-WWH). Since inflammation and HIV proteins induce the enzyme indoleamine 2,3-dioxygenase (IDO), converting tryptophan (T) to kynurenine (K) while producing downstream neurotoxic metabolites, we investigated IDO activation (KT ratio) in relation to cognition in VS-WWH and demographically similar women without HIV (WWoH). METHODS Ninety-nine VS-WWH on stable antiretroviral therapy and 102 WWoH (median age 52 vs 54 years; 73% vs 74% Black, respectively) from the New York and Chicago sites of the Women's Interagency HIV Study (WIHS) completed a neuropsychological test battery assessing motor function, processing speed, attention/working memory, verbal fluency, verbal learning and memory, and executive function and had plasma measured for tryptophan-kynurenine metabolites through liquid chromatography-tandem mass spectrometry and monocyte-derived [soluble cluster of differentiation-14 (sCD14), soluble cluster of differentiation-163 (sCD163), monocyte chemoattractant protein-1 (MCP-1)] plus general inflammatory markers [tumor necrosis factor alpha-2 receptor (TNF-R2), high-sensitivity C-reactive protein, high-sensitivity interleukin-6] through enzyme-linked immunosorbent assays between 2017 and 2020. RESULTS VS-WWH had a higher KT ratio (P < 0.01) and higher sCD14 levels (P < 0.05) compared with WWoH. Higher sCD163 was associated with higher KT ratio (R = 0.29, P < 0.01) and worse fine motor function in VS-WWH; after adjusting for sCD163 and sCD14 in multivariable regressions, higher KT ratio remained significantly associated with impaired fine motor function in VS-WWH only (standardized β = -0.29, P < 0.05). IDO activation was not associated with cognition in WWoH. CONCLUSIONS IDO activation (K:T) was associated with worse fine motor control in VS-WWH independent of measured systemic inflammation. Further studies investigating biological mechanisms linking IDO activation to fine motor function among VS-WWH are warranted.
Collapse
Affiliation(s)
| | | | - Audrey L. French
- Department of Medicine, Stroger Hospital of Cook County, Chicago IL
| | | | | | | | - Clary Clish
- Metabolomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Deborah Gustafson
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Andre Rogando
- Hektoen Institute of Medicine, Chicago, IL
- College of Science and Health, Charles R. Drew University of Medicine and Science, Los Angeles, CA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Helen Burgess
- Department of Psychiatry, University of Michigan, Ann Arbor, MI; and
| | - Leah H. Rubin
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD
- Departments of Psychiatry and Behavioral Sciences
- Molecular and Comparative Pathobiology; and
- Epidemiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
19
|
Shafqat A, Masters MC, Tripathi U, Tchkonia T, Kirkland JL, Hashmi SK. Long COVID as a disease of accelerated biological aging: An opportunity to translate geroscience interventions. Ageing Res Rev 2024; 99:102400. [PMID: 38945306 DOI: 10.1016/j.arr.2024.102400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
It has been four years since long COVID-the protracted consequences that survivors of COVID-19 face-was first described. Yet, this entity continues to devastate the quality of life of an increasing number of COVID-19 survivors without any approved therapy and a paucity of clinical trials addressing its biological root causes. Notably, many of the symptoms of long COVID are typically seen with advancing age. Leveraging this similarity, we posit that Geroscience-which aims to target the biological drivers of aging to prevent age-associated conditions as a group-could offer promising therapeutic avenues for long COVID. Bearing this in mind, this review presents a translational framework for studying long COVID as a state of effectively accelerated biological aging, identifying research gaps and offering recommendations for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shahrukh K Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Research and Innovation Center, Department of Health, Abu Dhabi, UAE; College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
20
|
Grunblatt E, Feinstein MJ. Precision Phenotyping of Heart Failure in People with HIV: Early Insights and Challenges. Curr Heart Fail Rep 2024; 21:417-427. [PMID: 38940893 DOI: 10.1007/s11897-024-00674-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE OF REVIEW People with HIV have an elevated risk of developing heart failure even with optimally controlled disease. In this review, we outline the various mechanisms through which HIV infection may directly and indirectly contribute to heart failure pathology and highlight the emerging relationship between HIV, chronic inflammation, and cardiometabolic disease. RECENT FINDINGS HIV infection leads to chronic inflammation, immune dysregulation, and metabolic imbalances even in those with well controlled disease. These dysregulations occur through several diverse mechanisms which may lead to manifestations of different phenotypes of heart failure in people with HIV. While it has long been known that people with HIV are at risk of developing heart failure, recent studies have suggested numerous complex mechanisms involving chronic inflammation, immune dysregulation, and metabolic derangement through which this may be mediated. Further comprehensive studies are needed to elucidate the precise relationship between these mechanisms and the development of different subtypes of heart failure in people with HIV.
Collapse
Affiliation(s)
- Eli Grunblatt
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA
| | - Matthew J Feinstein
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA.
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
21
|
Marini S, Huber A, Cash MN, Salemi M, Cook RL, Borsa P, Mavian CN. Oral Cannabidiol Treatment Is Associated with an Anti-Inflammatory Gene Expression Signature in Myeloid Cells of People Living with HIV. Cannabis Cannabinoid Res 2024; 9:1028-1037. [PMID: 38252549 DOI: 10.1089/can.2023.0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024] Open
Abstract
Introduction: HIV-related comorbidities appear to be related to chronic inflammation, a condition characterizing people living with HIV (PLWH). Prior work indicates that cannabidiol (CBD) might reduce inflammation; however, the genetics underpinning of this effect are not well investigated. Our main objective is to detect gene expression alterations in human peripheral blood mononuclear cells (PBMCs) from PLWH after at least 1 month of CBD treatment. Materials and Methods: We analyzed ∼41,000 PBMCs from three PLWH at baseline and after CBD treatment (27-60 days) through single-cell RNA sequencing. Results: We obtained a coherent signature, characterized by an anti-inflammatory activity, of differentially expressed genes in myeloid cells. Conclusions: Our study shows how CBD is associated with alterations of gene expression in myeloid cells after CBD treatment. Clinical Trial Registration: NCT05209867.
Collapse
Affiliation(s)
- Simone Marini
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Amanda Huber
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Melanie N Cash
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Marco Salemi
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Robert L Cook
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Paul Borsa
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Carla N Mavian
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
22
|
Bai F, Bono V, Borghi L, Bonazza F, Falcinella C, Vitaletti V, Miraglia F, Trunfio M, Calcagno A, Cusato J, Vegni E, d’Arminio Monforte A, Marchetti G. Association between tight junction proteins and cognitive performance in untreated persons with HIV. AIDS 2024; 38:1292-1303. [PMID: 38704619 PMCID: PMC11216391 DOI: 10.1097/qad.0000000000003923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/29/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND HIV-associated neurocognitive disorders (HAND) still affects persons with HIV (PWH) and their pathogenesis is not completely understood. We aimed to explore the association between plasma and cerebrospinal fluid (CSF) markers of blood-brain barrier (BBB) impairment and HAND in untreated PWH. DESIGN Cross-sectional study. METHODS We enrolled untreated PWH, who underwent blood examinations and lumbar puncture to measure inflammation (IL-15, TNF-α), BBB damage (zonulin and tight junction proteins, tight junction proteins: occludin, claudin-5) and endothelial adhesion molecules (VCAM-1, ICAM-1). A comprehensive neurocognitive battery was used to diagnose HAND (Frascati criteria). RESULTS Twenty-one patients (21/78, 26.9%) patients presented HAND (100% ANI). HAND patients displayed more frequently non-CNS AIDS-defining conditions, lower nadir CD4 + T cells and increased CD4 + T-cell exhaustion (lower CD4 + CD127 + and CD4 + CD45RA + T-cell percentages), in comparison to individuals without cognitive impairment. Furthermore, HAND was characterized by higher plasma inflammation (IL-15) but lower CSF levels of biomarkers of BBB impairment (zonulin and occludin). The association between BBB damage with HAND was confirmed by fitting a multivariable logistic regression. CSF/plasma endothelial adhesion molecules were not associated with HAND but with a poor performance in different cognitive domains. CONCLUSION By showing heightened inflammation and BBB impairment, our study suggests loss of BBB integrity as a possible factor contributing to the development of HAND in untreated PWH.
Collapse
Affiliation(s)
| | | | - Lidia Borghi
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Federica Bonazza
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Camilla Falcinella
- Unit of Infectious Diseases, ASST della Valle Olona, Busto Arsizio Hospital, Busto Arsizio
| | | | | | | | | | - Jessica Cusato
- Laboratory of Pharmacology and Pharmacotherapy, Amedeo di Savoia Hospital, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elena Vegni
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | | | | |
Collapse
|
23
|
Peters BA, Hanna DB, Xue X, Weber K, Appleton AA, Kassaye SG, Topper E, Tracy RP, Guillemette C, Caron P, Tien PC, Qi Q, Burk RD, Sharma A, Anastos K, Kaplan RC. Menopause and Estrogen Associations With Gut Barrier, Microbial Translocation, and Immune Activation Biomarkers in Women With and Without HIV. J Acquir Immune Defic Syndr 2024; 96:214-222. [PMID: 38905473 PMCID: PMC11196004 DOI: 10.1097/qai.0000000000003419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/29/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVES Estrogens may protect the gut barrier and reduce microbial translocation and immune activation, which are prevalent in HIV infection. We investigated relationships of the menopausal transition and estrogens with gut barrier, microbial translocation, and immune activation biomarkers in women with and without HIV. DESIGN Longitudinal and cross-sectional studies nested in the Women's Interagency HIV Study. METHODS Intestinal fatty acid binding protein, lipopolysaccharide binding protein, and soluble CD14 (sCD14) levels were measured in serum from 77 women (43 with HIV) before, during, and after the menopausal transition (∼6 measures per woman over ∼13 years). A separate cross-sectional analysis was conducted among 72 postmenopausal women with HIV with these biomarkers and serum estrogens. RESULTS Women in the longitudinal analysis were a median age of 43 years at baseline. In piecewise, linear, mixed-effects models with cutpoints 2 years before and after the final menstrual period to delineate the menopausal transition, sCD14 levels increased over time during the menopausal transition (Beta [95% CI]: 38 [12 to 64] ng/mL/yr, P = 0.004), followed by a decrease posttransition (-46 [-75 to -18], P = 0.001), with the piecewise model providing a better fit than a linear model (P = 0.0006). In stratified analyses, these results were only apparent in women with HIV. In cross-sectional analyses, among women with HIV, free estradiol inversely correlated with sCD14 levels (r = -0.26, P = 0.03). Lipopolysaccharide binding protein and intestinal fatty acid binding protein levels did not appear related to the menopausal transition and estrogen levels. CONCLUSIONS Women with HIV may experience heightened innate immune activation during menopause, possibly related to the depletion of estrogens.
Collapse
Affiliation(s)
- Brandilyn A. Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathleen Weber
- Cook County Health/Hektoen Institute of Medicine, Chicago, IL, USA
| | - Allison A. Appleton
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, Rensselaer, NY, United States
| | | | - Elizabeth Topper
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec - Université Laval Research Center, Cancer research center (CRC) and Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Patrick Caron
- Centre Hospitalier Universitaire (CHU) de Québec - Université Laval Research Center, Cancer research center (CRC) and Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Phyllis C. Tien
- Department of Veterans Affairs Medical Center, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert D. Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Microbiology and Immunology and Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
24
|
Lake JE, Feng H, Hyatt AN, Miao H, Debroy P, Funderburg N, Ailstock K, Dobs A, Haberlen S, Magnani JW, Margolick JB, McGowan K, Palella FJ, Witt MD, Bhasin S, Budoff MJ, Post WS, Brown TT. Transgender Women With Suppressed Testosterone Display Lower Burden of Coronary Disease Than Matched Cisgender Men. J Endocr Soc 2024; 8:bvae120. [PMID: 38974987 PMCID: PMC11223995 DOI: 10.1210/jendso/bvae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 07/09/2024] Open
Abstract
Context Cardiovascular disease (CVD) in transgender women (TW) may be affected by gender-affirming hormone therapy (GAHT) and HIV, but few data compare TW on contemporary GAHT to well-matched controls. Objective We compared CVD burden and biomarker profiles between TW and matched cisgender men (CM). Methods Adult TW on GAHT (n = 29) were recruited for a cross-sectional study (2018-2020). CM (n = 48) from the former Multicenter AIDS Cohort Study were matched 2:1 to TW on HIV serostatus, age ±5 years, race/ethnicity, BMI category and antiretroviral therapy (ART) type. Cardiac parameters were measured by CT and coronary atherosclerosis by coronary CT angiography; sex hormone and biomarker concentrations were measured centrally from stored samples. Results Overall, median age was 53 years and BMI 29 kg/m2; 69% were non-white. All participants with HIV (71%) had viral suppression on ART. Only 31% of TW had testosterone suppression (<50 ng/dL, TW-S). Traditional CVD risk factors were similar between groups, except that TW-S had higher BMI than TW with non-suppressed testosterone (TW-T). TW-S had no evidence of non-calcified coronary plaque or advanced coronary stenosis, whereas TW-T and CM had similar burden. TW had lower prevalence of any coronary plaque, calcified plaque and mixed plaque than CM, regardless of testosterone concentrations and HIV serostatus. Estradiol but not testosterone concentrations moderately and negatively correlated with the presence of coronary plaque and stenosis. Small sample size limited statistical power. Conclusion Older TW with suppressed total testosterone on GAHT had no CT evidence of non-calcified coronary plaque or advanced coronary stenosis. Longitudinal studies to understand relationships between GAHT and CVD risk in TW are needed.
Collapse
Affiliation(s)
- Jordan E Lake
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Han Feng
- Tulane Research and Innovation for Arrhythmia Discoveries-TRIAD Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ana N Hyatt
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Hongyu Miao
- Department of Statistics and College of Nursing, Florida State University, Tallahassee, FL 32306, USA
| | - Paula Debroy
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Nicholas Funderburg
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Kate Ailstock
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Adrian Dobs
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sabina Haberlen
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jared W Magnani
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joseph B Margolick
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kate McGowan
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Frank J Palella
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mallory D Witt
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Shalender Bhasin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Wendy S Post
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Todd T Brown
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
25
|
Riitho V, Connon R, Gwela A, Namusanje J, Nhema R, Siika A, Bwakura-Dangarembizi M, Musiime V, Berkley JA, Szubert AJ, Gibb DM, Walker AS, Klein N, Prendergast AJ. Biomarkers of mortality in adults and adolescents with advanced HIV in sub-Saharan Africa. Nat Commun 2024; 15:5492. [PMID: 38944653 PMCID: PMC11214617 DOI: 10.1038/s41467-024-49317-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/30/2024] [Indexed: 07/01/2024] Open
Abstract
One-third of people with HIV in sub-Saharan Africa start antiretroviral therapy (ART) with advanced disease. We investigated associations between immune biomarkers and mortality in participants with advanced HIV randomised to cotrimoxazole or enhanced antimicrobial prophylaxis in the Reduction of Early Mortality in HIV-Infected Adults and Children Starting Antiretroviral Therapy (REALITY) trial (ISRCTN43622374). Biomarkers were assayed using ELISA and Luminex. Associations between baseline values and all-cause 24-week mortality were analysed using Cox models, and for cause-specific mortality used Fine & Gray models, including prophylaxis randomisation, viral load, CD4, WHO stage, age, BMI, and site as covariates; and weighted according to inverse probability of selection into the substudy. Higher baseline CRP, IFN-γ, IL-6 and IP-10 were associated with higher all-cause mortality; and higher IL-23, IL-2 and RANTES with lower all-cause mortality. Associations varied by cause of death: tuberculosis-associated mortality was most strongly associated with higher CRP and sST2, and cryptococcosis-associated mortality with higher IL-4 and lower IL-8. Changes in I-FABP (p = 0.002), faecal alpha-1 antitrypsin (p = 0.01) and faecal myeloperoxidase (p = 0.005) between baseline and 4 weeks post-ART were greater in those receiving enhanced versus cotrimoxazole prophylaxis. Our findings highlight how the immune milieu shapes outcomes following ART initiation, and how adjunctive antimicrobials can modulate the gut environment in advanced HIV.
Collapse
Affiliation(s)
- Victor Riitho
- Blizard Institute, Queen Mary University of London, London, UK
- Center for Epidemiological Modelling and Analysis (CEMA), Institute of Tropical and Infectious Diseases (UNITID), University of Nairobi, Nairobi, Kenya
| | | | - Agnes Gwela
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Ruth Nhema
- University of Zimbabwe, Harare, Zimbabwe
| | | | | | - Victor Musiime
- Joint Clinical Research Centre, Kampala, Uganda
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | | | | | | | | | | | | |
Collapse
|
26
|
Nemphos SM, Green HC, Prusak JE, Fell SL, Goff K, Varnado M, Didier K, Guy N, Moström MJ, Tatum C, Massey C, Barnes MB, Rowe LA, Allers C, Blair RV, Embers ME, Maness NJ, Marx PA, Grasperge B, Kaur A, De Paris K, Shaffer JG, Hensley-McBain T, Londono-Renteria B, Manuzak JA. Elevated Inflammation Associated with Markers of Neutrophil Function and Gastrointestinal Disruption in Pilot Study of Plasmodium fragile Co-Infection of ART-Treated SIVmac239+ Rhesus Macaques. Viruses 2024; 16:1036. [PMID: 39066199 PMCID: PMC11281461 DOI: 10.3390/v16071036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Human immunodeficiency virus (HIV) and malaria, caused by infection with Plasmodium spp., are endemic in similar geographical locations. As a result, there is high potential for HIV/Plasmodium co-infection, which increases the pathology of both diseases. However, the immunological mechanisms underlying the exacerbated disease pathology observed in co-infected individuals are poorly understood. Moreover, there is limited data available on the impact of Plasmodium co-infection on antiretroviral (ART)-treated HIV infection. Here, we used the rhesus macaque (RM) model to conduct a pilot study to establish a model of Plasmodium fragile co-infection during ART-treated simian immunodeficiency virus (SIV) infection, and to begin to characterize the immunopathogenic effect of co-infection in the context of ART. We observed that P. fragile co-infection resulted in parasitemia and anemia, as well as persistently detectable viral loads (VLs) and decreased absolute CD4+ T-cell counts despite daily ART treatment. Notably, P. fragile co-infection was associated with increased levels of inflammatory cytokines, including monocyte chemoattractant protein 1 (MCP-1). P. fragile co-infection was also associated with increased levels of neutrophil elastase, a plasma marker of neutrophil extracellular trap (NET) formation, but significant decreases in markers of neutrophil degranulation, potentially indicating a shift in the neutrophil functionality during co-infection. Finally, we characterized the levels of plasma markers of gastrointestinal (GI) barrier permeability and microbial translocation and observed significant correlations between indicators of GI dysfunction, clinical markers of SIV and Plasmodium infection, and neutrophil frequency and function. Taken together, these pilot data verify the utility of using the RM model to examine ART-treated SIV/P. fragile co-infection, and indicate that neutrophil-driven inflammation and GI dysfunction may underlie heightened SIV/P. fragile co-infection pathogenesis.
Collapse
Affiliation(s)
- Sydney M. Nemphos
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Hannah C. Green
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - James E. Prusak
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Sallie L. Fell
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Megan Varnado
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Kaitlin Didier
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Natalie Guy
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Matilda J. Moström
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Coty Tatum
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Chad Massey
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Mary B. Barnes
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Lori A. Rowe
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Carolina Allers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Robert V. Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Monica E. Embers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Nicholas J. Maness
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Preston A. Marx
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| | - Brooke Grasperge
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27559, USA
| | - Jeffrey G. Shaffer
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | | | - Berlin Londono-Renteria
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| | - Jennifer A. Manuzak
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| |
Collapse
|
27
|
Premeaux TA, Bowler S, Friday CM, Moser CB, Hoenigl M, Lederman MM, Landay AL, Gianella S, Ndhlovu LC. Machine learning models based on fluid immunoproteins that predict non-AIDS adverse events in people with HIV. iScience 2024; 27:109945. [PMID: 38812553 PMCID: PMC11134891 DOI: 10.1016/j.isci.2024.109945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/12/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024] Open
Abstract
Despite the success of antiretroviral therapy (ART), individuals with HIV remain at risk for experiencing non-AIDS adverse events (NAEs), including cardiovascular complications and malignancy. Several surrogate immune biomarkers in blood have shown predictive value in predicting NAEs; however, composite panels generated using machine learning may provide a more accurate advancement for monitoring and discriminating NAEs. In a nested case-control study, we aimed to develop machine learning models to discriminate cases (experienced an event) and matched controls using demographic and clinical characteristics alongside 49 plasma immunoproteins measured prior to and post-ART initiation. We generated support vector machine (SVM) classifier models for high-accuracy discrimination of individuals aged 30-50 years who experienced non-fatal NAEs at pre-ART and one-year post-ART. Extreme gradient boosting generated a high-accuracy model at pre-ART, while K-nearest neighbors performed poorly all around. SVM modeling may offer guidance to improve disease monitoring and elucidate potential therapeutic interventions.
Collapse
Affiliation(s)
- Thomas A. Premeaux
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Scott Bowler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Courtney M. Friday
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Carlee B. Moser
- Center for Biostatistics in AIDS Research in the Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Martin Hoenigl
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, San Diego, CA, USA
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Michael M. Lederman
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sara Gianella
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
28
|
Brenchley JM, Serrano-Villar S. From dysbiosis to defense: harnessing the gut microbiome in HIV/SIV therapy. MICROBIOME 2024; 12:113. [PMID: 38907315 PMCID: PMC11193286 DOI: 10.1186/s40168-024-01825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/26/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Although the microbiota has been extensively associated with HIV pathogenesis, the majority of studies, particularly those using omics techniques, are largely correlative and serve primarily as a basis for hypothesis generation. Furthermore, most have focused on characterizing the taxonomic composition of the bacterial component, often overlooking other levels of the microbiome. The intricate mechanisms by which the microbiota influences immune responses to HIV are still poorly understood. Interventional studies on gut microbiota provide a powerful tool to test the hypothesis of whether we can harness the microbiota to improve health outcomes in people with HIV. RESULTS Here, we review the multifaceted role of the gut microbiome in HIV/SIV disease progression and its potential as a therapeutic target. We explore the complex interplay between gut microbial dysbiosis and systemic inflammation, highlighting the potential for microbiome-based therapeutics to open new avenues in HIV management. These include exploring the efficacy of probiotics, prebiotics, fecal microbiota transplantation, and targeted dietary modifications. We also address the challenges inherent in this research area, such as the difficulty in inducing long-lasting microbiome alterations and the complexities of study designs, including variations in probiotic strains, donor selection for FMT, antibiotic conditioning regimens, and the hurdles in translating findings into clinical practice. Finally, we speculate on future directions for this rapidly evolving field, emphasizing the need for a more granular understanding of microbiome-immune interactions, the development of personalized microbiome-based therapies, and the application of novel technologies to identify potential therapeutic agents. CONCLUSIONS Our review underscores the importance of the gut microbiome in HIV/SIV disease and its potential as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jason M Brenchley
- Barrier Immunity Section, Lab of Viral Diseases, NIAID, NIH, Bethesda, MA, USA.
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramon y Cajal, IRYCIS and CIBERInfec, Madrid, Spain.
| |
Collapse
|
29
|
Sehl ME, Breen EC, Shih R, Li F, Zhang J, Langfelder P, Horvath S, Bream JH, Duggal P, Martinson J, Wolinsky SM, Martinez-Maza O, Ramirez CM, Jamieson BD. Decreased but persistent epigenetic age acceleration is associated with changes in T-cell subsets after initiation of highly active antiretroviral therapy in persons living with HIV. FRONTIERS IN BIOINFORMATICS 2024; 4:1356509. [PMID: 38855141 PMCID: PMC11157435 DOI: 10.3389/fbinf.2024.1356509] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/25/2024] [Indexed: 06/11/2024] Open
Abstract
Introduction Persons living with HIV (PLWH) experience the early onset of age-related illnesses, even in the setting of successful human immunodeficiency virus (HIV) suppression with highly active antiretroviral therapy (HAART). HIV infection is associated with accelerated epigenetic aging as measured using DNA methylation (DNAm)-based estimates of biological age and of telomere length (TL). Methods DNAm levels (Infinium MethylationEPIC BeadChip) from peripheral blood mononuclear cells from 200 PLWH and 199 HIV-seronegative (SN) participants matched on chronologic age, hepatitis C virus, and time intervals were used to calculate epigenetic age acceleration, expressed as age-adjusted acceleration residuals from 4 epigenetic clocks [Horvath's pan-tissue age acceleration residual (AAR), extrinsic epigenetic age acceleration (EEAA), phenotypic epigenetic age acceleration (PEAA), and grim epigenetic age acceleration (GEAA)] plus age-adjusted DNAm-based TL (aaDNAmTL). Epigenetic age acceleration was compared for PLWH and SN participants at two visits: up to 1.5 years prior and 2-3 years after HAART (or equivalent visits). Flow cytometry was performed in PLWH and SN participants at both visits to evaluate T-cell subsets. Results Epigenetic age acceleration in PLWH decreased after the initiation of HAART but remained greater post-HAART than that in age-matched SN participants, with differences in medians of 6.6, 9.1, and 7.7 years for AAR, EEAA, and PEAA, respectively, and 0.39 units of aaDNAmTL shortening (all p < 0.001). Cumulative HIV viral load after HAART initiation was associated with some epigenetic acceleration (EEAA, PEAA, and aaDNAmTL), but even PLWH with undetectable HIV post-HAART showed persistent epigenetic age acceleration compared to SN participants (p < 0.001). AAR, EEAA, and aaDNAmTL showed significant associations with total, naïve, and senescent CD8 T-cell counts; the total CD4 T-cell counts were associated with AAR, EEAA, and PEAA (p = 0.04 to <0.001). In an epigenome-wide analysis using weighted gene co-methylation network analyses, 11 modules demonstrated significant DNAm differences pre- to post-HAART initiation. Of these, nine were previously identified as significantly different from pre- to post-HIV infection but in the opposite direction. Discussion In this large longitudinal study, we demonstrated that, although the magnitude of the difference decreases with HAART is associated with the cumulative viral load, PLWH are persistently epigenetically older than age-matched SN participants even after the successful initiation of HAART, and these changes are associated with changes in T-cell subsets.
Collapse
Affiliation(s)
- Mary E. Sehl
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
| | - Elizabeth Crabb Breen
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, United States
| | - Roger Shih
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
| | - Fengxue Li
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joshua Zhang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, United States
| | - Peter Langfelder
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, United States
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, United States
- Altos Labs, San Diego Institute of Science, San Diego, CA, United States
| | - Jay H. Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Immunology Training Program, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jeremy Martinson
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Steven M. Wolinsky
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Otoniel Martinez-Maza
- Departments of Obstetrics and Gynecology and Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, United States
| | - Christina M. Ramirez
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Beth D. Jamieson
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Anidi IU, Sakai S, Brooks K, Fling SP, Wagner MJ, Lurain K, Lindestam Arlehamn CS, Sette A, Knox KS, Brenchley JM, Uldrick TS, Sharon E, Barber DL. Exacerbation of CMV and Nontuberculous Mycobacterial Infections Following PD-1 Blockade for HIV-Associated Kaposi Sarcoma. Open Forum Infect Dis 2024; 11:ofae183. [PMID: 38680611 PMCID: PMC11049581 DOI: 10.1093/ofid/ofae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Blockade of the co-inhibitory receptor PD-1 enhances antitumor responses by boosting the function of antigen-specific T cells. Although rare, PD-1 blockade in patients with cancer can lead to exacerbation of infection-associated pathology. Here, we detail the case of a 38-year-old man who was enrolled in a clinical trial for assessment of the safety and activity of anti-PD-1 therapy for Kaposi sarcoma in people with HIV well-controlled on antiretroviral therapy. Less than a week after receiving the first dose of anti-PD-1 antibody (pembrolizumab), he presented with severe abdominal pain associated with sudden exacerbations of preexisting cytomegalovirus (CMV) enteritis and nontuberculous mycobacterial mesenteric lymphadenitis. Plasma biomarkers of gastrointestinal tract damage were highly elevated compared with healthy controls, consistent with HIV-associated loss of gut epithelial barrier integrity. Moreover, CMV-specific CD8 T cells expressed high levels of PD-1, and 7 days following PD-1 blockade, there was an increase in the frequency of activated CD38+ Ki67+ CMV-specific CD8 T cells. This case highlights the potential for PD-1 blockade to drive rapid exacerbations of inflammatory symptoms when administered to individuals harboring multiple unresolved infections.
Collapse
Affiliation(s)
- Ifeanyichukwu U Anidi
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Shunsuke Sakai
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven P Fling
- Cancer Immunotherapy Trials Network, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michael J Wagner
- Division of Medical Oncology, University of Washington and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Cecilia S Lindestam Arlehamn
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, California, USA
| | - Kenneth S Knox
- Department of Internal Medicine, College of Medicine Phoenix, University of Arizona Health Sciences, Phoenix, Arizona, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas S Uldrick
- Cancer Immunotherapy Trials Network, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Elad Sharon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Daniel L Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Jabs DA, Schneider MF, Pak JW, Beck-Engeser G, Chan F, Ambayec GC, Hunt PW. Association of Intermediate-Stage Age-Related Macular Degeneration with Plasma Inflammatory Biomarkers in Persons with AIDS. OPHTHALMOLOGY SCIENCE 2024; 4:100437. [PMID: 38304607 PMCID: PMC10831313 DOI: 10.1016/j.xops.2023.100437] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 02/03/2024]
Abstract
Purpose To evaluate associations of plasma levels of inflammatory biomarkers with age-related macular degeneration (AMD) and cataract in persons with AIDS. Design Nested case-control study (analysis 1) and nested cohort study (analysis 2). Participants Analysis 1: persons with AIDS and incident intermediate-stage AMD (n = 26) and controls without AMD matched for age, race/ethnicity, and gender (n = 49) from The Longitudinal Study of Ocular Complications of AIDS. Analysis 2: 475 persons from LSOCA with baseline plasma biomarker levels followed prospectively for cataract. Methods In both analyses, cryopreserved plasma specimens obtained at baseline were assayed for monocyte chemoattractant protein (MCP)-1 (CC motif chemokine ligand [CCL] 2), macrophage inflammatory protein (MIP)-1β (CCL4), soluble tumor necrosis factor receptor (sTNFR) 2, interleukin (IL)-18, and fractalkine (CX3 motif chemokine ligand 1 [CX3CL1]). Main Outcome Measures Analysis 1: mean difference (cases - controls) in plasma biomarker levels. Analysis 2: incident cataract. Results After adjusting for plasma human immunodeficiency virus RNA level, CD4+ T-cell count, and smoking, elevated baseline plasma levels of sTNFR2 and IL-18 (mean differences [cases - controls] 0.11 log10[pg/mL]; 95% confidence interval [CI], 0.01-0.20; P = 0.024 and 0.13 log10[pg/mL]; 95% CI, 0.01-0.24; P = 0.037, respectively) each were associated with incident AMD. In a competing risk (with mortality) analysis, elevated baseline standardized log10 plasma levels of MCP-1, sTNFR2, IL-18, and fractalkine each were associated with a decreased cataract risk. Conclusions When combined with previous data suggesting that AMD is associated with elevated plasma levels of C-reactive protein, soluble CD14, and possibly IL-6, the association of elevated plasma levels of sTNFR2 and IL-18 with incident AMD, but not with incident cataract, suggests that innate immune system activation, and possibly NLRP3 inflammasome activation, may play a role in the pathogenesis of AMD in this population. Financial Disclosures The authors have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Douglas A. Jabs
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Wilmer Eye Institute, the Department of Ophthalmology, the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael F. Schneider
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jeong Won Pak
- Department of Ophthalmology and Visual Sciences, the University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Gabriele Beck-Engeser
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California
| | - Fay Chan
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California
| | - Gabrielle C. Ambayec
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California
| | - Peter W. Hunt
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California
| |
Collapse
|
32
|
Evans C, Mutasa K, Rukobo S, Govha M, Mushayanembwa P, Chasekwa B, Majo FD, Tavengwa NV, Broad J, Noble C, Gough EK, Kelly P, Bourke CD, Humphrey JH, Ntozini R, Prendergast AJ. Inflammation and cytomegalovirus viremia during pregnancy drive sex-differentiated differences in mortality and immune development in HIV-exposed infants. Nat Commun 2024; 15:2909. [PMID: 38632279 PMCID: PMC11024190 DOI: 10.1038/s41467-023-44166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 12/04/2023] [Indexed: 04/19/2024] Open
Abstract
Children who are HIV-exposed but uninfected have increased infectious mortality compared to HIV-unexposed children, raising the possibility of immune abnormalities following exposure to maternal viraemia, immune dysfunction, and co-infections during pregnancy. In a secondary analysis of the SHINE trial in rural Zimbabwe we explored biological pathways underlying infant mortality, and maternal factors shaping immune development in HIV-exposed uninfected infants. Maternal inflammation and cytomegalovirus viraemia were independently associated with infant deaths: mortality doubled for each log10 rise in maternal C-reactive protein (adjusted hazard ratio (aHR) 2.09; 95% CI 1.33-3.27), and increased 1.6-fold for each log10 rise in maternal cytomegalovirus viral load (aHR 1.62; 95% CI 1.11-2.36). In girls, mortality was more strongly associated with maternal C-reactive protein than cytomegalovirus; in boys, mortality was more strongly associated with cytomegalovirus than C-reactive protein. At age one month, HIV-exposed uninfected infants had a distinct immune milieu, characterised by raised soluble CD14 and an altered CD8 + T-cell compartment. Alterations in immunophenotype and systemic inflammation were generally greater in boys than girls. Collectively, these findings show how the pregnancy immune environment in women with HIV underlies mortality and immune development in their offspring in a sex-differentiated manner, and highlights potential new intervention strategies to transform outcomes of HIV-exposed children. ClinicalTrials.gov/NCT01824940.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.
- Blizard Institute, Queen Mary University of London, London, UK.
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jonathan Broad
- Blizard Institute, Queen Mary University of London, London, UK
| | - Christie Noble
- Blizard Institute, Queen Mary University of London, London, UK
| | - Ethan K Gough
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Paul Kelly
- Blizard Institute, Queen Mary University of London, London, UK
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, UK
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, UK
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
33
|
Zhou J, Yang Y, Xie Z, Lu D, Huang J, Lan L, Guo B, Yang X, Wang Q, Li Z, Zhang Y, Yang X, Ai S, Liu N, Cui P, Liang H, Ye L, Huang J. Dysbiosis of gut microbiota and metabolites during AIDS: implications for CD4 + T cell reduction and immune activation. AIDS 2024; 38:633-644. [PMID: 38061029 PMCID: PMC10942204 DOI: 10.1097/qad.0000000000003812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/11/2023] [Accepted: 11/29/2023] [Indexed: 03/16/2024]
Abstract
OBJECTIVE Identifying the gut microbiota associated with host immunity in the AIDS stage. DESIGN We performed a cross-sectional study. METHODS We recruited people with HIV (PWH) in the AIDS or non-AIDS stage and evaluated their gut microbiota and metabolites by using 16S ribosomal RNA (rRNA) sequencing and liquid chromatography-mass spectrometry (LC-MS). Machine learning models were used to analyze the correlations between key bacteria and CD4 + T cell count, CD4 + T cell activation, bacterial translocation, gut metabolites, and KEGG functional pathways. RESULTS We recruited 114 PWH in the AIDS stage and 203 PWH in the non-AIDS stage. The α-diversity of gut microbiota was downregulated in the AIDS stage ( P < 0.05). Several machine learning models could be used to identify key gut microbiota associated with AIDS, including the logistic regression model with area under the curve (AUC), sensitivity, specificity, and Brier scores of 0.854, 0.813, 0.813, and 0.160, respectively. The decreased key bacteria ASV1 ( Bacteroides sp.), ASV8 ( Fusobacterium sp.), ASV30 ( Roseburia sp.), ASV37 ( Bacteroides sp.), and ASV41 ( Lactobacillus sp.) in the AIDS stage were positively correlated with the CD4 + T cell count, the EndoCAb IgM level, 4-hydroxyphenylpyruvic acid abundance, and the predicted cell growth pathway, and negatively correlated with the CD3 + CD4 + CD38 + HLA-DR + T cell count and the sCD14 level. CONCLUSION Machine learning has the potential to recognize key gut microbiota related to AIDS. The key five bacteria in the AIDS stage and their metabolites might be related to CD4 + T cell reduction and immune activation.
Collapse
Affiliation(s)
- Jie Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Yuecong Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | | | - Dongjia Lu
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | | | - Liuyang Lan
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Baodong Guo
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Xiping Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Qing Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Zhuoxin Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Yu Zhang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Xing Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| | - Sufang Ai
- The Fourth People's Hospital of Nanning
| | | | - Ping Cui
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease
| |
Collapse
|
34
|
Tincati C, Bono V, Cannizzo ES, Tosi D, Savi F, Falcinella C, Casabianca A, Orlandi C, Luigiano C, Augello M, Rusconi S, Muscatello A, Bandera A, Calcagno A, Gori A, Nozza S, Marchetti G. Primary HIV infection features colonic damage and neutrophil inflammation yet containment of microbial translocation. AIDS 2024; 38:623-632. [PMID: 38016163 PMCID: PMC10942218 DOI: 10.1097/qad.0000000000003799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/20/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION Impairment of the gastrointestinal barrier leads to microbial translocation and peripheral immune activation, which are linked to disease progression. Data in the setting of primary HIV/SIV infection suggest that gut barrier damage is one of the first events of the pathogenic cascade, preceding mucosal immune dysfunction and microbial translocation. We assessed gut structure and immunity as well as microbial translocation in acutely and chronically-infected, combination antiretroviral therapy (cART)-naive individuals. METHODS Fifteen people with primary HIV infection (P-HIV) and 13 with chronic HIV infection (C-HIV) c-ART-naive participants were cross-sectionally studied. Gut biopsies were analysed in terms of gut reservoirs (total, integrated and unintegrated HIV DNA); tight junction proteins (E-cadherin, Zonula Occludens-1), CD4 + expression, neutrophil myeloperoxidase (histochemical staining); collagen deposition (Masson staining). Flow cytometry was used to assess γδ T-cell frequency (CD3 + panγδ+Vδ1+/Vδ2+). In plasma, we measured microbial translocation (LPS, sCD14, EndoCAb) and gut barrier function (I-FABP) markers (ELISA). RESULTS P-HIV displayed significantly higher tissue HIV DNA, yet neutrophil infiltration and collagen deposition in the gut were similar in the two groups. In contrast, microbial translocation markers were significantly lower in P-HIV compared with C-HIV. A trend to higher mucosal E-cadherin, and gut γδ T-cells was also observed in P-HIV. CONCLUSION Early HIV infection features higher HIV DNA in the gut, yet comparable mucosal alterations to those observed in chronic infection. In contrast, microbial translocation is contained in primary HIV infection, likely because of a partial preservation of E-cadherin and mucosal immune subsets, namely γδ T-cells.
Collapse
Affiliation(s)
- Camilla Tincati
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan
| | - Valeria Bono
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan
| | | | - Delfina Tosi
- Pathology Unit, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan
| | - Federica Savi
- Pathology Unit, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan
| | - Camilla Falcinella
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan
| | - Anna Casabianca
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Fano
| | - Chiara Orlandi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Fano
| | | | - Matteo Augello
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan
| | - Stefano Rusconi
- UOC Malattie Infettive, Ospedale Civile di Legnano, Department of Biomedical and Clinical Biosciences, University of Milan
| | - Antonio Muscatello
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan
| | - Andrea Calcagno
- Unit of Infectious Diseases Unit, Department of Medical Sciences, University of Turin, Turin
| | - Andrea Gori
- Clinic of Infectious Diseases, Department of Pathophysiology and Transplantation, ASST Fatebenefratelli Sacco University of Milan
| | - Silvia Nozza
- Infectious Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases, Department of Health Sciences, University of Milan
| |
Collapse
|
35
|
Clemente T, Galli L, Lolatto R, Gagliardini R, Lagi F, Ferrara M, Cattelan AM, Focà E, Di Biagio A, Cervo A, Calza L, Maggiolo F, Marchetti G, Cenderello G, Rusconi S, Zazzi M, Santoro MM, Spagnuolo V, Castagna A. Cohort profile: PRESTIGIO, an Italian prospective registry-based cohort of people with HIV-1 resistant to reverse transcriptase, protease and integrase inhibitors. BMJ Open 2024; 14:e080606. [PMID: 38341206 PMCID: PMC10862296 DOI: 10.1136/bmjopen-2023-080606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/04/2024] [Indexed: 02/12/2024] Open
Abstract
PURPOSE The PRESTIGIO Registry was established in 2017 to collect clinical, virological and immunological monitoring data from people living with HIV (PLWH) with documented four-class drug resistance (4DR). Key research purposes include the evaluation of residual susceptibility to specific antiretrovirals and the validation of treatment and monitoring strategies in this population. PARTICIPANTS The PRESTIGIO Registry collects annual plasma and peripheral blood mononuclear cell samples and demographic, clinical, virological, treatment and laboratory data from PLWH followed at 39 Italian clinical centres and characterised by intermediate-to-high genotypic resistance to ≥1 nucleoside reverse transcriptase inhibitors, ≥1 non-nucleoside reverse transcriptase inhibitors, ≥1 protease inhibitors, plus either intermediate-to-high genotypic resistance to ≥1 integrase strand transfer inhibitors (INSTIs) or history of virological failure to an INSTI-containing regimen. To date, 229 people have been recorded in the cohort. Most of the data are collected from the date of the first evidence of 4DR (baseline), with some prebaseline information obtained retrospectively. Samples are collected from the date of enrollment in the registry. FINDINGS TO DATE The open-ended cohort has been used to assess (1) prognosis in terms of survival or development of AIDS-related or non-AIDS-related clinical events; (2) long-term efficacy and safety of different antiretroviral regimens and (3) virological and immunological factors predictive of clinical outcome and treatment efficacy, especially through analysis of plasma and cell samples. FUTURE PLANS The registry can provide new knowledge on how to implement an integrated approach to study PLWH with documented resistance to the four main antiretroviral classes, a population with a limited number of individuals characterised by a high degree of frailty and complexity in therapeutic management. Given the scheduled annual updates of PLWH data, the researchers who collaborate in the registry can send study proposals at any time to the steering committee of the registry, which evaluates every 3 months whether the research studies can be conducted on data and biosamples from the registry and whether they are aimed at a better understanding of a specific health condition, the emergence of comorbidities or the effect of potential treatments or experimental drugs that may have an impact on disease progression and quality of life. Finally, the research studies should aim to be inclusive, innovative and in touch with the communities and society as a whole. TRIAL REGISTRATION NUMBER NCT04098315.
Collapse
Affiliation(s)
- Tommaso Clemente
- Vita-Salute San Raffaele University, Milan, Italy
- Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Galli
- Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Lolatto
- Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Gagliardini
- National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Filippo Lagi
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Micol Ferrara
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Maria Cattelan
- Infectious Diseases Unit, Department of Molecular Medicine, Padua University Hospital, Padua, Italy
| | - Emanuele Focà
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, ASST Spedali di Brescia, Brescia, Italy
| | - Antonio Di Biagio
- Clinic of Infectious Diseases, IRCCS Policlinico San Martino Hospital, University of Genoa, Genoa, Italy
| | - Adriana Cervo
- Infectious Diseases Unit, Policlinico di Modena, Università Degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Leonardo Calza
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, S. Orsola Hospital, "Alma Mater Studiorum" University of Bologna, Bologna, Italy
| | - Franco Maggiolo
- Unit of HIV-related Diseases and Experimental Therapies, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases, Department of Health Sciences, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | | | - Stefano Rusconi
- Infectious Diseases Unit, ASST Ovest Milanese, Legnano General Hospital, Legnano, Italy
- DIBIC, University of Milan, Milan, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | | | - Vincenzo Spagnuolo
- Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Castagna
- Vita-Salute San Raffaele University, Milan, Italy
- Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
36
|
Rocafort M, Gootenberg DB, Luévano JM, Paer JM, Hayward MR, Bramante JT, Ghebremichael MS, Xu J, Rogers ZH, Munoz AR, Okello S, Kim JH, Sentongo R, Wagubi R, Lankowski A, Maruapula S, Zhao G, Handley SA, Mosepele M, Siedner MJ, Kwon DS. HIV-associated gut microbial alterations are dependent on host and geographic context. Nat Commun 2024; 15:1055. [PMID: 38316748 PMCID: PMC10844288 DOI: 10.1038/s41467-023-44566-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 12/19/2023] [Indexed: 02/07/2024] Open
Abstract
HIV-associated changes in intestinal microbiota are believed to be important drivers of disease progression. However, the majority of studies have focused on populations in high-income countries rather than in developing regions where HIV burden is greatest. To better understand the impact of HIV on fecal microbiota globally, we compare the fecal microbial community of individuals in the U.S., Uganda, and Botswana. We identify significant bacterial taxa alterations with both treated and untreated HIV infection with a high degree of uniqueness in each cohort. HIV-associated taxa alterations are also significantly different between populations that report men who have sex with men (MSM) behavior and non-MSM populations. Additionally, while we find that HIV infection is consistently associated with higher soluble markers of immune activation, most specific bacterial taxa associated with these markers in each region are not shared and none are shared across all three geographic locations in our study. Our findings demonstrate that HIV-associated changes in fecal microbiota are overall distinct among geographical locations and sexual behavior groups, although a small number of taxa shared between pairs of geographic locations warrant further investigation, highlighting the importance of considering host context to fully assess the impact of the gut microbiome on human health and disease.
Collapse
Affiliation(s)
- Muntsa Rocafort
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - David B Gootenberg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Harvard Medical School, Boston, MA, 02114, USA
| | - Jesús M Luévano
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Harvard Medical School, Boston, MA, 02114, USA
| | - Jeffrey M Paer
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | | | | | - Jiawu Xu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Zoe H Rogers
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Samson Okello
- Department of Medicine, Mbarara University of Science and Technology, 1956, Mbarara, Uganda
| | - June-Ho Kim
- Harvard Medical School, Boston, MA, 02114, USA
| | - Ruth Sentongo
- Department of Medicine, Mbarara University of Science and Technology, 1956, Mbarara, Uganda
| | - Robert Wagubi
- Department of Medicine, Mbarara University of Science and Technology, 1956, Mbarara, Uganda
| | - Alex Lankowski
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Segametsi Maruapula
- Department of Family & Consumer Sciences, University of Botswana, 0022, Gaborone, Botswana
| | - Guoyan Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Mosepele Mosepele
- Faculty of Medicine, University of Botswana, 0022, Gaborone, Botswana
| | - Mark J Siedner
- Harvard Medical School, Boston, MA, 02114, USA
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, 02114, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Douglas S Kwon
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA.
- Harvard Medical School, Boston, MA, 02114, USA.
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA.
| |
Collapse
|
37
|
Srinivasa S, Abohashem S, Walpert AR, Dunderdale CN, Iyengar S, Shen G, Jerosch-Herold M, deFilippi CR, Robbins GK, Lee H, Kwong RY, Adler GK, Tawakol A, Grinspoon SK. Mineralocorticoid Receptor Antagonism by Eplerenone and Arterial Inflammation in HIV: The MIRABELLA HIV Study. JAMA Cardiol 2024; 9:189-194. [PMID: 38090987 PMCID: PMC10719827 DOI: 10.1001/jamacardio.2023.4578] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/19/2023] [Indexed: 12/17/2023]
Abstract
Importance The risk for atherosclerotic disease is increased 1.5- to 2.0-fold among persons with HIV (PWH). Increased activation of the renin-angiotensin-aldosterone system may contribute to increased arterial inflammation in this population. Objective To determine the effects of eplerenone on arterial inflammation among well-treated PWH without known cardiovascular disease (CVD). Design, Setting, and Participants Well-treated PWH who participated in the double-blinded, placebo-controlled, Mineralocorticoid Receptor Antagonism for Cardiovascular Health in HIV (MIRACLE HIV) study between February 2017 and March 2022 assessing the effects of eplerenone on myocardial perfusion were invited to participate in the Mineralocorticoid Receptor Antagonism By Eplerenone to Lower Arterial Inflammation in HIV (MIRABELLA) substudy if there was no current statin use. Participants were enrolled in the MIRABELLA study and underwent additional 18F-fludeoxyglucose-positron emission tomography/computed tomography (18F-FDG PET/CT) imaging of the aorta and carotid arteries to assess arterial inflammation over 12 months of treatment with eplerenone vs placebo. Interventions Eplerenone, 50 mg, twice a day vs identical placebo. Main Outcomes and Measures The primary outcome was change in target to background ratio (TBR), a measure of arterial wall inflammation, in the index vessel after 12 months of treatment. The index vessel was defined as the vessel (aorta, left carotid artery, or right carotid artery) with the highest TBR at baseline in each participant. Results A total of 26 participants (mean [SD] age, 54 [7] years; 18 male [69%]) were enrolled in the study. Treatment groups (eplerenone, 13 vs placebo, 13) were of similar age, sex, and body mass index. Eplerenone was associated with a reduction in TBR of the primary end point, the index vessel (eplerenone vs placebo: model treatment effect, -0.31; 95% CI, -0.50 to -0.11; P = .006; percentage change, -12.4% [IQR, -21.9% to -2.6%] vs 5.1% [IQR, -1.6% to 11.0%]; P = .003). We further observed a significant reduction of the TBR of the most diseased segment (MDS) of the index vessel (eplerenone vs placebo: -19.1% [IQR, -27.0% to -11.9%] vs 6.8% [IQR, -9.1% to 12.1%]; P = .007). A similar result was seen assessing the index vessel of the carotids (eplerenone vs placebo: -10.0% [IQR, -21.8% to 3.6%] vs 9.7% [IQR, -9.8% to 15.9%]; P = .046). Reduction in the TBR of MDS of the index vessel on 18F-FDG PET/CT correlated with improvement in the stress myocardial blood flow on cardiac magnetic resonance imaging (Spearman ρ = -0.67; P = .01). Conclusion and Relevance In this small randomized clinical trial, eplerenone was associated with reduction in arterial inflammation among well-treated PWH without known CVD. In addition, reductions in arterial inflammation as measured by 18F-FDG PET/CT were related to improvements in stress myocardial perfusion. Further larger studies should explore whether eplerenone is a potential treatment strategy for inflammatory-mediated CVD in PWH. Trial Registration ClinicalTrials.gov Identifier: NCT02740179.
Collapse
Affiliation(s)
- Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Shady Abohashem
- Cardiovascular Imaging Research Center and Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Allie R. Walpert
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | | | - Sanjna Iyengar
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Grace Shen
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michael Jerosch-Herold
- Cardiovascular Division of Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Gregory K. Robbins
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond Y. Kwong
- Cardiovascular Division of Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gail K. Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center and Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
38
|
Ron R, Moreno E, Rosas Cancio-Suárez M, Serrano-Villar S. The microbiome as a biomarker of anal precancerous lesions in people with HIV. Curr Opin Infect Dis 2024; 37:17-25. [PMID: 37889583 DOI: 10.1097/qco.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
PURPOSE OF REVIEW Early detection and treatment of human papillomavirus (HPV)-related anal dysplasia in some high-risk groups can help anal cancer prevention, but new tools to improve diagnostic and risk assessment are needed. Here, we aim to discuss the evidence on the role of the microbiome as a potential biomarker for anal high-grade squamous intraepithelial lesions (HSILs) in people with HIV (PWH). RECENT FINDINGS This review covers relevant studies on the links between the microbiome and HPV infection, cervical dysplasia/cancer, and anal HPV disease. It focuses on anal samples and precancerous lesions. SUMMARY The review highlights the promising potential of the anal microbiome as a novel biomarker for precancerous lesions in people with HIV, while also discussing limitations and future research needs.
Collapse
Affiliation(s)
- Raquel Ron
- Infectious Diseases Department, Hospital Universitario Ramón y Cajal, Madrid, Spain, CIBERINFEC
| | | | | | | |
Collapse
|
39
|
Lembas A, Załęski A, Peller M, Mikuła T, Wiercińska-Drapało A. Human Immunodeficiency Virus as a Risk Factor for Cardiovascular Disease. Cardiovasc Toxicol 2024; 24:1-14. [PMID: 37982976 PMCID: PMC10838226 DOI: 10.1007/s12012-023-09815-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The developments in HIV treatments have increased the life expectancy of people living with HIV (PLWH), a situation that makes cardiovascular disease (CVD) in that population as relevant as ever. PLWH are at increased risk of CVD, and our understanding of the underlying mechanisms is continually increasing. HIV infection is associated with elevated levels of multiple proinflammatory molecules, including IL-6, IL-1β, VCAM-1, ICAM-1, TNF-α, TGF-β, osteopontin, sCD14, hs-CRP, and D-dimer. Other currently examined mechanisms include CD4 + lymphocyte depletion, increased intestinal permeability, microbial translocation, and altered cholesterol metabolism. Antiretroviral therapy (ART) leads to decreases in the concentrations of the majority of proinflammatory molecules, although most remain higher than in the general population. Moreover, adverse effects of ART also play an important role in increased CVD risk, especially in the era of rapid advancement of new therapeutical options. Nevertheless, it is currently believed that HIV plays a more significant role in the development of metabolic syndromes than treatment-associated factors. PLWH being more prone to develop CVD is also due to the higher prevalence of smoking and chronic coinfections with viruses such as HCV and HBV. For these reasons, it is crucial to consider HIV a possible causal factor in CVD occurrence, especially among young patients or individuals without common CVD risk factors.
Collapse
Affiliation(s)
- Agnieszka Lembas
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Poland
- Hospital for Infectious Diseases, Warsaw, Poland
| | - Andrzej Załęski
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Poland.
- Hospital for Infectious Diseases, Warsaw, Poland.
| | - Michał Peller
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Mikuła
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Poland
- Hospital for Infectious Diseases, Warsaw, Poland
| | - Alicja Wiercińska-Drapało
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Poland
- Hospital for Infectious Diseases, Warsaw, Poland
| |
Collapse
|
40
|
Sereti I, Verburgh ML, Gifford J, Lo A, Boyd A, Verheij E, Verhoeven A, Wit FWNM, Schim van der Loeff MF, Giera M, Kootstra NA, Reiss P, Vujkovic-Cvijin I. Impaired gut microbiota-mediated short-chain fatty acid production precedes morbidity and mortality in people with HIV. Cell Rep 2023; 42:113336. [PMID: 37918403 PMCID: PMC10872975 DOI: 10.1016/j.celrep.2023.113336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Antiretroviral therapy (ART) has dramatically lengthened lifespan among people with HIV (PWH), but this population experiences heightened rates of inflammation-related comorbidities. HIV-associated inflammation is linked with an altered microbiome; whether such alterations precede inflammation-related comorbidities or occur as their consequence remains unknown. We find that ART-treated PWH exhibit depletion of gut-resident bacteria that produce short-chain fatty acids (SCFAs)-crucial microbial metabolites with anti-inflammatory properties. Prior reports establish that fecal SCFA concentrations are not depleted in PWH. We find that gut-microbiota-mediated SCFA production capacity is better reflected in serum than in feces and that PWH exhibit reduced serum SCFA, which associates with inflammatory markers. Leveraging stool and serum samples collected prior to comorbidity onset, we find that HIV-specific microbiome alterations precede morbidity and mortality in ART-treated PWH. Among these microbiome alterations, reduced microbiome-mediated conversion of lactate to propionate precedes mortality in PWH. Thus, gut microbial fiber/lactate conversion to SCFAs may modulate HIV-associated comorbidity risk.
Collapse
Affiliation(s)
- Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, NIAID/NIH, Rockville, MD, USA; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Myrthe L Verburgh
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Jacob Gifford
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alice Lo
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anders Boyd
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; HIV Monitoring Foundation, Amsterdam, the Netherlands; Public Health Service of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
| | - Eveline Verheij
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Aswin Verhoeven
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, the Netherlands
| | - Ferdinand W N M Wit
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; HIV Monitoring Foundation, Amsterdam, the Netherlands
| | - Maarten F Schim van der Loeff
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Public Health Service of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, the Netherlands
| | - Neeltje A Kootstra
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Experimental Immunology, Amsterdam, the Netherlands
| | - Peter Reiss
- Amsterdam University Medical Centers, University of Amsterdam, Infectious Diseases, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands; Amsterdam University Medical Centers, University of Amsterdam, Department of Global Health, Meibergdreef 9, Amsterdam, the Netherlands
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Munjoma PT, Chandiwana P, Wyss J, Mazhandu AJ, Jordi SBU, Gutsire R, Katsidzira L, Yilmaz B, Misselwitz B, Duri K. Immune activation and inflammation in lactating women on combination antiretroviral therapy: role of gut dysfunction and gut microbiota imbalance. Front Immunol 2023; 14:1280262. [PMID: 38045684 PMCID: PMC10693333 DOI: 10.3389/fimmu.2023.1280262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/17/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Combination antiretroviral therapy (cART) effectively controls HIV; however, chronic low-level viremia and gut microbiota dysbiosis remain significant drivers of gut and systemic inflammation. In this study, we explored the relationship between gut microbiota composition, intestinal inflammation, microbial translocation, and systemic inflammation in women on cART in Sub-Saharan Africa. Methods We conducted a study in HIV-infected and HIV-uninfected lactating women followed up at 6 weeks and 6 months postpartum in Harare, Zimbabwe. We used 16S ribosomal Ribonucleic Acid (rRNA) sequencing and MesoScale Discovery V-Plex assays to examine the gut microbiome and to quantify plasma inflammatory biomarkers, respectively. In addition, we measured fecal calprotectin, plasma lipopolysaccharide-binding protein (LBP), and soluble cluster of differentiation 14 (sCD14) by enzyme-linked immunosorbent assay to assess gut inflammation, microbial translocation, and monocyte/macrophage activation. Results A group of 77 lactating women were studied, of which 35% were HIV-infected. Fecal calprotectin levels were similar by HIV status at both follow-up time points. In the HIV-infected group at 6 weeks postpartum, fecal calprotectin was elevated: median (interquartile range) [158.1 µg/g (75.3-230.2)] in women who had CD4+ T-lymphocyte counts <350 cells/µL compared with those with ≥350 cells/µL [21.1 µg/g (0-58.4)], p = 0.032. Plasma sCD14 levels were significantly higher in the HIV-infected group at both 6 weeks and 6 months postpartum, p < 0.001. Plasma LBP levels were similar, but higher levels were observed in HIV-infected women with elevated fecal calprotectin. We found significant correlations between fecal calprotectin, LBP, and sCD14 with proinflammatory cytokines. Gut microbial alpha diversity was not affected by HIV status and was not affected by use of antibiotic prophylaxis. HIV significantly affected microbial beta diversity, and significant differences in microbial composition were noted. The genera Slackia and Collinsella were relatively more abundant in the HIV-infected group, whereas a lower relative abundance of Clostriduim sensu_stricto_1 was observed. Our study also found correlations between gut microbial taxa abundance and systemic inflammatory biomarkers. Discussion and conclusion HIV-infected lactating women had increased immune activation and increased microbial translocation associated with increased gut inflammation. We identified correlations between the gut inflammation and microbial composition, microbial translocation, and systemic inflammation. The interplay of these parameters might affect the health of this vulnerable population.
Collapse
Affiliation(s)
- Privilege Tendai Munjoma
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Panashe Chandiwana
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Jacqueline Wyss
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Arthur John Mazhandu
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Rutendo Gutsire
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Leolin Katsidzira
- Department of Internal Medicine, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, Maurice Müller Laboratories, University of Bern, Bern, Switzerland
| | - Kerina Duri
- Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| |
Collapse
|
42
|
Lake JE, Miao H, Bowman ER, Clark JL, Hyatt AN, Kettelhut A, Lama JR, Reisner SL, Mayer KH, Perez-Brumer A, Funderburg N. Gender-affirming hormone therapy decreases d-dimer but worsens insulin sensitivity in transgender women. HIV Med 2023; 24:1144-1149. [PMID: 37386803 PMCID: PMC10755063 DOI: 10.1111/hiv.13522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023]
Abstract
OBJECTIVES Gender-affirming hormonal therapies (GAHT) and HIV increase cardiovascular risk for transgender women (TW), yet there is a paucity of data quantifying cardiometabolic changes following GAHT initiation, particularly among TW with HIV. METHODS The Féminas study enrolled TW from October 2016 to March 2017 in Lima, Peru. Participants reported sexual activity that was high risk for HIV acquisition or transmission. All were tested for HIV/ sexually transmitted infection and were given access to GAHT (oestradiol valerate and spironolactone), HIV pre-exposure prophylaxis (PrEP) or antiretroviral therapy (ART) for 12 months. Biomarker measurement was done on stored serum, whereas fasting glucose and lipids were measured in real time. RESULTS In all, 170 TW (32 with HIV, 138 without HIV) had median age 27 years and 70% prior GAHT use. At baseline, PCSK9, sCD14, sCD163, IL-6, sTNFRI/II, CRP and EN-RAGE levels were significantly higher in TW with HIV than in TW without HIV. High-density lipoprotein and total cholesterol were lower and insulin and glucose parameters were similar. All TW with HIV started ART, but only five achieved virological suppression at any time. No TW without HIV initiated PrEP. Over 6 months, all participants initiated GAHT and had worsening insulin, glucose and HOMA-IR. Large d-dimer decreases also occurred. Similar changes occurred in TW with and without HIV. CONCLUSIONS In this unique cohort of TW, GAHT decreased d-dimer but worsened insulin sensitivity. Because PrEP uptake and ART adherence were very low, observed effects are primarily attributed to GAHT use. Further study is needed to better understand cardiometabolic changes in TW by HIV serostatus.
Collapse
Affiliation(s)
- Jordan E. Lake
- University of Texas Health Science Center at Houston, 6431 Fannin St. MSB 2.112, Houston, TX 77030, USA
| | - Hongyu Miao
- Florida State University, 600 W College Ave, Tallahassee, FL 32306, USA
| | - Emily R. Bowman
- The Ohio State University, 453 West 10 Ave, Columbus, OH 43210, USA
| | - Jesse L. Clark
- University of California, Los Angeles, 405 Hilgard Ave., Los Angeles 90095, CA, USA
| | - Ana N. Hyatt
- University of Texas Health Science Center at Houston, 6431 Fannin St. MSB 2.112, Houston, TX 77030, USA
| | - Aaren Kettelhut
- The Ohio State University, 453 West 10 Ave, Columbus, OH 43210, USA
| | - Javier R. Lama
- Asociación Civil Impacta Salud y Educación, Av. Almte. Miguel Grau 1010, Lima 15063, Peru
| | - Sari L. Reisner
- Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
43
|
Dwivedi AK, Gornalusse GG, Siegel DA, Barbehenn A, Thanh C, Hoh R, Hobbs KS, Pan T, Gibson EA, Martin J, Hecht F, Pilcher C, Milush J, Busch MP, Stone M, Huang ML, Reppetti J, Vo PM, Levy CN, Roychoudhury P, Jerome KR, Hladik F, Henrich TJ, Deeks SG, Lee SA. A cohort-based study of host gene expression: tumor suppressor and innate immune/inflammatory pathways associated with the HIV reservoir size. PLoS Pathog 2023; 19:e1011114. [PMID: 38019897 PMCID: PMC10712869 DOI: 10.1371/journal.ppat.1011114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 12/11/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
The major barrier to an HIV cure is the HIV reservoir: latently-infected cells that persist despite effective antiretroviral therapy (ART). There have been few cohort-based studies evaluating host genomic or transcriptomic predictors of the HIV reservoir. We performed host RNA sequencing and HIV reservoir quantification (total DNA [tDNA], unspliced RNA [usRNA], intact DNA) from peripheral CD4+ T cells from 191 ART-suppressed people with HIV (PWH). After adjusting for nadir CD4+ count, timing of ART initiation, and genetic ancestry, we identified two host genes for which higher expression was significantly associated with smaller total DNA viral reservoir size, P3H3 and NBL1, both known tumor suppressor genes. We then identified 17 host genes for which lower expression was associated with higher residual transcription (HIV usRNA). These included novel associations with membrane channel (KCNJ2, GJB2), inflammasome (IL1A, CSF3, TNFAIP5, TNFAIP6, TNFAIP9, CXCL3, CXCL10), and innate immunity (TLR7) genes (FDR-adjusted q<0.05). Gene set enrichment analyses further identified significant associations of HIV usRNA with TLR4/microbial translocation (q = 0.006), IL-1/NRLP3 inflammasome (q = 0.008), and IL-10 (q = 0.037) signaling. Protein validation assays using ELISA and multiplex cytokine assays supported these observed inverse host gene correlations, with P3H3, IL-10, and TNF-α protein associations achieving statistical significance (p<0.05). Plasma IL-10 was also significantly inversely associated with HIV DNA (p = 0.016). HIV intact DNA was not associated with differential host gene expression, although this may have been due to a large number of undetectable values in our study. To our knowledge, this is the largest host transcriptomic study of the HIV reservoir. Our findings suggest that host gene expression may vary in response to the transcriptionally active reservoir and that changes in cellular proliferation genes may influence the size of the HIV reservoir. These findings add important data to the limited host genetic HIV reservoir studies to date.
Collapse
Affiliation(s)
- Ashok K. Dwivedi
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Germán G. Gornalusse
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - David A. Siegel
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Alton Barbehenn
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Cassandra Thanh
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, California, United States of America
| | - Rebecca Hoh
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Kristen S. Hobbs
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, California, United States of America
| | - Tony Pan
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, California, United States of America
| | - Erica A. Gibson
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, California, United States of America
| | - Jeffrey Martin
- Department of Biostatistics & Epidemiology, University of California San Francisco, California, United States of America
| | - Frederick Hecht
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Christopher Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, University of California San Francisco, California, United States of America
| | - Michael P. Busch
- Vitalant Blood Bank, San Francisco, California, United States of America
| | - Mars Stone
- Vitalant Blood Bank, San Francisco, California, United States of America
| | - Meei-Li Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Julieta Reppetti
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO- Houssay), Buenos Aires, Argentina
| | - Phuong M. Vo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Claire N. Levy
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Pavitra Roychoudhury
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Keith R. Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Florian Hladik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Timothy J. Henrich
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Steven G. Deeks
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| | - Sulggi A. Lee
- Department of Medicine, Division of HIV, Infectious Diseases & Global Medicine, University of California, San Francisco, California, United States of America
| |
Collapse
|
44
|
Guo X, Wang Z, Qu M, Guo Y, Yu M, Hong W, Zhang C, Fan X, Song J, Xu R, Zhang J, Huang H, Linghu E, Wang FS, Sun L, Jiao YM. Abnormal blood microbiota profiles are associated with inflammation and immune restoration in HIV/AIDS individuals. mSystems 2023; 8:e0046723. [PMID: 37698407 PMCID: PMC10654078 DOI: 10.1128/msystems.00467-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/22/2023] [Indexed: 09/13/2023] Open
Abstract
IMPORTANCE The characteristics of blood microbiota in HIV-infected individuals and their relevance to disease progression are still unknown, despite alterations in gut microbiota diversity and composition in HIV-infected individuals. Here, we present evidence of increased blood microbiota diversity in HIV-infected individuals, which may result from gut microbiota translocation. Also, we identify a group of microbes, Porphyromonas gingivalis, Prevotella sp. CAG:5226, Eubacterium sp. CAG:251, Phascolarctobacterium succinatutens, Anaerobutyricum hallii, Prevotella sp. AM34-19LB, and Phocaeicola plebeius, which are linked to poor immunological recovery. This work provides a scientific foundation toward therapeutic strategies targeting blood microbiota for immune recovery of HIV infection.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Zerui Wang
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Mengmeng Qu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yuntian Guo
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Minrui Yu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Weiguo Hong
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jinwen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruonan Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jiyuan Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Huihuang Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Enqiang Linghu
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Lijun Sun
- Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| |
Collapse
|
45
|
Taramasso L, Andreoni M, Antinori A, Bandera A, Bonfanti P, Bonora S, Borderi M, Castagna A, Cattelan AM, Celesia BM, Cicalini S, Cingolani A, Cossarizza A, D'Arminio Monforte A, D'Ettorre G, Di Biagio A, Di Giambenedetto S, Di Perri G, Esposito V, Focà E, Gervasoni C, Gori A, Gianotti N, Guaraldi G, Gulminetti R, Lo Caputo S, Madeddu G, Maggi P, Marandola G, Marchetti GC, Mastroianni CM, Mussini C, Perno CF, Rizzardini G, Rusconi S, Santoro M, Sarmati L, Zazzi M, Maggiolo F. Pillars of long-term antiretroviral therapy success. Pharmacol Res 2023; 196:106898. [PMID: 37648103 DOI: 10.1016/j.phrs.2023.106898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Meeting the challenge of antiretroviral therapy (ART) whose efficacy can last a lifetime requires continuous updating of the virological, pharmacological, and quality of life outcomes to be pursued and a continuous review of literature data on the efficacy and tolerability of new drugs and therapeutic strategies. METHODS With the aim of identifying open questions and answers about the current controversies in modern ART, we adapted the Design Thinking methodology to the needs of the design phase of a scientific article, involving a team of experts in HIV care. RESULTS Five main pillars of treatment success were discussed: sustained virologic suppression over time; immunological recovery; pharmacological attributes; long-term tolerability and safety of ART; and people's satisfaction and quality of life. The definition of the outcomes to be achieved in each thematic area and the tools to achieve them were reviewed and discussed. CONCLUSIONS Long-term treatment success should be intended as a combination of HIV-RNA suppression, immune recovery, and high quality of life. To achieve this, the regimen should be well-tolerated, with high potency, genetic barrier, and forgiveness, and should be tailored by a person-centered perspective, based on individual needs, preferences, and therapeutic history.
Collapse
Affiliation(s)
- Lucia Taramasso
- IRCCS Ospedale Policlinico San Martino di Genova, Genova, Italy.
| | | | - Andrea Antinori
- Istituto Nazionale per le Malattie Infettive Lazzaro Spallanzani IRCCS, Roma, Italy
| | - Alessandra Bandera
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | - Paolo Bonfanti
- Università degli Studi di Milano-Bicocca, Fondazione IRCCS San Gerardo dei Tintori - Monza, Monza, Italy
| | - Stefano Bonora
- Ospedale Amedeo di Savoia, Università degli Studi di Torino, Torino, Italy
| | - Marco Borderi
- Azienda Ospedaliero-Universitaria Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | | | | | - Stefania Cicalini
- Istituto Nazionale per le Malattie Infettive Lazzaro Spallanzani IRCCS, Roma, Italy
| | | | | | | | - Gabriella D'Ettorre
- Department of Public Health and Infectious Diseases AOU Policlinico Umberto I Sapienza, Rome, Italy
| | - Antonio Di Biagio
- Department of Health Sciences, Clinic of Infectious Diseases, University of Genoa, Genoa, Italy
| | | | - Giovanni Di Perri
- Ospedale Amedeo di Savoia, Università degli Studi di Torino, Torino, Italy
| | - Vincenzo Esposito
- UOC di Malattie infettive e Medicina di Genere P.O. Cotugno-A.O. dei Colli, Napoli, Italy
| | - Emanuele Focà
- Università degli Studi di Brescia e ASST Spedali Civili di Brescia, Brescia, Italy
| | | | - Andrea Gori
- Università degli Studi di Milano, Milano, Italy; ASST Fatebenefratelli Sacco, Ospedale Luigi Sacco, Milano, Italy
| | | | - Giovanni Guaraldi
- Azienda Ospedaliero-Universitaria Policlinico di Modena, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | | | | | - Giordano Madeddu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Paolo Maggi
- Università degli Studi della Campania Luigi Vanvitelli, AORN S. Anna e S. Sebastiano Caserta, Caserta, Italy
| | | | - Giulia Carla Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | | | - Cristina Mussini
- Azienda Ospedaliero-Universitaria Policlinico di Modena, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | | | | | - Stefano Rusconi
- Ospedale Civile di Legnano ASST Ovest Milanese - Università degli Studi di Milano, Legnano, Italy
| | - Maria Santoro
- Università degli Studi di Roma "Tor Vergata", Roma, Italy
| | | | | | | |
Collapse
|
46
|
Herbert NG, Goulder PJR. Impact of early antiretroviral therapy, early life immunity and immune sex differences on HIV disease and posttreatment control in children. Curr Opin HIV AIDS 2023; 18:229-236. [PMID: 37421384 PMCID: PMC10399946 DOI: 10.1097/coh.0000000000000807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
PURPOSE OF REVIEW To review recent insights into the factors affecting HIV disease progression in children living with HIV, contrasting outcomes: following early ART initiation with those in natural, antiretroviral therapy (ART)-naive infection; in children versus adults; and in female individuals versus male individuals. RECENT FINDINGS Early life immune polarization and several factors associated with mother-to-child transmission of HIV result in an ineffective HIV-specific CD8+ T-cell response and rapid disease progression in most children living with HIV. However, the same factors result in low immune activation and antiviral efficacy mediated mainly through natural killer cell responses in children and are central features of posttreatment control. By contrast, rapid activation of the immune system and generation of a broad HIV-specific CD8+ T-cell response in adults, especially in the context of 'protective' HLA class I molecules, are associated with superior disease outcomes in ART-naive infection but not with posttreatment control. The higher levels of immune activation in female individuals versus male individuals from intrauterine life onwards increase HIV infection susceptibility in females in utero and may favour ART-naive disease outcomes rather than posttreatment control. SUMMARY Early-life immunity and factors associated with mother-to-child transmission typically result in rapid HIV disease progression in ART-naive infection but favour posttreatment control in children following early ART initiation.
Collapse
Affiliation(s)
- Nicholas G Herbert
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
47
|
Shive CL, Kowal CM, Desotelle AF, Nguyen Y, Carbone S, Kostadinova L, Davitkov P, O’Mara M, Reihs A, Siddiqui H, Wilson BM, Anthony DD. Endotoxemia Associated with Liver Disease Correlates with Systemic Inflammation and T Cell Exhaustion in Hepatitis C Virus Infection. Cells 2023; 12:2034. [PMID: 37626844 PMCID: PMC10453378 DOI: 10.3390/cells12162034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/29/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Both acute and chronic hepatitis C virus (HCV) infections are characterized by inflammation. HCV and reduced liver blood filtration contribute to inflammation; however, the mechanisms of systemic immune activation and dysfunction as a result of HCV infection are not clear. We measured circulating inflammatory mediators (IL-6, IP10, sCD163, sCD14), indices of endotoxemia (EndoCab, LBP, FABP), and T cell markers of exhaustion and senescence (PD-1, TIGIT, CD57, KLRG-1) in HCV-infected participants, and followed a small cohort after direct-acting anti-viral therapy. IL-6, IP10, Endocab, LBP, and FABP were elevated in HCV participants, as were T cell co-expression of exhaustion and senescence markers. We found positive associations between IL-6, IP10, EndoCab, LBP, and co-expression of T cell markers of exhaustion and senescence. We also found numerous associations between reduced liver function, as measured by plasma albumin levels, and T cell exhaustion/senescence, inflammation, and endotoxemia. We found positive associations between liver stiffness (TE score) and plasma levels of IL-6, IP10, and LBP. Lastly, plasma IP10 and the proportion of CD8 T cells co-expressing PD-1 and CD57 decreased after initiation of direct-acting anti-viral therapy. Although associations do not prove causality, our results support the model that translocation of microbial products, resulting from decreased liver blood filtration, during HCV infection drives chronic inflammation that results in T cell exhaustion/senescence and contributes to systemic immune dysfunction.
Collapse
Affiliation(s)
- Carey L. Shive
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
- Pathology Department, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Corinne M. Kowal
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Alexandra F. Desotelle
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Ynez Nguyen
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Sarah Carbone
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Lenche Kostadinova
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Perica Davitkov
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Megan O’Mara
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Alexandra Reihs
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Hinnah Siddiqui
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
| | - Brigid M. Wilson
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Donald D. Anthony
- Cleveland VA Medical Center, Cleveland, OH 44106, USA; (C.M.K.); (A.F.D.); (Y.N.); (S.C.); (L.K.); (P.D.); (M.O.); (A.R.); (H.S.); (B.M.W.); (D.D.A.)
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
48
|
Finnegan D, Tocmo R, Loscher C. Targeted Application of Functional Foods as Immune Fitness Boosters in the Defense against Viral Infection. Nutrients 2023; 15:3371. [PMID: 37571308 PMCID: PMC10421353 DOI: 10.3390/nu15153371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
In recent times, the emergence of viral infections, including the SARS-CoV-2 virus, the monkeypox virus, and, most recently, the Langya virus, has highlighted the devastating effects of viral infection on human life. There has been significant progress in the development of efficacious vaccines for the prevention and control of viruses; however, the high rates of viral mutation and transmission necessitate the need for novel methods of control, management, and prevention. In recent years, there has been a shift in public awareness on health and wellbeing, with consumers making significant dietary changes to improve their immunity and overall health. This rising health awareness is driving a global increase in the consumption of functional foods. This review delves into the benefits of functional foods as potential natural means to modulate the host immune system to enhance defense against viral infections. We provide an overview of the functional food market in Europe and discuss the benefits of enhancing immune fitness in high-risk groups, including the elderly, those with obesity, and people with underlying chronic conditions. We also discuss the immunomodulatory mechanisms of key functional foods, including dairy proteins and hydrolysates, plant-based functional foods, fermentates, and foods enriched with vitamin D, zinc, and selenium. Our findings reveal four key immunity boosting mechanisms by functional foods, including inhibition of viral proliferation and binding to host cells, modulation of the innate immune response in macrophages and dendritic cells, enhancement of specific immune responses in T cells and B cells, and promotion of the intestinal barrier function. Overall, this review demonstrates that diet-derived nutrients and functional foods show immense potential to boost viral immunity in high-risk individuals and can be an important approach to improving overall immune health.
Collapse
Affiliation(s)
| | | | - Christine Loscher
- School of Biotechnology, Dublin City University, D09 DX63 Dublin, Ireland; (D.F.); (R.T.)
| |
Collapse
|
49
|
Funderburg NT, Shive CL, Chen Z, Tatsuoka C, Bowman ER, Longenecker CT, McComsey GA, Clagett BM, Dorazio D, Freeman ML, Sieg SF, Moisi D, Anthony DD, Jacobson JM, Stein SL, Calabrese LH, Landay A, Flexner C, Crawford KW, Capparelli EV, Rodriguez B, Lederman MM. Interleukin 6 Blockade With Tocilizumab Diminishes Indices of Inflammation That Are Linked to Mortality in Treated Human Immunodeficiency Virus Infection. Clin Infect Dis 2023; 77:272-279. [PMID: 37011013 PMCID: PMC10371305 DOI: 10.1093/cid/ciad199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/23/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND People with human immunodeficiency virus (PWH) are at increased risk for comorbidities, and plasma interleukin 6 (IL-6) levels are among the most robust predictors of these outcomes. Tocilizumab (TCZ) blocks the receptor for IL-6, inhibiting functions of this cytokine. METHODS This was a 40-week, placebo-controlled, crossover trial (NCT02049437) where PWH on stable antiretroviral therapy (ART) were randomized to receive 3 monthly doses of TCZ or matching placebo intravenously. Following a 10-week treatment period and a 12-week washout, participants were switched to the opposite treatment. The primary endpoints were safety and posttreatment levels of C-reactive protein (CRP) and CD4+ T-cell cycling. Secondary endpoints included changes in inflammatory indices and lipid levels. RESULTS There were 9 treatment-related toxicities of grade 2 or greater during TCZ administration (mostly neutropenia) and 2 during placebo administration. Thirty-one of 34 participants completed the study and were included in a modified intent-to-treat analysis. TCZ reduced levels of CRP (median decrease, 1819.9 ng/mL, P < .0001; effect size, 0.87) and reduced inflammatory markers in PWH, including D-dimer, soluble CD14, and tumor necrosis factor receptors. T-cell cycling tended to decrease in all maturation subsets after TCZ administration, but was only significant among naive CD4 T cells. Lipid levels, including lipid classes that have been related to cardiovascular disease risk, increased during TCZ treatment. CONCLUSIONS TCZ is safe and decreases inflammation in PWH; IL-6 is a key driver of the inflammatory environment that predicts morbidity and mortality in ART-treated PWH. The clinical significance of lipid elevations during TCZ treatment requires further study. Clinical Trials Registration. NCT02049437.
Collapse
Affiliation(s)
- Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio, USA
| | - Carey L Shive
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Zhengyi Chen
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Curtis Tatsuoka
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emily R Bowman
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio, USA
| | - Chris T Longenecker
- Department of Medicine and Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Grace A McComsey
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Brian M Clagett
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Dominic Dorazio
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Michael L Freeman
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Scott F Sieg
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Daniela Moisi
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Donald D Anthony
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
- Rheumatology Section, MetroHealth Medical Center, Cleveland, Ohio, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Sharon L Stein
- Department of Surgery, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | | | - Alan Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Keith W Crawford
- Therapeutic Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Edmund V Capparelli
- Clinical Pediatrics and Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Benigno Rodriguez
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Michael M Lederman
- Department of Medicine, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
50
|
Mboumba Bouassa RS, Comeau E, Alexandrova Y, Pagliuzza A, Yero A, Samarani S, Needham J, Singer J, Lee T, Bobeuf F, Vertzagias C, Sebastiani G, Margolese S, Mandarino E, Klein MB, Lebouché B, Routy JP, Chomont N, Costiniuk CT, Jenabian MA. Effects of Oral Cannabinoids on Systemic Inflammation and Viral Reservoir Markers in People with HIV on Antiretroviral Therapy: Results of the CTN PT028 Pilot Clinical Trial. Cells 2023; 12:1811. [PMID: 37508476 PMCID: PMC10378564 DOI: 10.3390/cells12141811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic HIV infection is characterized by persistent inflammation despite antiretroviral therapy (ART). Cannabinoids may help reduce systemic inflammation in people with HIV (PWH). To assess the effects of oral cannabinoids during HIV, ten PWH on ART were randomized (n = 5/group) to increasing doses of oral Δ9-tetrahydrocannabinol (THC): cannabidiol (CBD) combination (2.5:2.5-15:15 mg/day) capsules or CBD-only (200-800 mg/day) capsules for 12 weeks. Blood specimens were collected prospectively 7-21 days prior to treatment initiation and at weeks 0 to 14. Plasma cytokine levels were determined via Luminex and ELISA. Immune cell subsets were characterized by flow cytometry. HIV DNA/RNA were measured in circulating CD4 T-cells and sperm by ultra-sensitive qPCR. Results from both arms were combined for statistical analysis. Plasma levels of IFN-γ, IL-1β, sTNFRII, and REG-3α were significantly reduced at the end of treatment (p ˂ 0.05). A significant decrease in frequencies of PD1+ memory CD4 T-cells, CD73+ regulatory CD4 T-cells, and M-DC8+ intermediate monocytes was also observed (p ˂ 0.05), along with a transient decrease in CD28-CD57+ senescent CD4 and CD8 T-cells. Ki-67+ CD4 T-cells, CCR2+ non-classical monocytes, and myeloid dendritic cells increased over time (p ˂ 0.05). There were no significant changes in other inflammatory markers or HIV DNA/RNA levels. These findings can guide future large clinical trials investigating cannabinoid anti-inflammatory properties.
Collapse
Affiliation(s)
- Ralph-Sydney Mboumba Bouassa
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Eve Comeau
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Yulia Alexandrova
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Amélie Pagliuzza
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Suzanne Samarani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Judy Needham
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Joel Singer
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Terry Lee
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Florian Bobeuf
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Claude Vertzagias
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Giada Sebastiani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Shari Margolese
- CIHR Canadian HIV Trials Network, Vancouver, BC V6Z 1Y6, Canada
| | | | - Marina B Klein
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Bertrand Lebouché
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
- Centre for Outcomes Research & Evaluation, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Cecilia T Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|