1
|
Takyi A, Soma A, Przybylska M, Harriss E, Barnes KI, Dahal P, Guérin PJ, Stepniewska K, Carrara VI. Efficacy of artemisinin-based combination therapy (ACT) in people living with HIV (PLHIV) diagnosed with uncomplicated Plasmodium falciparum malaria in Africa: a WWARN systematic review. Malar J 2025; 24:153. [PMID: 40380136 DOI: 10.1186/s12936-025-05393-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 04/29/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Africa bears the highest double burden of HIV and malaria worldwide. In 2023, an estimated 25.9 million people were living with HIV (PLHIV), and 246 million malaria cases were diagnosed in Africa. Malaria patients co-infected with HIV are considered at a higher risk of failing malaria treatment, according to the World Health Organization (WHO) guidelines. This systematic literature review aims to assess the treatment outcomes following artemisinin-based combination therapy (ACT) in PLHIV. METHODS The literature search was conducted up to April 2022 in the following databases: MEDLINE, EMBASE, Web of Science, Cochrane Central, WHO Global Index Medicus, Clinicaltrials.gov, and the WorldWide Antimalarial Resistance Network (WWARN) Clinical Trial Library. Studies describing any malaria treatment outcomes or anti-malarial drug exposure in PLHIV treated for uncomplicated Plasmodium falciparum malaria infection were eligible for inclusion. RESULTS A total of 26 articles describing 19 studies conducted between 2003 and 2017 in six countries were included in this review; it represented 2850 malaria episodes in PLHIV across various transmission settings. The most studied artemisinin-based combination was artemether-lumefantrine (in 16 studies). PLHIV were treated with various antiretroviral therapy (ART) regimens, namely efavirenz (EFV), nevirapine (NVP), atazanavir-ritonavir (ATVr), lopinavir-ritonavir (LPV/r), and/or on prophylaxis with trimethoprim-sulfamethoxazole (TS), or were untreated (in 3 studies). There was no evidence of an increased risk of recrudescence in PLHIV compared to those without HIV. When treated with artemether-lumefantrine, PLHIV receiving LPV/r had a lower risk of malaria recurrence compared to PLHIV on NVP-based or EFV-based ART, or those without HIV. LPV/r increased lumefantrine exposure and EFV-treated patients had a reduced exposure to both artemether and lumefantrine; NVP reduced artemether exposure only. CONCLUSIONS Limited data on ACT outcomes or drug exposure in PLHIV in Africa remains a reality to date, and the effect of antivirals appears inconsistent in the literature. Considering the heterogeneity in study designs, these review's findings support conducting an individual patient data meta-analysis to explore the impact of antiretroviral therapy on anti-malarial treatment. TRIAL REGISTRATION The protocol for the original search was published on PROSPERO with registration number CRD42018089860.
Collapse
Affiliation(s)
- Abena Takyi
- Department of Child Health, Korle Bu Teaching Hospital, Accra, Ghana
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Aboubakar Soma
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Centre MURAZ/Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
- Département Médecine-Pharmacopée Traditionnelle/Pharmacie (MEPHATRA/Ph), Institut de Recherche en Sciences de la Santé (IRSS), Ouagadougou, Burkina Faso
| | | | - Eli Harriss
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Karen I Barnes
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Prabin Dahal
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Philippe J Guérin
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Kasia Stepniewska
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
| | - Verena I Carrara
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Infectious Diseases Data Observatory (IDDO), Oxford, UK.
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.
- Institute of Global Health, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
2
|
Li X, Sun L, He Y, Zhao F, Luo Y, Liu C, Hu Y, Jiang Y, Lu H, Liu J. HDL-C as a novel predictor of immune reconstitution in people living with HIV: insights from a baseline-to-dynamic change cohort study in China, 2005-2022. Front Immunol 2025; 16:1520615. [PMID: 40421011 PMCID: PMC12104299 DOI: 10.3389/fimmu.2025.1520615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
Background High-density lipoprotein cholesterol (HDL-C) is a well-established marker of lipid metabolism and increasingly recognized as an indicator of inflammatory status. This study investigates HDL-C's role as a predictor of immune reconstitution in people living with HIV (PLWH). Methods We performed a prospective cohort study of 15,434 PLWH initiating antiretroviral therapy at the Third People's Hospital of Shenzhen, China, between 2005 and 2022. Baseline quartile grouping and Group-Based Trajectory Modeling (GBTM) explored the relationship between HDL-C and immune reconstitution. Restricted cubic spline plots identified nonlinear association. Results Over a median follow-up of 17.9 months, 9,609 PLWH achieved the CD4+ T-cell count of 500 cells/μL or higher. Multivariate-adjusted Cox proportional hazards regression model showed that hazard ratios (HR) [95% confidence intervals (CI)] for CD4+ T-cell recovery in Q2, Q3, and Q4 versus Q1 of the HDL-C were 0.94 (95% CI: 0.88-0.99), 0.92 (95% CI: 0.87-0.98), and 0.85 (95% CI: 0.80-0.91), respectively. GBTM identified two HDL-C trajectories: Low-floating and High-floating. Relative to Low-floating, High-floating demonstrated a reduced likelihood of CD4+ T-cell recovery (HR=0.86, 95% CI: 0.82-0.90, p < 0.001). A nonlinear association was observed between HDL-C and the outcome (p for nonlinear association = 0.037, p for overall < 0.001), with a threshold at 1.13 mmol/L. Negative correlations between HDL-C and CD4+ T-cell recovery were observed both below the threshold (HR=0.72, 95% CI: 0.57-0.92) and above the threshold (HR=0.78, 95% CI: 0.69-0.87) (both p < 0.05). Conclusions Our study highlights HDL-C's role in immune recovery, suggesting its potential in guiding prevention and treatment strategies for PLWH.
Collapse
Affiliation(s)
- Xiaorui Li
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| | - Liqin Sun
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Yun He
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Fang Zhao
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Yinsong Luo
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| | - Chenye Liu
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| | - Yiyao Hu
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| | - Yuxin Jiang
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| | - Hongzhou Lu
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Jiaye Liu
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
3
|
Liu X, Zhang L, Li X, Chen L, Lu L, Yang Y, Wu Y, Zheng L, Tang J, Wang F, Han Y, Song X, Cao W, Li T. Single-cell multi-omics profiling uncovers the immune heterogeneity in HIV-infected immunological non-responders. EBioMedicine 2025; 115:105667. [PMID: 40184908 PMCID: PMC12002939 DOI: 10.1016/j.ebiom.2025.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Immunological non-responders (INRs) are people living with HIV-1 who fail to achieve full immune reconstitution despite long-term effective antiretroviral therapy (ART). This incomplete recovery of CD4+ T cells increase the risk of opportunistic infections and non-AIDS-related morbidity and mortality. Understanding the mechanisms driving this immune dysfunction is critical for developing targeted therapies. METHODS We performed single-cell RNA sequencing (scRNA-seq) and single-cell VDJ sequencing (scVDJ-seq) on peripheral blood mononuclear cells (PBMCs) from INRs, immune responders (IRs), and healthy controls (HCs). We developed scGeneANOVA, a novel mixed model differential gene analysis tool, to detect differentially expressed genes and pathways. In addition, we developed the Viral Identification and Load Detection Analysis (VILDA) tool to quantify HIV-1 transcripts and investigate their relationship with interferon (IFN) pathway activation. FINDINGS Our analysis revealed that INRs exhibit a dysregulated IFN response, closely associated with CD4+ T cell exhaustion and immune recovery failure. The scGeneANOVA tool identified critical genes and pathways that were missed by traditional analysis methods, while VILDA showed higher levels of HIV-1 transcripts in INRs, which may drive the heightened IFN response. These findings support a potential contribution of IFN signalling in INR-related immune dysfunction. INTERPRETATION Our study provides new insights into the pathogenic mechanisms behind immune recovery failure in INRs, suggesting that IFN signalling might be involved in the development of CD4+ T cell exhaustion. The identification of key genes and pathways offers potential biomarkers and therapeutic targets for improving immune recovery in this vulnerable population. FUNDING This study was supported by the grants from Special Research Fund for the Central High-level Hospitals of Peking Union Medical College Hospital (Grant No. 2022-PUMCH-D-008), Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (Grant No. 2021-I2M-1-037), National Key Technologies R&D Program for the 13th Five-year Plan (Grant No. 2017ZX10202101-001). The funders played no role in the design, experiment conduction, data analysis and preparation of the manuscript of this work.
Collapse
Affiliation(s)
- Xiaosheng Liu
- School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China; Centre for Life Sciences, Tsinghua University, 100084, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Leidan Zhang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Ling Chen
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yang Yang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yuanni Wu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Liyuan Zheng
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Jia Tang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Fada Wang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China; State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, 100730, Beijing, China; Center for AIDS Research, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Taisheng Li
- School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China; Centre for Life Sciences, Tsinghua University, 100084, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China; State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, 100730, Beijing, China; Center for AIDS Research, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
4
|
Sun J, Yang M, Su G, Wang L, Hu X, Zhou Y, Cui G, Qian G, Yuan Y, Hu X, Li S, Luo H, Zhang S, Li G, Zhang D, Li G, Cheng M, Yu Z, Ren Z. The Antiviral Efficacy and Safety of Azvudine in Hospitalized SARS-CoV-2 Infected Patients with Liver Diseases Based on a Multicenter, Retrospective Cohort Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405679. [PMID: 39985372 PMCID: PMC12005779 DOI: 10.1002/advs.202405679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/31/2024] [Indexed: 02/24/2025]
Abstract
Despite azvudine being prioritized for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, its effectiveness and safety remain inadequately substantiated in hospitalized SARS-CoV-2 infected patients with liver diseases. A retrospective nine-center cohort study along with an independent validation cohort is conducted to examine the efficacy of azvudine (Clinical Trial Registration Number: NCT06349655). The primary outcome is all-cause mortality and the secondary outcome is composite disease progression. Efficacy is assessed via Kaplan-Meier analysis and Cox regression, with subgroup and sensitivity analyses for further validation. Among 32 864 hospitalized SARS-CoV-2 infected patients, 1022 eligible azvudine recipients, and 1022 controls are included through propensity score match. Kaplan-Meier analysis reveals that azvudine treatment is associated with a lower risk of all-cause mortality and composite disease progression (both p<0.0001). Cox regression analysis suggests azvudine recipients could have a 39% lower risk of all-cause mortality than controls (95% confidence interval [CI]: 0.468-0.795, p<0.001), but with no notable significance in composite disease progression (hazard ratio: 0.85, 95% CI: 0.686-1.061, p = 0.154). Subgroup analysis suggests that azvudine has a greater benefit for both all-cause mortality and composite disease progression in patients with kidney diseases or without autoimmune diseases. Three sensitivity analyses and validation cohorts confirm the robustness of the findings. Safety analysis observes few adverse events in azvudine recipients. Within 15 days after azvudine administration, no significant difference in liver function indexes and kidney function indexes is observed between the two groups except for a few time points. These findings demonstrate that azvudine shows potential clinical efficacy in improving all-cause mortality in hospitalized SARS-CoV-2 infected patients with liver diseases, with acceptable adverse effects.
Collapse
Affiliation(s)
- Junyi Sun
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Mengzhao Yang
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Guanyue Su
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Ling Wang
- Department of Cardiovascular MedicineHenan Provincial Chest Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Xiaobo Hu
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Yongjian Zhou
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Guangying Cui
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Guowu Qian
- Department of Gastrointestinal SurgeryNanyang Central HospitalNanyang473009China
| | - Yiqiang Yuan
- Department of Cardiovascular MedicineHenan Provincial Chest Hospital Affiliated to Zhengzhou UniversityZhengzhou450008China
| | - Xinjun Hu
- Department of Infectious DiseasesThe First Affiliated HospitalCollege of Clinical MedicineHenan University of Science and TechnologyLuoyang471003China
| | - Silin Li
- Department of Respiratory and Critical Care MedicineFengqiu County People's HospitalXinxiang453300China
| | - Hong Luo
- Guangshan County People's HospitalGuangshan CountyXinyang465450China
| | - Shixi Zhang
- Department of Infectious DiseasesShangqiu Municipal HospitalShangqiu476000China
| | - Guangming Li
- Department of Liver DiseaseThe Affiliated Infectious Disease Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Donghua Zhang
- Department of Infectious DiseasesAnyang City Fifth People's HospitalAnyang455000China
| | - Guotao Li
- Department of Infectious DiseasesLuoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang471000China
| | - Ming Cheng
- Department of Medical InformationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Zujiang Yu
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Zhigang Ren
- Department of Infectious DiseasesState Key Laboratory of Antiviral DrugsPingyuan LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| |
Collapse
|
5
|
Chéret A. Acute HIV-1 Infection: Paradigm and Singularity. Viruses 2025; 17:366. [PMID: 40143294 PMCID: PMC11945883 DOI: 10.3390/v17030366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Acute HIV-1 infection (AHI) is a transient period where the virus causes evident damage to the immune system, including an extensive apoptosis of CD4+ T cells associated with a high level of activation and a major cytokine storm to fight the invading virus. HIV infection establishes persistence by integrating the viral genome into host cell DNA in both replicating and non-replicating forms, effectively hiding from immune surveillance within infected lymphocytes as cellular reservoirs. The measurement of total HIV-1 DNA in peripheral blood mononuclear cells (PBMCs) is a reliable reflection of this reservoir. Initiating treatments during AHI with nucleoside reverse transcriptase inhibitors (NRTIs) and/or integrase strand transfer inhibitors (INSTIs) is essential to alter the dynamics of the global reservoir expansion, and to reduce the establishment of long-lived cellular and tissue reservoirs, while preserving and enhancing specific and non-specific immune responses. Furthermore, some of the patients treated at the AHI stage may become post-treatment controllers and should be informative regarding the mechanism of viral control, so patients treated during AHI are undoubtedly the best candidates to test innovative remission strategies toward a functional cure that could play a pivotal role in long-term HIV control. AHI is characterized by high levels of viral replication, with a significant increase in the risk of HIV transmission. Detecting AHI and initiating early treatment following diagnosis provides a window of opportunity to control the epidemic, particularly in high-risk populations.
Collapse
Affiliation(s)
- Antoine Chéret
- Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris Descartes, 75014 Paris, France;
- Service Plateforme de Diagnostic et Thérapeutique Pluridisciplinaire, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
- INSERM-CIC-1424, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
| |
Collapse
|
6
|
Nabipur L, Mouawad M, Venketaraman V. Additive Effects of Glutathione in Improving Antibiotic Efficacy in HIV- M.tb Co-Infection in the Central Nervous System: A Systematic Review. Viruses 2025; 17:127. [PMID: 39861915 PMCID: PMC11769047 DOI: 10.3390/v17010127] [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: 12/31/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND HIV and tuberculosis (TB) co-infection poses a significant health challenge, particularly when involving the central nervous system (CNS), where it leads to severe morbidity and mortality. Current treatments face challenges such as drug resistance, immune reconstitution inflammatory syndrome (IRIS), and persistent inflammation. Glutathione (GSH) has the therapeutic potential to enhance treatment outcomes by improving antibiotic efficacy, reducing inflammation, and mitigating immune dysfunction. METHODS Relevant studies were identified through systematic searches of PubMed, Elsevier, WHO, and related databases. Inclusion criteria focused on preclinical and clinical research examining GSH or its precursors in HIV, TB, or co-infection, with emphasis on microbial control, immune modulation, and CNS-related outcomes. RESULTS Preclinical studies showed that GSH improves macrophage antimicrobial function, reduces oxidative stress, and limits Mycobacterium tuberculosis (M.tb) growth. Animal models demonstrated reduced bacterial burden in the lungs, liver, and spleen with GSH supplementation, along with enhanced granuloma stability. Clinical studies highlighted increased TH1 cytokine production, reduced inflammatory markers, and improved CD4+ T cell counts in HIV-M.tb co-infected patients. N-acetylcysteine (NAC), a GSH precursor, was shown to significantly enhance the efficacy of first-line TB antibiotics and mitigate treatment-associated toxicity. DISCUSSION GSH shows promise as an adjunct therapy for HIV-M.tb co-infection, particularly for cases involving the CNS, where it may improve immune recovery and reduce inflammation. However, evidence is limited by small sample sizes and a lack of randomized trials. Future research should focus on developing CNS-directed GSH formulations and evaluating its integration into current treatment protocols to address the dual burden of HIV and TB, ultimately improving patient outcomes.
Collapse
Affiliation(s)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (L.N.); (M.M.)
| |
Collapse
|
7
|
Corbett C, van Rensburg R, Brey N, O'Hagan S, Esterhuizen TM, Chow FC, Decloedt EH. Timing of ART Initiation Associated With HIV-Associated Stroke. J Acquir Immune Defic Syndr 2025; 98:e1-e3. [PMID: 39288941 DOI: 10.1097/qai.0000000000003529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Affiliation(s)
- Craig Corbett
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
- Department of Medicine, Paarl Hospital, Paarl, Western Cape, South Africa
| | - Roland van Rensburg
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Naeem Brey
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Suzanne O'Hagan
- Division of Radiodiagnosis, Department of Medical Imaging and Clinical Oncology, Cape Town, Western Cape, South Africa
| | - Tonya M Esterhuizen
- Biostatistics Unit, Division of Epidemiology and Biostatistics, Cape Town, Western Cape, South Africa
| | - Felicia C Chow
- Departments of Neurology and Medicine (Infectious Diseases), University of California San Francisco, San Francisco, CA
| | - Eric H Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
8
|
Cui X, Yi Y, Lin Y, Zhu N, Li X. Clinical efficacy and safety of new compound single tablet antiviral drugs in the treatment of HIV/AIDS. Life Sci 2024; 358:123117. [PMID: 39424269 DOI: 10.1016/j.lfs.2024.123117] [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: 02/23/2024] [Revised: 06/06/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
AIMS Genvoya, Biktarvy and Dovato are novel single-tablet antiretroviral therapy(ART). The aim of this study is to explore the therapeutic effects of these novel drugs on HIV/AIDS. MAIN METHODS This retrospective cohort study, conducted at a single center, included a total of 200 HIV-treated patients who transitioned to these new antiretroviral drugs from July 2021 to August 2023. Data were extracted from electronic medical records at Ditan Hospital. The Genvoya group comprised 22 patients, and all subsequent switches in this group were to Biktarvy. The primary HAART group consisted of 178 patients initially treated with a first-line triple Highly Active Antiretroviral Therapy (HAART) regimen during the same period. This group was further subdivided into HAART+Dovato, HAART+Biktarvy, and HAART+Genvoya groups based on the switching regimen. The primary outcomes focused on changes in viral load and immune efficacy, while secondary safety indicators included blood/liver function, lipid parameters, renal function, blood glucose, blood uric acid, etc. KEY FINDINGS: The viral suppression rate was 100 % after the drug change treatment, and CD4+ T cell counts increased significantly across all four groups. Over the 6-month treatment period, there were increases in creatinine (Cr), low-density lipoprotein (LDL), high-density lipoprotein (HDL), erythrocyte count, and glomerular filtration rate (eGFR). Conversely, Alanine transaminase (ALT), Aspartate aminotransferase (AST), C-reactive protein (CRP), albumin (ALB), and blood glucose (Glu) levels decreased. SIGNIFICANCE Genvoya, Biktarvy and Dovato are recommended for the treatment of HIV/AIDS and have a good safety profile.
Collapse
Affiliation(s)
- Xinyu Cui
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yunyun Yi
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yingying Lin
- Department of Center of Integrated Traditional Chinese and Western Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Na Zhu
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin Li
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
9
|
Nelson M, Bracchi M, Hunter E, Ong E, Pozniak A, van Halsema C. British HIV Association guidelines on the management of opportunistic infection in people living with HIV: The clinical management of non-tuberculous mycobacteria 2024. HIV Med 2024; 25 Suppl 4:3-25. [PMID: 39822028 DOI: 10.1111/hiv.13727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 01/19/2025]
Affiliation(s)
- M Nelson
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
- Imperial College, London, UK
| | - M Bracchi
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - E Hunter
- The Newcastle-upon-Tyne Hospitals NHS Foundation Trust, UK
| | - E Ong
- The Newcastle-upon-Tyne Hospitals NHS Foundation Trust, UK
- Newcastle University Medicine Malaysia, Johor, Malaysia
| | - A Pozniak
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
- London School of Hygiene and Tropical Medicine, UK
| | | |
Collapse
|
10
|
Miedema J, Cinetto F, Smed-Sörensen A, Spagnolo P. The immunopathogenesis of sarcoidosis. J Autoimmun 2024; 149:103247. [PMID: 38734536 DOI: 10.1016/j.jaut.2024.103247] [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: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Sarcoidosis is a granulomatous multiorgan disease, thought to result from exposure to yet unidentified antigens in genetically susceptible individuals. The exaggerated inflammatory response that leads to granuloma formation is highly complex and involves the innate and adaptive immune system. Consecutive immunological studies using advanced technology have increased our understanding of aberrantly activated immune cells, mediators and pathways that influence the formation, maintenance and resolution of granulomas. Over the years, it has become increasingly clear that disease immunopathogenesis can only be understood if the clinical heterogeneity of sarcoidosis is taken into consideration, along with the distribution of immune cells in peripheral blood and involved organs. Most studies offer an immunological snapshot during disease course, while the cellular composition of both the circulation and tissue microenvironment may change over time. Despite these challenges, novel insights on the role of the immune system are continuously published, thus bringing the field forward. This review highlights current knowledge on the innate and adaptive immune responses involved in sarcoidosis pathogenesis, as well as the pathways involved in non-resolving disease and fibrosis development. Additionally, we describe proposed immunological mechanisms responsible for drug-induced sarcoid like reactions. Although many aspects of disease immunopathogenesis remain to be unraveled, the identification of crucial immune reactions in sarcoidosis may help identify new treatment targets. We therefore also discuss potential therapies and future strategies based on the latest immunological findings.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonary Medicine, Center of Expertise for Interstitial Lung Disease, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Italy; Department of Medicine - DIMED, University of Padova, Padova, Italy.
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| |
Collapse
|
11
|
Grant-McAuley W, Morgenlander WR, Ruczinski I, Kammers K, Laeyendecker O, Hudelson SE, Thakar M, Piwowar-Manning E, Clarke W, Breaud A, Ayles H, Bock P, Moore A, Kosloff B, Shanaube K, Meehan SA, van Deventer A, Fidler S, Hayes R, Larman HB, Eshleman SH, for the HPTN 071 (PopART) Study Team. Identification of antibody targets associated with lower HIV viral load and viremic control. PLoS One 2024; 19:e0305976. [PMID: 39288118 PMCID: PMC11407625 DOI: 10.1371/journal.pone.0305976] [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: 02/06/2024] [Accepted: 06/09/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND High HIV viral loads (VL) are associated with increased morbidity, mortality, and on-going transmission. HIV controllers maintain low VLs in the absence of antiretroviral therapy (ART). We previously used a massively multiplexed antibody profiling assay (VirScan) to compare antibody profiles in HIV controllers and persons living with HIV (PWH) who were virally suppressed on ART. In this report, we used VirScan to evaluate whether antibody reactivity to specific HIV targets and broad reactivity across the HIV genome was associated with VL and controller status 1-2 years after infection. METHODS Samples were obtained from participants who acquired HIV infection in a community-randomized trial in Africa that evaluated an integrated strategy for HIV prevention (HPTN 071 PopART). Controller status was determined using VL and antiretroviral (ARV) drug data obtained at the seroconversion visit and 1 year later. Viremic controllers had VLs <2,000 copies/mL at both visits; non-controllers had VLs >2,000 copies/mL at both visits. Both groups had no ARV drugs detected at either visit. VirScan testing was performed at the second HIV-positive visit (1-2 years after HIV infection). RESULTS The study cohort included 13 viremic controllers and 64 non-controllers. We identified ten clusters of homologous peptides that had high levels of antibody reactivity (three in gag, three in env, two in integrase, one in protease, and one in vpu). Reactivity to 43 peptides (eight unique epitopes) in six of these clusters was associated with lower VL; reactivity to six of the eight epitopes was associated with HIV controller status. Higher aggregate antibody reactivity across the eight epitopes (more epitopes targeted, higher mean reactivity across all epitopes) and across the HIV genome was also associated with lower VL and controller status. CONCLUSIONS We identified HIV antibody targets associated with lower VL and HIV controller status 1-2 years after infection. Robust aggregate responses to these targets and broad antibody reactivity across the HIV genome were also associated with lower VL and controller status. These findings provide novel insights into the relationship between humoral immunity and viral containment that could help inform the design of antibody-based approaches for reducing HIV VL.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William R. Morgenlander
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Kai Kammers
- Quantitative Sciences Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Manjusha Thakar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Helen Ayles
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Ayana Moore
- FHI 360, Durham, North Carolina, United States of America
| | - Barry Kosloff
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kwame Shanaube
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
| | - Sue-Ann Meehan
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Anneen van Deventer
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - H. Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | |
Collapse
|
12
|
Zaongo SD, Zongo AW, Chen Y. Mechanisms underlying the development of type 1 diabetes in ART-treated people living with HIV: an enigmatic puzzle. Front Immunol 2024; 15:1470308. [PMID: 39257582 PMCID: PMC11383789 DOI: 10.3389/fimmu.2024.1470308] [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: 07/25/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
The immunopathogenesis of HIV infection remains poorly understood. Despite the widespread use of effective modern antiretroviral therapy (ART), people living with HIV (PLWH) are known to develop several comorbidities, including type 1 diabetes (T1DM). However, the etiology and critical mechanisms accounting for the onset of T1DM in the preceding context remain unknown. This article proposes to address this topic in order to provide further understanding and future research directions.
Collapse
Affiliation(s)
- Silvere D Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Abel W Zongo
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
13
|
Arribas L, Menéndez-Arias L, Betancor G. May I Help You with Your Coat? HIV-1 Capsid Uncoating and Reverse Transcription. Int J Mol Sci 2024; 25:7167. [PMID: 39000271 PMCID: PMC11241228 DOI: 10.3390/ijms25137167] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) capsid is a protein core formed by multiple copies of the viral capsid (CA) protein. Inside the capsid, HIV-1 harbours all the viral components required for replication, including the genomic RNA and viral enzymes reverse transcriptase (RT) and integrase (IN). Upon infection, the RT transforms the genomic RNA into a double-stranded DNA molecule that is subsequently integrated into the host chromosome by IN. For this to happen, the viral capsid must open and release the viral DNA, in a process known as uncoating. Capsid plays a key role during the initial stages of HIV-1 replication; therefore, its stability is intimately related to infection efficiency, and untimely uncoating results in reverse transcription defects. How and where uncoating takes place and its relationship with reverse transcription is not fully understood, but the recent development of novel biochemical and cellular approaches has provided unprecedented detail on these processes. In this review, we present the latest findings on the intricate link between capsid stability, reverse transcription and uncoating, the different models proposed over the years for capsid uncoating, and the role played by other cellular factors on these processes.
Collapse
Affiliation(s)
- Laura Arribas
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain;
| | - Luis Menéndez-Arias
- Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid), 28049 Madrid, Spain;
| | - Gilberto Betancor
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain;
| |
Collapse
|
14
|
Wen Y, Hu XD. Comment on: Clinical Characteristics, Diagnosis, and Treatments for COVID-19. Curr Med Sci 2024; 44:667-668. [PMID: 38809378 DOI: 10.1007/s11596-024-2897-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Affiliation(s)
- Ying Wen
- Department of Infectious Diseases, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| | - Xin-Dan Hu
- Department of Infectious Diseases, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
15
|
Anderko RR, DePuyt AE, Bronson R, Bullotta AC, Aga E, Bosch RJ, Jones RB, Eron JJ, Mellors JW, Gandhi RT, McMahon DK, Macatangay BJ, Rinaldo CR, Mailliard RB. Persistence of a Skewed Repertoire of NK Cells in People with HIV-1 on Long-Term Antiretroviral Therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1564-1578. [PMID: 38551350 PMCID: PMC11073922 DOI: 10.4049/jimmunol.2300672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
HIV-1 infection greatly alters the NK cell phenotypic and functional repertoire. This is highlighted by the expansion of a rare population of FcRγ- NK cells exhibiting characteristics of traditional immunologic memory in people with HIV (PWH). Although current antiretroviral therapy (ART) effectively controls HIV-1 viremia and disease progression, its impact on HIV-1-associated NK cell abnormalities remains unclear. To address this, we performed a longitudinal analysis detailing conventional and memory-like NK cell characteristics in n = 60 PWH during the first 4 y of ART. Throughout this regimen, a skewed repertoire of cytokine unresponsive FcRγ- memory-like NK cells persisted and accompanied an overall increase in NK surface expression of CD57 and KLRG1, suggestive of progression toward immune senescence. These traits were linked to elevated serum inflammatory biomarkers and increasing Ab titers to human CMV, with human CMV viremia detected in approximately one-third of PWH at years 1-4 of ART. Interestingly, 40% of PWH displayed atypical NK cell subsets, representing intermediate stages of NK-poiesis based on single-cell multiomic trajectory analysis. Our findings indicate that NK cell irregularities persist in PWH despite long-term ART, underscoring the need to better understand the causative mechanisms that prevent full restoration of immune health in PWH.
Collapse
Affiliation(s)
- Renee R. Anderko
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Allison E. DePuyt
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rhianna Bronson
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Arlene C. Bullotta
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Evgenia Aga
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ronald J. Bosch
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - R. Brad Jones
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Joseph J. Eron
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John W. Mellors
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rajesh T. Gandhi
- Infectious Disease Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deborah K. McMahon
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bernard J. Macatangay
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles R. Rinaldo
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robbie B. Mailliard
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Stankiewicz LN, Rossi FMV, Zandstra PW. Rebuilding and rebooting immunity with stem cells. Cell Stem Cell 2024; 31:597-616. [PMID: 38593798 DOI: 10.1016/j.stem.2024.03.012] [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: 01/08/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
Advances in modern medicine have enabled a rapid increase in lifespan and, consequently, have highlighted the immune system as a key driver of age-related disease. Immune regeneration therapies present exciting strategies to address age-related diseases by rebooting the host's primary lymphoid tissues or rebuilding the immune system directly via biomaterials or artificial tissue. Here, we identify important, unanswered questions regarding the safety and feasibility of these therapies. Further, we identify key design parameters that should be primary considerations guiding technology design, including timing of application, interaction with the host immune system, and functional characterization of the target patient population.
Collapse
Affiliation(s)
- Laura N Stankiewicz
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
17
|
Windels EM, Wampande EM, Joloba ML, Boom WH, Goig GA, Cox H, Hella J, Borrell S, Gagneux S, Brites D, Stadler T. HIV co-infection is associated with reduced Mycobacterium tuberculosis transmissibility in sub-Saharan Africa. PLoS Pathog 2024; 20:e1011675. [PMID: 38696531 DOI: 10.1371/journal.ppat.1011675] [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: 09/12/2023] [Revised: 05/14/2024] [Accepted: 04/10/2024] [Indexed: 05/04/2024] Open
Abstract
Persons living with HIV are known to be at increased risk of developing tuberculosis (TB) disease upon infection with Mycobacterium tuberculosis (Mtb). However, it has remained unclear how HIV co-infection affects subsequent Mtb transmission from these patients. Here, we customized a Bayesian phylodynamic framework to estimate the effects of HIV co-infection on the Mtb transmission dynamics from sequence data. We applied our model to four Mtb genomic datasets collected in sub-Saharan African countries with a generalized HIV epidemic. Our results confirm that HIV co-infection is a strong risk factor for developing active TB. Additionally, we demonstrate that HIV co-infection is associated with a reduced effective reproductive number for TB. Stratifying the population by CD4+ T-cell count yielded similar results, suggesting that, in this context, CD4+ T-cell count is not a better predictor of Mtb transmissibility than HIV infection status alone. Together, our genome-based analyses complement observational household contact studies, and more firmly establish the negative association between HIV co-infection and Mtb transmissibility.
Collapse
Affiliation(s)
- Etthel M Windels
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | | | - W Henry Boom
- Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Galo A Goig
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Helen Cox
- University of Cape Town, Cape Town, South Africa
| | - Jerry Hella
- Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Sonia Borrell
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Sebastien Gagneux
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Daniela Brites
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Tanja Stadler
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
18
|
Quaresma MVLDS, Vicente BM, Balchiunas RE, Ribeiro SML. Sarcopenia risk, sarcopenia-related quality of life, and associated factors in people living with human immunodeficiency virus (HIV): A web-based survey. Nutrition 2024; 120:112352. [PMID: 38306734 DOI: 10.1016/j.nut.2024.112352] [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: 06/16/2023] [Revised: 11/17/2023] [Accepted: 01/02/2024] [Indexed: 02/04/2024]
Abstract
OBJECTIVES We aimed to screen for the risk of sarcopenia and sarcopenia-related quality of life and associated factors of people living with HIV (PLWH). RESEARCH METHODS AND PROCEDURES This nonprobabilistic web-based survey evaluated PLWH. The participants were invited directly from a university-based inpatient clinic and responded to a web questionnaire that included the SARC-F and SarQoL to screen people at risk of sarcopenia and their quality of life. People at risk of sarcopenia were defined by the proposed cutoff points for SARC-F (≥ 4 points), and SarQoL overall score was categorized according to the median. Moreover, we performed a logistic regression to investigate associations between HIV-, lifestyle-, and health-associated factors (i.e., physical activity, dietary pattern, sleep quality, gastrointestinal symptoms, HIV diagnosis, type, combinations, and duration of ART, smoking, drinking, BMI, and weight loss), and outcomes (SARC-F and SarQoL). RESULTS The sample comprised 202 PLWH, mainly middle-aged (50.6-60.5 y; n = 101). Only 5.9% (n = 12) are at risk of sarcopenia according to SARC-F, and only 17.3% (n = 35) exhibited lower sarcopenia-related quality of life according to SarQoL. In the multiple models, only the gastrointestinal symptoms increased the odds of sarcopenia risk (OR: 1.058; P = 0.01) and poor sarcopenia-associated quality of life (OR: 1.041; P = 0.013). CONCLUSIONS We verified that only 5.9% and 17.3% of PLWH are at risk of sarcopenia and presented lower sarcopenia-related quality of life, respectively. Only the gastrointestinal symptoms were associated with a risk of sarcopenia and lower sarcopenia-related quality of life, without significant differences between age groups.
Collapse
Affiliation(s)
- Marcus V L Dos Santos Quaresma
- Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil; Centro Universitário São Camilo, São Paulo, Brazil
| | | | - Roseli Espindola Balchiunas
- Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil; Centro Universitário São Camilo, São Paulo, Brazil
| | - Sandra M L Ribeiro
- Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil; Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Brazil.
| |
Collapse
|
19
|
Matsuda K, Maeda K. HIV Reservoirs and Treatment Strategies toward Curing HIV Infection. Int J Mol Sci 2024; 25:2621. [PMID: 38473868 DOI: 10.3390/ijms25052621] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Combination antiretroviral therapy (cART) has significantly improved the prognosis of individuals living with human immunodeficiency virus (HIV). Acquired immunodeficiency syndrome has transformed from a fatal disease to a treatable chronic infection. Currently, effective and safe anti-HIV drugs are available. Although cART can reduce viral production in the body of the patient to below the detection limit, it cannot eliminate the HIV provirus integrated into the host cell genome; hence, the virus will be produced again after cART discontinuation. Therefore, research into a cure (or remission) for HIV has been widely conducted. In this review, we focus on drug development targeting cells latently infected with HIV and assess the progress including our current studies, particularly in terms of the "Shock and Kill", and "Block and Lock" strategies.
Collapse
Affiliation(s)
- Kouki Matsuda
- Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Kenji Maeda
- Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
20
|
Liu H, He S, Yang T, Lu C, Yao Y, Zhou R, Yin K, He Y, Cheng J. Tolerability and effectiveness of albuvirtide combined with dolutegravir for hospitalized people living with HIV/AIDS. Medicine (Baltimore) 2023; 102:e35344. [PMID: 37960773 PMCID: PMC10637561 DOI: 10.1097/md.0000000000035344] [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: 03/21/2023] [Accepted: 08/21/2023] [Indexed: 11/15/2023] Open
Abstract
Treatment options for hospitalized people living with HIV/AIDS (PLWHA) with opportunistic infections and comorbidities are limited in China. Albuvirtide (ABT), a new peptide drug, is a long-acting HIV fusion inhibitor with limited drug-drug interactions and fast onset time. This single-center, retrospective cohort study investigated the effectiveness and safety of ABT plus dolutegravir (DTG) therapy in a real-world setting. We performed a chart review on the electronic patient records for hospitalized PLWHA using ABT plus DTG between April and December 2020. The clinical outcomes were retrospectively analyzed. Among 151 PLWHA (mean age 47.6 ± 15.9 years), 140 (93%) had at least 1 episode of bacterial and/or fungal infections and 64 (42%) had other comorbidities including syphilis, hepatitis B, and/or hypertension. ABT plus DTG was given to 87 treatment-naïve (TN) and 64 treatment-experienced (TE) PLWHA. Regardless of treatment history, mean HIV-1 RNA levels significantly decreased from 4.32 log10copies/mL to 2.24 log10copies/mL, 2.10 log10copies/mL and 1.89 log10copies/mL after 2, 4 and 8 weeks of treatment, respectively (P < .0001). Compared with baseline mean CD4 + T-cell counts of 122.72 cells/μL, it increased to 207.87 cells/μL (P = .0067) and 218.69 cells/μL (P = .0812) after 4 and 8 weeks of treatment. Except for limited laboratory abnormalities such as hyperuricemia, increased creatinine level, and hyperglycemia observed after treatment, no other clinical adverse events were considered related to ABT plus DTG. Data suggests that ABT plus DTG is safe and effective for critically-ill hospitalized PLWHA. In view of the rapid viral load suppression and restoration of CD4 + count within 8 weeks of treatment, its clinical application warrants further investigation.
Collapse
Affiliation(s)
- Huanxia Liu
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Shenghua He
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Tongtong Yang
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Chunrong Lu
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Yuan Yao
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Ruifeng Zhou
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Ke Yin
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Yuanhong He
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Jing Cheng
- Clinical Section 1, Department of Infectious Disease, Public Health Clinical Center of Chengdu, Chengdu, China
| |
Collapse
|
21
|
Zhang W, Yan J, Luo H, Wang X, Ruan L. Incomplete immune reconstitution and its predictors in people living with HIV in Wuhan, China. BMC Public Health 2023; 23:1808. [PMID: 37716975 PMCID: PMC10505310 DOI: 10.1186/s12889-023-16738-w] [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: 04/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023] Open
Abstract
OBJECTIVE This study aimed to build and validate a nomogram model to predict the risk of incomplete immune reconstitution in people living with HIV (PLWH). METHODS Totally 3783 individuals with a confirmed diagnosis of HIV/AIDS were included. A predictive model was developed based on a retrospective set (N = 2678) and was validated using the remaining cases (N = 1105). Univariate and multivariate logistic regression analyses were performed to determine valuable predictors among the collected clinical and laboratory variables. The predictive model is presented in the form of a nomogram, which is internally and externally validated with two independent datasets. The discrimination of nomograms was assessed by calculating the area under the curve (AUC). Besides, calibration curve and decision curve (DCA) analyses were performed in the training and validation sets. RESULTS The final model comprised 5 predictors, including baseline CD4, age at ART initiation, BMI, HZ and TBIL. The AUC of the nomogram model was 0.902, 0.926, 0.851 in the training cohort, internal validation and external cohorts. The calibration accuracy and diagnostic performance were satisfactory in both the training and validation sets. CONCLUSIONS This predictive model based on a retrospective study was externally validated using 5 readily available clinical indicators. It showed high performance in predicting the risk of incomplete immune reconstitution in people living with HIV.
Collapse
Affiliation(s)
- Wenyuan Zhang
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, Hubei, China
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, 430023, Hubei, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, Hubei, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, Hubei, China
| | - Jisong Yan
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, 430023, Hubei, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, Hubei, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, Hubei, China
- Department of Respiratory and Critical Care Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, Hubei, China
| | - Hong Luo
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, 430023, Hubei, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, Hubei, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, Hubei, China
- Department of Respiratory and Critical Care Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, Hubei, China
| | - Xianguang Wang
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, 430023, Hubei, China.
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, Hubei, China.
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, Hubei, China.
- Department of Respiratory and Critical Care Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, Hubei, China.
| | - Lianguo Ruan
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, Hubei, China.
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, 430023, Hubei, China.
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, 430023, Hubei, China.
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, Hubei, China.
| |
Collapse
|
22
|
Bekker LG, Beyrer C, Mgodi N, Lewin SR, Delany-Moretlwe S, Taiwo B, Masters MC, Lazarus JV. HIV infection. Nat Rev Dis Primers 2023; 9:42. [PMID: 37591865 DOI: 10.1038/s41572-023-00452-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 08/19/2023]
Abstract
The AIDS epidemic has been a global public health issue for more than 40 years and has resulted in ~40 million deaths. AIDS is caused by the retrovirus, HIV-1, which is transmitted via body fluids and secretions. After infection, the virus invades host cells by attaching to CD4 receptors and thereafter one of two major chemokine coreceptors, CCR5 or CXCR4, destroying the host cell, most often a T lymphocyte, as it replicates. If unchecked this can lead to an immune-deficient state and demise over a period of ~2-10 years. The discovery and global roll-out of rapid diagnostics and effective antiretroviral therapy led to a large reduction in mortality and morbidity and to an expanding group of individuals requiring lifelong viral suppressive therapy. Viral suppression eliminates sexual transmission of the virus and greatly improves health outcomes. HIV infection, although still stigmatized, is now a chronic and manageable condition. Ultimate epidemic control will require prevention and treatment to be made available, affordable and accessible for all. Furthermore, the focus should be heavily oriented towards long-term well-being, care for multimorbidity and good quality of life. Intense research efforts continue for therapeutic and/or preventive vaccines, novel immunotherapies and a cure.
Collapse
Affiliation(s)
- Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, RSA, Cape Town, South Africa.
| | - Chris Beyrer
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Nyaradzo Mgodi
- University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | | | - Babafemi Taiwo
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Jeffrey V Lazarus
- CUNY Graduate School of Public Health and Health Policy, New York, NY, USA
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
23
|
Zhang W, Ruan L. Recent advances in poor HIV immune reconstitution: what will the future look like? Front Microbiol 2023; 14:1236460. [PMID: 37608956 PMCID: PMC10440441 DOI: 10.3389/fmicb.2023.1236460] [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: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Abstract
Combination antiretroviral therapy has demonstrated proved effectiveness in suppressing viral replication and significantly recovering CD4+ T cell count in HIV type-1 (HIV-1)-infected patients, contributing to a dramatic reduction in AIDS morbidity and mortality. However, the factors affecting immune reconstitution are extremely complex. Demographic factors, co-infection, baseline CD4 cell level, abnormal immune activation, and cytokine dysregulation may all affect immune reconstitution. According to report, 10-40% of HIV-1-infected patients fail to restore the normalization of CD4+ T cell count and function. They are referred to as immunological non-responders (INRs) who fail to achieve complete immune reconstitution and have a higher mortality rate and higher risk of developing other non-AIDS diseases compared with those who achieve complete immune reconstitution. Heretofore, the mechanisms underlying incomplete immune reconstitution in HIV remain elusive, and INRs are not effectively treated or mitigated. This review discusses the recent progress of mechanisms and factors responsible for incomplete immune reconstitution in AIDS and summarizes the corresponding therapeutic strategies according to different mechanisms to improve the individual therapy.
Collapse
Affiliation(s)
| | - Lianguo Ruan
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, Hubei, China
| |
Collapse
|
24
|
Rodriguez MT, McLaurin KA, Shtutman M, Kubinak JL, Mactutus CF, Booze RM. Therapeutically targeting the consequences of HIV-1-associated gastrointestinal dysbiosis: Implications for neurocognitive and affective alterations. Pharmacol Biochem Behav 2023; 229:173592. [PMID: 37390973 PMCID: PMC10494709 DOI: 10.1016/j.pbb.2023.173592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Approximately 50 % of the individuals living with human immunodeficiency virus type 1 (HIV-1) are plagued by debilitating neurocognitive impairments (NCI) and/or affective alterations. Sizeable alterations in the composition of the gut microbiome, or gastrointestinal dysbiosis, may underlie, at least in part, the NCI, apathy, and/or depression observed in this population. Herein, two interrelated aims will be critically addressed, including: 1) the evidence for, and functional implications of, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals; and 2) the potential for therapeutically targeting the consequences of this dysbiosis for the treatment of HIV-1-associated NCI and affective alterations. First, gastrointestinal microbiome dysbiosis in HIV-1 seropositive individuals is characterized by decreased alpha (α) diversity, a decreased relative abundance of bacterial species belonging to the Bacteroidetes phylum, and geographic-specific alterations in Bacillota (formerly Firmicutes) spp. Fundamentally, changes in the relative abundance of Bacteroidetes and Bacillota spp. may underlie, at least in part, the deficits in γ-aminobutyric acid and serotonin neurotransmission, as well as prominent synaptodendritic dysfunction, observed in this population. Second, there is compelling evidence for the therapeutic utility of targeting synaptodendritic dysfunction as a method to enhance neurocognitive function and improve motivational dysregulation in HIV-1. Further research is needed to determine whether the therapeutics enhancing synaptic efficacy exert their effects by altering the gut microbiome. Taken together, understanding gastrointestinal microbiome dysbiosis resulting from chronic HIV-1 viral protein exposure may afford insight into the mechanisms underlying HIV-1-associated neurocognitive and/or affective alterations; mechanisms which can be subsequently targeted via novel therapeutics.
Collapse
Affiliation(s)
- Mason T Rodriguez
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Kristen A McLaurin
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Michael Shtutman
- Drug Discovery and Biomedical Sciences, College of Pharmacy, 715 Sumter Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Jason L Kubinak
- Pathology, Microbiology & Immunology, School of Medicine Columbia, 6311 Garners Ferry Road, Building 2, Columbia, SC 29209, United States of America
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, 1512 Pendleton Street, University of South Carolina, Columbia, SC 29208, United States of America.
| |
Collapse
|
25
|
Li H, McLaurin KA, Mactutus CF, Booze RM. Microglia proliferation underlies synaptic dysfunction in the prefrontal cortex: implications for the pathogenesis of HIV-1-associated neurocognitive and affective alterations. J Neurovirol 2023; 29:460-471. [PMID: 37222970 PMCID: PMC10629500 DOI: 10.1007/s13365-023-01147-x] [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: 01/20/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/25/2023]
Abstract
Microglia, which are productively infected by HIV-1, are critical for brain development and maturation, as well as synaptic plasticity. The pathophysiology of HIV-infected microglia and their role in the pathogenesis of HIV-1-associated neurocognitive and affective alterations, however, remains understudied. Three complementary aims were undertaken to critically address this knowledge gap. First, the expression of HIV-1 mRNA in the dorsolateral prefrontal cortex of postmortem HIV-1 seropositive individuals with HAND was investigated. Utilization of immunostaining and/or RNAscope multiplex fluorescent assays revealed prominent HIV-1 mRNA in microglia of postmortem HIV-1 seropositive individuals with HAND. Second, measures of microglia proliferation and neuronal damage were evaluated in chimeric HIV (EcoHIV) rats. Eight weeks after EcoHIV inoculation, enhanced microglial proliferation was observed in the medial prefrontal cortex (mPFC) of EcoHIV rats, evidenced by an increased number of cells co-localized with both Iba1 + and Ki67 + relative to control animals. Neuronal damage in EcoHIV infected rats was evidenced by pronounced decreases in both synaptophysin and postsynaptic density protein 95 (PSD-95), markers of presynaptic and postsynaptic damage, respectively. Third, regression analyses were conducted to evaluate whether microglia proliferation mechanistically underlies neuronal damage in EcoHIV and control animals. Indeed, microglia proliferation accounted for 42-68.6% of the variance in synaptic dysfunction. Collectively, microglia proliferation induced by chronic HIV-1 viral protein exposure may underlie the profound synaptodendritic alterations in HIV-1. Understanding how microglia are involved in the pathogenesis of HAND and HIV-1-associated affective disorders affords a key target for the development of novel therapeutics.
Collapse
Affiliation(s)
- Hailong Li
- Cognitive and Neural Science Program, Department of Psychology, University of South Carolina, Barnwell College, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Kristen A McLaurin
- Cognitive and Neural Science Program, Department of Psychology, University of South Carolina, Barnwell College, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, University of South Carolina, Barnwell College, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, University of South Carolina, Barnwell College, 1512 Pendleton Street, Columbia, SC, 29208, USA.
| |
Collapse
|
26
|
Carmona-Pérez L, Dagenais-Lussier X, Mai LT, Stögerer T, Swaminathan S, Isnard S, Rice MR, Barnes BJ, Routy JP, van Grevenynghe J, Stäger S. The TLR7/IRF-5 axis sensitizes memory CD4+ T cells to Fas-mediated apoptosis during HIV-1 infection. JCI Insight 2023; 8:e167329. [PMID: 37227774 PMCID: PMC10371351 DOI: 10.1172/jci.insight.167329] [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: 11/18/2022] [Accepted: 05/23/2023] [Indexed: 05/27/2023] Open
Abstract
HIV-1 infection is characterized by inflammation and a progressive decline in CD4+ T cell count. Despite treatment with antiretroviral therapy (ART), the majority of people living with HIV (PLWH) maintain residual levels of inflammation, a low degree of immune activation, and higher sensitivity to cell death in their memory CD4+ T cell compartment. To date, the mechanisms responsible for this high sensitivity remain elusive. We have identified the transcription factor IRF-5 to be involved in impairing the maintenance of murine CD4+ T cells during chronic infection. Here, we investigate whether IRF-5 also contributes to memory CD4+ T cell loss during HIV-1 infection. We show that TLR7 and IRF-5 were upregulated in memory CD4+ T cells from PLWH, when compared with naturally protected elite controllers and HIVfree participants. TLR7 was upstream of IRF-5, promoting Caspase 8 expression in CD4+ T cells from ART HIV-1+ but not from HIVfree donors. Interestingly, the TLR7/IRF-5 axis acted synergistically with the Fas/FasL pathway, suggesting that TLR7 and IRF-5 expression in ART HIV-1+ memory CD4+ T cells represents an imprint that predisposes cells to Fas-mediated apoptosis. This predisposition could be blocked using IRF-5 inhibitory peptides, suggesting IRF-5 blockade as a possible therapy to prevent memory CD4+ T cell loss in PLWH.
Collapse
Affiliation(s)
- Liseth Carmona-Pérez
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Xavier Dagenais-Lussier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Linh T. Mai
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Tanja Stögerer
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Sharada Swaminathan
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Stéphane Isnard
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec, Canada
| | - Matthew R. Rice
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Betsy J. Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, Quebec, Canada
| | - Julien van Grevenynghe
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| | - Simona Stäger
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, and Infectiopôle-INRS, Laval, Quebec, Canada
| |
Collapse
|
27
|
Enichen E, Adams RB, Demmig-Adams B. Physical Activity as an Adjunct Treatment for People Living with HIV? Am J Lifestyle Med 2023; 17:502-517. [PMID: 37426740 PMCID: PMC10328202 DOI: 10.1177/15598276221078222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
This review evaluates physical activity as a candidate for an adjunct treatment, in conjunction with antiretroviral therapy (ART), for people living with HIV (PLWH). Evidence is summarized that chronic, non-resolving inflammation (a principal feature of immune system dysfunction) and a dysfunctional state of the gut environment are key factors in HIV infection that persist despite treatment with ART. In addition, evidence is summarized that regular physical activity may restore normal function of both the immune system and the gut environment and may thereby ameliorate symptoms and non-resolving inflammation-associated comorbidities that burden PLWH. Physicians who care for PLWH could thus consider incorporating physical activity into treatment plans to complement ART. It is also discussed that different types of physical activity can have different effects on the gut environment and immune function, and that future research should establish more specific criteria for the design of exercise regimens tailored to PLWH.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Robert B. Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| |
Collapse
|
28
|
Bao Q, Zhou J. Various strategies for developing APOBEC3G protectors to circumvent human immunodeficiency virus type 1. Eur J Med Chem 2023; 250:115188. [PMID: 36773550 DOI: 10.1016/j.ejmech.2023.115188] [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: 11/28/2022] [Revised: 01/18/2023] [Accepted: 02/04/2023] [Indexed: 02/09/2023]
Abstract
Host restriction factor APOBEC3G (A3G) efficiently restricts Vif-deficient HIV-1 by being packaged with progeny virions and causing the G to A mutation during HIV-1 viral DNA synthesis as the progeny virus infects new cells. HIV-1 expresses Vif protein to resist the activity of A3G by mediating A3G degradation. This process requires the self-association of Vif in concert with A3G proteins, protein chaperones, and factors of the ubiquitination machinery, which are potential targets to discover novel anti-HIV drugs. This review will describe compounds that have been reported so far to inhibit viral replication of HIV-1 by protecting A3G from Vif-mediated degradation.
Collapse
Affiliation(s)
- Qiqi Bao
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China.
| |
Collapse
|
29
|
(5R)-5-hydroxytriptolide for HIV immunological non-responders receiving ART: a randomized, double-blinded, placebo-controlled phase II study. THE LANCET REGIONAL HEALTH - WESTERN PACIFIC 2023. [DOI: 10.1016/j.lanwpc.2023.100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
30
|
Thymic Exhaustion and Increased Immune Activation Are the Main Mechanisms Involved in Impaired Immunological Recovery of HIV-Positive Patients under ART. Viruses 2023; 15:v15020440. [PMID: 36851655 PMCID: PMC9961132 DOI: 10.3390/v15020440] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Decades of studies in antiretroviral therapy (ART) have passed, and the mechanisms that determine impaired immunological recovery in HIV-positive patients receiving ART have not been completely elucidated yet. Thus, T-lymphocytes immunophenotyping and cytokines levels were analyzed in 44 ART-treated HIV-positive patients who had a prolonged undetectable plasma viral load. The patients were classified as immunological non-responders (INR = 13) and immunological responders (IR = 31), according to their CD4+ T cell levels. Evaluating pre-CD4+ levels, we observed a statistically significant trend between lower CD4+ T cell levels and INR status (Z = 3.486, p < 0.001), and during 18 months of ART, the CD4+ T cell levels maintained statistical differences between the INR and IR groups (WTS = 37.252, p < 0.001). Furthermore, the INRs were associated with an elevated age at ART start; a lower pre-treatment CD4+ T cell count and a percentage that remained low even after 18 months of ART; lower levels of recent thymic emigrant (RTE) CD4+ T cell (CD45RA + CD31+) and a naïve CD4+ T cell (CD45RA + CD62L+); higher levels of central memory CD4+ T cells (CD45RA-CD62L+); and higher immune activation by CD4+ expressing HLA-DR+ or both (HLA-DR+ and CD38+) when compared with IRs. Our study demonstrates that thymic exhaustion and increased immune activation are two mechanisms substantially implicated in the impaired immune recovery of ART-treated HIV patients.
Collapse
|
31
|
Anderko RR, Mailliard RB. Mapping the interplay between NK cells and HIV: therapeutic implications. J Leukoc Biol 2023; 113:109-138. [PMID: 36822173 PMCID: PMC10043732 DOI: 10.1093/jleuko/qiac007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Indexed: 01/18/2023] Open
Abstract
Although highly effective at durably suppressing plasma HIV-1 viremia, combination antiretroviral therapy (ART) treatment regimens do not eradicate the virus, which persists in long-lived CD4+ T cells. This latent viral reservoir serves as a source of plasma viral rebound following treatment interruption, thus requiring lifelong adherence to ART. Additionally, challenges remain related not only to access to therapy but also to a higher prevalence of comorbidities with an inflammatory etiology in treated HIV-1+ individuals, underscoring the need to explore therapeutic alternatives that achieve sustained virologic remission in the absence of ART. Natural killer (NK) cells are uniquely positioned to positively impact antiviral immunity, in part due to the pleiotropic nature of their effector functions, including the acquisition of memory-like features, and, therefore, hold great promise for transforming HIV-1 therapeutic modalities. In addition to defining the ability of NK cells to contribute to HIV-1 control, this review provides a basic immunologic understanding of the impact of HIV-1 infection and ART on the phenotypic and functional character of NK cells. We further delineate the qualities of "memory" NK cell populations, as well as the impact of HCMV on their induction and subsequent expansion in HIV-1 infection. We conclude by highlighting promising avenues for optimizing NK cell responses to improve HIV-1 control and effect a functional cure, including blockade of inhibitory NK receptors, TLR agonists to promote latency reversal and NK cell activation, CAR NK cells, BiKEs/TriKEs, and the role of HIV-1-specific bNAbs in NK cell-mediated ADCC activity against HIV-1-infected cells.
Collapse
Affiliation(s)
- Renee R. Anderko
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robbie B. Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| |
Collapse
|
32
|
Li H, McLaurin KA, Mactutus CF, Booze RM. Microglia Proliferation Underlies Synaptic Dysfunction in the Prefrontal Cortex: Implications for the Pathogenesis of HIV-1-Associated Neurocognitive and Affective Alterations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524942. [PMID: 36711456 PMCID: PMC9882316 DOI: 10.1101/2023.01.20.524942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Microglia, which are productively infected by HIV-1, are critical for brain development and maturation, as well as synaptic plasticity. The pathophysiology of HIV-infected microglia and their role in the pathogenesis of HIV-1-associated neurocognitive and affective alterations, however, remains understudied. Three complementary aims were undertaken to critically address this knowledge gap. First, the predominant cell type expressing HIV-1 mRNA in the dorsolateral prefrontal cortex of postmortem HIV-1 seropositive individuals with HAND was investigated. Utilization of a combined RNAscope multiplex fluorescent and immunostaining assay revealed prominent HIV-1 mRNA in microglia of postmortem HIV-1 seropositive individuals with HAND. Second, measures of microglia proliferation and neuronal damage were evaluated in chimeric HIV (EcoHIV) rats. Eight weeks after EcoHIV innoculation, enhanced microglial proliferation was observed in the medial prefrontal cortex (mPFC) of EcoHIV rats, evidenced by an increased number of cells co-localized with both Iba1+ and Ki67+ relative to control animals. Neuronal damage in EcoHIV infected rats was evidenced by pronounced decreases in both synaptophysin and post synaptic density protein 95 (PSD-95), markers of pre-synaptic and post-synaptic damage, respectively. Third, regression analyses were conducted to evaluate whether microglia proliferation mechanistically underlies neuronal damage in EcoHIV and control animals. Indeed, microglia proliferation accounts for 42-68.6% of the variance in synaptic dysfunction. Collectively, microglia proliferation induced by chronic HIV-1 viral protein exposure may underlie the profound synaptodendritic alterations in HIV-1. Understanding how microglia are involved in the pathogenesis of HAND and HIV-1-associated affective disorders affords a key target for the development of novel therapeutics.
Collapse
|
33
|
Brown AN, Lang Y, Zhou J, Franco EJ, Hanrahan KC, Bulitta JB, Drusano GL. Why Molnupiravir Fails in Hospitalized Patients. mBio 2022; 13:e0291622. [PMID: 36374076 PMCID: PMC9765607 DOI: 10.1128/mbio.02916-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has radically altered daily life. Effective antiviral therapies to combat COVID-19, especially severe disease, remain scarce. Molnupiravir is an antiviral that has shown clinical efficacy against mild-to-moderate COVID-19 but failed to provide benefit to hospitalized patients with severe disease. Here, we explained the mechanism behind the failure of molnupiravir in hospitalized patients and identified alternative dosing strategies that would improve therapeutic outcomes in all patients with COVID-19. We showed that delaying therapy initiation markedly decreased the antiviral effect of molnupiravir, and these results were directly related to intracellular drug triphosphate pools and intracellular viral burden at the start of therapy. The adverse influence of therapeutic delay could be overcome by increasing drug exposure, which increased intracellular molnupiravir triphosphate concentrations that inhibited viral replication. These findings illustrated that molnupiravir must be administered as early as possible following COVID-19 symptom onset to maximize therapeutic efficacy. Higher doses may be effective in patients hospitalized with severe disease, but the safety of high-dose molnupiravir regimens is unknown. Our findings could be extended to design effective regimens with nucleoside analogs for other RNA viruses, especially those with pandemic potential. IMPORTANCE In this study, we showed that early intervention with molnupiravir resulted in a greater antiviral effect, and we explained the mechanism behind this phenomenon. Our results predicted and explained the failure of molnupiravir in hospitalized patients and highlighted the utility of preclinical pharmacodynamic studies to design optimal antiviral regimens for the treatment of viral diseases. This contrasts with the procedure that was implemented early in the pandemic in which clinical studies were conducted in the absence of preclinical experimentation. These findings are significant and demonstrated the importance of experimental approaches in antiviral development for treatments against COVID-19 as well as other viral diseases.
Collapse
Affiliation(s)
- Ashley N. Brown
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
- Department of Medicine, College of Medicine, University of Florida, Orlando, Florida, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Yinzhi Lang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jieqiang Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Evelyn J. Franco
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
- Department of Medicine, College of Medicine, University of Florida, Orlando, Florida, USA
| | - Juergen B. Bulitta
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - George L. Drusano
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
- Department of Medicine, College of Medicine, University of Florida, Orlando, Florida, USA
| |
Collapse
|
34
|
Lu L, Li X, Liu X, Qiu Z, Han Y, Song X, Li Y, Li X, Cao W, Lv W, Dou Z, Li T. The pattern and magnitude of T cell subsets reconstitution during ten years of ART with viral suppression in HIV-infected patients. Aging (Albany NY) 2022; 14:9647-9667. [PMID: 36490352 PMCID: PMC9792206 DOI: 10.18632/aging.204416] [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: 06/07/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND The extent of immune reconstitution in human immunodeficiency virus (HIV) infected persons receiving long-term antiretroviral therapy (ART) with controlled viral load has been controversial. We studied the extent and speed of T cell subsets retrieval after long-term antiretroviral treatment. METHODS 662 HIV-infected patients followed at least 2 years whose plasma HIV-1 RNA load <50 copies/mL were evaluated for longitudinal and functional phenotypic indices of immune restoration. Determinants of change in magnitude and importance of recovery have been evaluated using mixed linear regression models. RESULTS Almost all robust immune restorations achieved occurred after 2-3 years of ART. The median CD4 lymphocyte count increased 449 cells/μl (IQR 303-604) from 226 cells/μl (IQR 83-336) at baseline during the third year (P < 0.001); CD4+T lymphocyte rises during the sixth and tenth years were not significant. Naive and memory CD4+T cells'reconstitution occurred in the sixth and eighth years of ART but no significant change thereafter. The change of CD45RA+Naïve and CD45RA-memory CD4+T cell reconstitution is different in baseline CD4+T cell counts <100 cells/μl group and in baseline CD4+T cell counts >100 cells/μl group. Activation antigen expression (CD38 or HLA-DR) on CD8 lymphocytes declined mostly during the first till second year, and after 4 years, activation antigen expression on patient lymphocytes showed no significant change. The proportion of CD4 cells expressing CD28 climbed during the first years and reached normal levels in the second year. CONCLUSIONS Immune restoration was dependent on the capacity of immune system during the first 2-3 year of ART. But the significant change of CD4 and compartments of CD4+T cells could persist until 6-8 years. The pattern of CD38+CD8+, HLA-DR+CD8+, CD28+CD4+ T cells could quickly return to normal level and no significant change after sufficient time of ART. In general, the immune response compared to the baseline status may be the overall effect from the age and time of antiretroviral treatment.
Collapse
Affiliation(s)
- Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosheng Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhifeng Qiu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxia Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Lv
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihui Dou
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Valyaeva AA, Tikhomirova MA, Potashnikova DM, Bogomazova AN, Snigiryova GP, Penin AA, Logacheva MD, Arifulin EA, Shmakova AA, Germini D, Kachalova AI, Saidova AA, Zharikova AA, Musinova YR, Mironov AA, Vassetzky YS, Sheval EV. Ectopic expression of HIV-1 Tat modifies gene expression in cultured B cells: implications for the development of B-cell lymphomas in HIV-1-infected patients. PeerJ 2022; 10:e13986. [PMID: 36275462 PMCID: PMC9586123 DOI: 10.7717/peerj.13986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023] Open
Abstract
An increased frequency of B-cell lymphomas is observed in human immunodeficiency virus-1 (HIV-1)-infected patients, although HIV-1 does not infect B cells. Development of B-cell lymphomas may be potentially due to the action of the HIV-1 Tat protein, which is actively released from HIV-1-infected cells, on uninfected B cells. The exact mechanism of Tat-induced B-cell lymphomagenesis has not yet been precisely identified. Here, we ectopically expressed either Tat or its TatC22G mutant devoid of transactivation activity in the RPMI 8866 lymphoblastoid B cell line and performed a genome-wide analysis of host gene expression. Stable expression of both Tat and TatC22G led to substantial modifications of the host transcriptome, including pronounced changes in antiviral response and cell cycle pathways. We did not find any strong action of Tat on cell proliferation, but during prolonged culturing, Tat-expressing cells were displaced by non-expressing cells, indicating that Tat expression slightly inhibited cell growth. We also found an increased frequency of chromosome aberrations in cells expressing Tat. Thus, Tat can modify gene expression in cultured B cells, leading to subtle modifications in cellular growth and chromosome instability, which could promote lymphomagenesis over time.
Collapse
Affiliation(s)
- Anna A. Valyaeva
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Tikhomirova
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Daria M. Potashnikova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra N. Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | | | | | - Maria D. Logacheva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Eugene A. Arifulin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Anna A. Shmakova
- Koltzov Institute of Developmental Biology, Moscow, Russia,UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Diego Germini
- UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Anastasia I. Kachalova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Aleena A. Saidova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Anastasia A. Zharikova
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana R. Musinova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Andrey A. Mironov
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Institute for Information Transmission Problems, Moscow, Russia
| | - Yegor S. Vassetzky
- Koltzov Institute of Developmental Biology, Moscow, Russia,UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Eugene V. Sheval
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
36
|
Sitoe N, Ahmed MIM, Enosse M, Bakuli A, Chissumba RM, Held K, Hoelscher M, Nhassengo P, Khosa C, Rachow A, Geldmacher C, on behalf of TB Sequel Consortium. Tuberculosis Treatment Response Monitoring by the Phenotypic Characterization of MTB-Specific CD4+ T-Cells in Relation to HIV Infection Status. Pathogens 2022; 11:pathogens11091034. [PMID: 36145465 PMCID: PMC9506022 DOI: 10.3390/pathogens11091034] [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: 08/16/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
HIV infection causes systemic immune activation, impacts TB disease progression and hence may influence the diagnostic usability of Mycobacterium tuberculosis-specific T cell profiling. We investigated changes of activation and maturation markers on MTB-specific CD4+ T-cells after anti-tuberculosis treatment initiation in relation to HIV status and the severity of lung impairment. Thawed peripheral blood mononuclear cells from TB patients with (n = 27) and without HIV (n = 17) were analyzed using an intracellular IFN-γ assay and flow cytometry 2 and 6 months post-TB treatment initiation. H37Rv antigen was superior to the profile MTB-specific CD4+ T-cells phenotype when compared to PPD and ESAT6/CFP10. Regardless of HIV status and the severity of lung impairment, activation markers (CD38, HLA-DR and Ki67) on MTB-specific CD4+ T-cells declined after TB treatment initiation (p < 0.01), but the expression of the maturation marker CD27 did not change over the course of TB treatment. The MTB-specific T cell phenotype before, during and after treatment completion was similar between people living with and without HIV, as well as between subjects with severe and mild lung impairment. These data suggest that the assessment of activation and maturation markers on MTB-specific CD4+ T-cells can be useful for TB treatment monitoring, regardless of HIV status and the severity of lung disease.
Collapse
Affiliation(s)
- Nádia Sitoe
- Instituto Nacional de Saúde, Marracuene 3943, Mozambique
- CIH LMU Center for International Health, Ludwig-Maximilians University, 80802 Munich, Germany
- Correspondence: ; Tel.: +258-840784833
| | - Mohamed I. M. Ahmed
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | - Maria Enosse
- Instituto Nacional de Saúde, Marracuene 3943, Mozambique
| | - Abhishek Bakuli
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | | | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | | | - Celso Khosa
- Instituto Nacional de Saúde, Marracuene 3943, Mozambique
| | - Andrea Rachow
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), 80802 Munich, Germany
- German Center for Infection Research, Partner Site Munich, 80802 Munich, Germany
| | | |
Collapse
|
37
|
Petit C. D’un protocole de soin au succès d’un essai clinique. Med Sci (Paris) 2022; 38:707-713. [DOI: 10.1051/medsci/2022109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Les innovations pour traiter l’infection par le virus de l’immunodéficience humaine (VIH) n’ont pas cessé depuis les premières monothérapies et, en 1996, les premières trithérapies. L’une d’elles vient d’être validée par l’essai ANRS QUATUOR. Elle consiste à prendre deux fois moins de médicaments, en rendant le traitement intermittent. À la demande des patients non adhérents à sa prescription standard, Jacques Leibowitch a encadré cette pratique dès 2002, en s’appuyant sur une étude transgressant le dogme de l’adhésion stricte au traitement quotidien. Ce concept de traitement à temps partiel provenait des travaux du groupe d’Anthony Fauci, mais il le revisitera pour le pousser à son apogée avec la cohorte Iccarre. Son intention strictement thérapeutique s’inscrivit initialement dans le cadre du protocole de soin Iccarre qui, en 2020, comptait 96 patients, majoritairement en réduction médicamenteuse de 70 % grâce à l’ultra-intermittence thérapeutique. Il a posé les bases de l’essai contrôlé QUATUOR dont le résultat, récemment publié, montre la non infériorité des traitements intermittents à 4 jours/7 de médicaments par rapport au traitement standard.
Collapse
|
38
|
Zhang L, Wei Y, Wang D, Du J, Wang X, Li B, Jiang M, Zhang M, Chen N, Deng M, Song C, Chen D, Wu L, Xiao J, Liang H, Zhao H, Kong Y. Elevated Foxp3+ double-negative T cells are associated with disease progression during HIV infection. Front Immunol 2022; 13:947647. [PMID: 35967422 PMCID: PMC9365964 DOI: 10.3389/fimmu.2022.947647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/29/2022] [Indexed: 11/14/2022] Open
Abstract
Persistent immune activation, which occurs during the whole course of HIV infection, plays a pivotal role in CD4+ T cells depletion and AIDS progression. Furthermore, immune activation is a key factor that leads to impaired immune reconstitution after long-term effective antiretroviral therapy (ART), and is even responsible for the increased risk of developing non-AIDS co-morbidities. Therefore, it’s imperative to identify an effective intervention targeting HIV-associated immune activation to improve disease management. Double negative T cells (DNT) were reported to provide immunosuppression during HIV infection, but the related mechanisms remained puzzled. Foxp3 endows Tregs with potent suppressive function to maintain immune homeostasis. However, whether DNT cells expressed Foxp3 and the accurate function of these cells urgently needed to be investigated. Here, we found that Foxp3+ DNT cells accumulated in untreated people living with HIV (PLWH) with CD4+ T cell count less than 200 cells/µl. Moreover, the frequency of Foxp3+ DNT cells was negatively correlated with CD4+ T cell count and CD4/CD8 ratio, and positively correlated with immune activation and systemic inflammation in PLWH. Of note, Foxp3+ DNT cells might exert suppressive regulation by increased expression of CD39, CD25, or vigorous proliferation (high levels of GITR and ki67) in ART-naive PLWH. Our study underlined the importance of Foxp3+ DNT cells in the HIV disease progression, and suggest that Foxp3+ DNT may be a potential target for clinical intervention for the control of immune activation during HIV infection.
Collapse
Affiliation(s)
- Leidan Zhang
- Peking University Ditan Teaching Hospital, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Wei
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Di Wang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Juan Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xinyue Wang
- Peking University Ditan Teaching Hospital, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Bei Li
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Meiqing Jiang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Mengyuan Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Na Chen
- Peking University Ditan Teaching Hospital, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Meiju Deng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chuan Song
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Danying Chen
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liang Wu
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jiang Xiao
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hongyuan Liang
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hongxin Zhao
- Peking University Ditan Teaching Hospital, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yaxian Kong, ; Hongxin Zhao,
| | - Yaxian Kong
- Peking University Ditan Teaching Hospital, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yaxian Kong, ; Hongxin Zhao,
| |
Collapse
|
39
|
LEVI LI, SHARMA S, SCHLEISS MR, FURRER H, NIXON DE, BLACKSTAD M, HERNANDEZ-ALVARADO N, DWYER DE, BORGES AH, LANE HC, LUNDGREN J, NEATON JD, MOLINA JM, INSIGHT and ANRS study groups. Cytomegalovirus viremia and risk of disease progression and death in HIV-positive patients starting antiretroviral therapy. AIDS 2022; 36:1265-1272. [PMID: 35442221 PMCID: PMC9980657 DOI: 10.1097/qad.0000000000003238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The aim of this study was to assess the prevalence of cytomegalovirus (CMV) viremia in HIV-positive patients starting antiretroviral therapy (ART) and to evaluate its impact on clinical outcomes. DESIGN A retrospective analysis of four clinical trials (INSIGHT FIRST, SMART, START, and ANRS REFLATE TB). METHODS Stored plasma samples from participants were used to measure CMV viremia at baseline prior to initiating ART and at visits through 1 year of follow-up after ART initiation. CMV viremia was measured centrally using a quantitative PCR assay. Within FIRST, associations of CMV viremia at baseline and through 8 months of ART were examined with a composite clinical outcome of AIDS, serious non-AIDS events, or death using Cox proportional hazards regression. RESULTS Samples from a total of 3176 participants, 1169 from FIRST, 137 from ANRS REFLATE TB, 54 from SMART, and 1816 from START were available with baseline CMV viremia prevalence of 17, 26, 0, and 1%, respectively. Pooled across trials, baseline CMV viremia was associated with low CD4 + T-cell counts and high HIV RNA levels. In FIRST, CMV viremia was detected in only 5% of participants between baseline and month 8. After adjustment for CD4 + T-cell count and HIV RNA levels, hazard ratios for risk of clinical outcomes was 1.15 (0.86-1.54) and 2.58 (1.68-3.98) in FIRST participants with baseline and follow-up CMV viremia, respectively. CONCLUSION Baseline CMV viremia in HIV-positive patients starting ART is associated with advanced infection and only persistent CMV viremia after ART initiation is associated with a higher risk of morbidity and mortality.
Collapse
Affiliation(s)
- Laura I. LEVI
- University of Paris and Hôpital Saint-Louis and Lariboisière, INSERM U944, Paris, France
| | - Shweta SHARMA
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Mark R. SCHLEISS
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hansjakob FURRER
- Bern University Hospital, Inselspital, University Hospital of Bern, Bern, Switzerland
| | | | - Mark BLACKSTAD
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | - Dominic E. DWYER
- Westmead Hospital and University of Sydney, Westmead, NSW, Australia
| | | | - H. Clifford LANE
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jens LUNDGREN
- CHIP, Department of Infectious Diseases, Copenhagen, Denmark
| | - James D. NEATON
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Jean-Michel MOLINA
- University of Paris and Hôpital Saint-Louis and Lariboisière, INSERM U944, Paris, France
| | | |
Collapse
|
40
|
Ahodantin J, Nio K, Funaki M, Zhai X, Wilson E, Kottilil S, Cheng L, Li G, Su L. Type I interferons and TGF-β cooperate to induce liver fibrosis during HIV-1 infection under antiretroviral therapy. JCI Insight 2022; 7:e152738. [PMID: 35639478 PMCID: PMC9310524 DOI: 10.1172/jci.insight.152738] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
Liver diseases have become a major comorbidity health concern for people living with HIV-1 (PLWH) treated with combination antiretroviral therapy (cART). To investigate if HIV-1 infection and cART interact to lead to liver diseases, humanized mice reconstituted with progenitor cells from human fetal livers were infected with HIV-1 and treated with cART. We report here that chronic HIV-1 infection with cART induced hepatitis and liver fibrosis in humanized mice, associated with accumulation of M2-like macrophages (M2LMs), elevated TGF-β, and IFN signaling in the liver. Interestingly, IFN-I and TGF-β cooperatively activated human hepatic stellate cells (HepSCs) in vitro. Mechanistically, IFN-I enhanced TGF-β-induced SMAD2/3 activation in HepSCs. Finally, blockade of IFN-I signaling reversed HIV/cART-induced liver diseases in humanized mice. Consistent with the findings in humanized mice with HIV-1 and cART, we detected elevated markers of liver injury, M2LMs, and of IFN signaling in blood specimens from PLWH compared with those of healthy individuals. These findings identify the IFN-I/M2LM/HepSC axis in HIV/cART-induced liver diseases and suggest that inhibiting IFN-I signaling or M2LM may provide a novel therapeutic strategy for treating HIV/cART-associated liver diseases in PLWH treated with antiretroviral therapy.
Collapse
Affiliation(s)
- James Ahodantin
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kouki Nio
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Masaya Funaki
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xuguang Zhai
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eleanor Wilson
- Division of Clinical Care and Research, Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Shyamasundaran Kottilil
- Division of Clinical Care and Research, Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Liang Cheng
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guangming Li
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lishan Su
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, Departments of Pharmacology and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
41
|
Zaongo SD, Ouyang J, Chen Y, Jiao YM, Wu H, Chen Y. HIV Infection Predisposes to Increased Chances of HBV Infection: Current Understanding of the Mechanisms Favoring HBV Infection at Each Clinical Stage of HIV Infection. Front Immunol 2022; 13:853346. [PMID: 35432307 PMCID: PMC9010668 DOI: 10.3389/fimmu.2022.853346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Human immunodeficiency virus (HIV) selectively targets and destroys the infection-fighting CD4+ T-lymphocytes of the human immune system, and has a life cycle that encompasses binding to certain cells, fusion to that cell, reverse transcription of its genome, integration of its genome into the host cell DNA, replication of the HIV genome, assembly of the HIV virion, and budding and subsequent release of free HIV virions. Once a host is infected with HIV, the host’s ability to competently orchestrate effective and efficient immune responses against various microorganisms, such as viral infections, is significantly disrupted. Without modern antiretroviral therapy (ART), HIV is likely to gradually destroy the cellular immune system, and thus the initial HIV infection will inexorably evolve into acquired immunodeficiency syndrome (AIDS). Generally, HIV infection in a patient has an acute phase, a chronic phase, and an AIDS phase. During these three clinical stages, patients are found with relatively specific levels of viral RNA, develop rather distinctive immune conditions, and display unique clinical manifestations. Convergent research evidence has shown that hepatitis B virus (HBV) co-infection, a common cause of chronic liver disease, is fairly common in HIV-infected individuals. HBV invasion of the liver can be facilitated by HIV infection at each clinical stage of the infection due to a number of contributing factors, including having identical transmission routes, immunological suppression, gut microbiota dysbiosis, poor vaccination immune response to hepatitis B immunization, and drug hepatotoxicity. However, there remains a paucity of research investigation which critically describes the influence of the different HIV clinical stages and their consequences which tend to favor HBV entrenchment in the liver. Herein, we review advances in the understanding of the mechanisms favoring HBV infection at each clinical stage of HIV infection, thus paving the way toward development of potential strategies to reduce the prevalence of HBV co-infection in the HIV-infected population.
Collapse
Affiliation(s)
- Silvere D. Zaongo
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaling Chen
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Yan-Mei Jiao
- Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hao Wu
- Department of Infectious Diseases, You’an Hospital, Capital Medical University, Beijing, China
| | - Yaokai Chen
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- *Correspondence: Yaokai Chen,
| |
Collapse
|
42
|
Zaongo SD, Harypursat V, Chen Y. Single-Cell Sequencing Facilitates Elucidation of HIV Immunopathogenesis: A Review of Current Literature. Front Immunol 2022; 13:828860. [PMID: 35185920 PMCID: PMC8850777 DOI: 10.3389/fimmu.2022.828860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022] Open
Abstract
Knowledge gaps remain in the understanding of HIV disease establishment and progression. Scientists continue to strive in their endeavor to elucidate the precise underlying immunopathogenic mechanisms of HIV-related disease, in order to identify possible preventive and therapeutic targets. A useful tool in the quest to reveal some of the enigmas related to HIV infection and disease is the single-cell sequencing (scRNA-seq) technique. With its proven capacity to elucidate critical processes in cell formation and differentiation, to decipher critical hematopoietic pathways, and to understand the regulatory gene networks that predict immune function, scRNA-seq is further considered to be a potentially useful tool to explore HIV immunopathogenesis. In this article, we provide an overview of single-cell sequencing platforms, before delving into research findings gleaned from the use of single cell sequencing in HIV research, as published in recent literature. Finally, we describe two important avenues of research that we believe should be further investigated using the single-cell sequencing technique.
Collapse
Affiliation(s)
- Silvere D Zaongo
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
43
|
Yang Y, Iwasaki A. Impact of Chronic HIV Infection on SARS-CoV-2 Infection, COVID-19 Disease and Vaccines. Curr HIV/AIDS Rep 2022; 19:5-16. [PMID: 34843064 PMCID: PMC8628277 DOI: 10.1007/s11904-021-00590-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has developed into a global pandemic that affect the health of hundreds of millions worldwide. In particular, SARS-CoV-2 infection in people with chronic human immune deficiency virus (HIV) infection is of concern, due to their already immunocompromised status. Yet, whether and how the immunological changes brought about by HIV will affect the immune responses against SARS-CoV-2 acute infection and impact the effectiveness of vaccines remain unclear. We discuss the intersection of COVID-19 in HIV-infected individuals. RECENT FINDINGS People living with HIV (PLWH) may be at increased risk of severe SARS-CoV-2 mediated disease complication due to functional impairment of the immune system and persistent inflammation, which can be ameliorated by antiretroviral therapy. Importantly, limited data suggest that current approved vaccines may be safe and efficacious in PLWH. To address remaining questions and supplement limited experimental evidence, more studies examining the interplay between HIV and SARS-CoV-2 through their impact on the host immune system are required.
Collapse
Affiliation(s)
- Yexin Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
44
|
Noiman A, Esber A, Wang X, Bahemana E, Adamu Y, Iroezindu M, Kiweewa F, Maswai J, Owuoth J, Maganga L, Ganesan A, Maves RC, Lalani T, Colombo RE, Okulicz JF, Polyak C, Crowell TA, Ake JA, Agan BK. Clinical factors and outcomes associated with immune non-response among virally suppressed adults with HIV from Africa and the United States. Sci Rep 2022; 12:1196. [PMID: 35075147 PMCID: PMC8786968 DOI: 10.1038/s41598-022-04866-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 12/24/2021] [Indexed: 11/21/2022] Open
Abstract
A significant minority of people living with HIV (PLWH) achieve viral suppression (VS) on antiretroviral therapy (ART) but do not regain healthy CD4 counts. Clinical factors affecting this immune non-response (INR) and its effect on incident serious non-AIDS events (SNAEs) have been challenging to understand due to confounders that are difficult to control in many study settings. The U.S. Military HIV Natural History Study (NHS) and African Cohort Study (AFRICOS). PLWH with sustained VS (< 400 copies/ml for at least two years) were evaluated for INR (CD4 < 350 cells/µl at the time of sustained VS). Logistic regression estimated adjusted odds ratios (aORs) and 95% confidence intervals (CIs) for factors associated with INR. Cox proportional hazards regression produced adjusted hazard ratios (aHRs) for factors associated with incident SNAE after sustained VS. INR prevalence was 10.8% and 25.8% in NHS and AFRICOS, respectively. Higher CD4 nadir was associated with decreased odds of INR (aOR = 0.34 [95% CI 0.29, 0.40] and aOR = 0.48 [95% CI 0.40, 0.57] per 100 cells/µl in NHS and AFRICOS, respectively). After adjustment, INR was associated with a 61% increase in relative risk of SNAE [95% CI 1.12, 2.33]. Probability of "SNAE-free" survival at 15 years since sustained VS was approximately 20% lower comparing those with and without INR; nearly equal to the differences observed by 15-year age groups. CD4 monitoring before and after VS is achieved can help identify PLWH at risk for INR. INR may be a useful clinical indicator of future risk for SNAEs.
Collapse
Affiliation(s)
- Adi Noiman
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA. .,Infectious Disease Clinical Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, 11300 Rockville Pike, Suite 600, Rockville, MD, 20852, USA.
| | - Allahna Esber
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Xun Wang
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Emmanuel Bahemana
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Henry M. Jackson Foundation MRI, Mbeya, Tanzania
| | - Yakubu Adamu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,U.S. Army Medical Research Directorate-Africa, Nairobi, Kenya.,Henry M. Jackson Foundation MRI, Abuja, Nigeria
| | - Michael Iroezindu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,U.S. Army Medical Research Directorate-Africa, Nairobi, Kenya.,Henry M. Jackson Foundation MRI, Abuja, Nigeria
| | | | - Jonah Maswai
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Kenya Medical Research Institute, Nairobi, Kenya.,Henry M. Jackson Foundation MRI, Kericho, Kenya
| | - John Owuoth
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Kenya Medical Research Institute, Nairobi, Kenya.,Henry M. Jackson Foundation MRI, Kisumu, Kenya
| | - Lucas Maganga
- National Institute of Medical Research-Mbeya Medical Research Centre, Mbeya, Tanzania
| | - Anuradha Ganesan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Ryan C Maves
- Naval Medical Center San Diego, San Diego, CA, USA
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | - Rhonda E Colombo
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Madigan Army Medical Center, Joint Base Lewis-McChord, WA, USA
| | - Jason F Okulicz
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Brooke Army Medical Center, San Antonio, TX, USA
| | - Christina Polyak
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Trevor A Crowell
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Division of Infectious Diseases, Department of Medicine, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Julie A Ake
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Division of Infectious Diseases, Department of Medicine, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brian K Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.
| |
Collapse
|
45
|
Zaongo SD, Sun F, Chen Y. Are HIV-1-Specific Antibody Levels Potentially Useful Laboratory Markers to Estimate HIV Reservoir Size? A Review. Front Immunol 2021; 12:786341. [PMID: 34858439 PMCID: PMC8632222 DOI: 10.3389/fimmu.2021.786341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
Despite the benefits achieved by the widespread availability of modern antiretroviral therapy (ART), HIV RNA integration into the host cell genome is responsible for the creation of latent HIV reservoirs, and represents a significant impediment to completely eliminating HIV infection in a patient via modern ART alone. Several methods to measure HIV reservoir size exist; however, simpler, cheaper, and faster tools are required in the quest for total HIV cure. Over the past few years, measurement of HIV-specific antibodies has evolved into a promising option for measuring HIV reservoir size, as they can be measured via simple, well-known techniques such as the western blot and enzyme-linked immunosorbent assay (ELISA). In this article, we re-visit the dynamic evolution of HIV-1-specific antibodies and the factors that may influence their levels in the circulation of HIV-positive individuals. Then, we describe the currently-known relationship between HIV-1-specific antibodies and HIV reservoir size based on study of data from contemporary literature published during the past 5 years. We conclude by highlighting current trends, and discussing the individual HIV-specific antibody that is likely to be the most reliable antibody for potential future utilization for quantification of HIV reservoir size.
Collapse
Affiliation(s)
- Silvere D Zaongo
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Feng Sun
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Division of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
46
|
Mbuya W, Held K, Mcharo RD, Haule A, Mhizde J, Mnkai J, Mahenge A, Mwakatima M, Sembo M, Mwalongo W, Agrea P, Hoelscher M, Maboko L, Saathoff E, Geisenberger O, Rwegoshora F, Torres L, Koup RA, Kroidl A, Chachage M, Geldmacher C. Depletion of Human Papilloma Virus E6- and E7-Oncoprotein-Specific T-Cell Responses in Women Living With HIV. Front Immunol 2021; 12:742861. [PMID: 34759925 PMCID: PMC8573218 DOI: 10.3389/fimmu.2021.742861] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background Cervical cancer - caused by persistent High Risk Human Papilloma Virus (HR HPV) infections - is the second most common cancer affecting women globally. HIV infection increases the risk for HPV persistence, associated disease progression and malignant cell transformation. We therefore hypothesized that this risk increase is directly linked to HIV infection associated dysfunction or depletion of HPV-oncoprotein-specific T-cell responses. Methods The 2H study specifically included HIV+ and HIV- women with and without cervical lesions and cancer to analyze HPV oncogene-specific T cell responses in relation to HPV infection, cervical lesion status and HIV status. Oncoprotein E6 and E7 specific T-cell responses were quantified for the most relevant types HPV16, 18 and 45 and control antigens (CMV-pp65) and M.tb-PPD in 373 women, using fresh peripheral blood mononuclear cells in an IFN-γ release ELISpot assay. Results Overall, systemic E6- and E7-oncoprotein-specific T-cell responses were infrequent and of low magnitude, when compared to CMV-pp65 and M.tb-PPD (p < 0.001 for all HR HPV types). Within HIV negative women infected with either HPV16, 18 or 45, HPV16 infected women had lowest frequency of autologous-type-E6/E7-specific T-cell responses (33%, 16/49), as compared to HPV18 (46% (6/13), p = 0.516) and HPV45 (69% (9/13), p = 0.026) infected women. Prevalent HPV18 and 45, but not HPV16 infections were linked to detectable oncoprotein-specific T-cell responses, and for these infections, HIV infection significantly diminished T-cell responses targeting the autologous infecting genotype. Within women living with HIV, low CD4 T-cell counts, detectable HIV viremia as well as cancerous and precancerous lesions were significantly associated with depletion of HPV oncoprotein-specific T-cell responses. Discussion Depletion of HPV-oncoprotein-specific T-cell responses likely contributes to the increased risk for HR HPV persistence and associated cancerogenesis in women living with HIV. The low inherent immunogenicity of HPV16 oncoproteins may contribute to the exceptional potential for cancerogenesis associated with HPV16 infections.
Collapse
Affiliation(s)
- Wilbert Mbuya
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ruby D Mcharo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Antelmo Haule
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Jacklina Mhizde
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Jonathan Mnkai
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Anifrid Mahenge
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Maria Mwakatima
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Margareth Sembo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Wolfram Mwalongo
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Peter Agrea
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Leonard Maboko
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Tanzania Commission for AIDS (TACAIDS), Dar es Salaam, Tanzania
| | - Elmar Saathoff
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Otto Geisenberger
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - France Rwegoshora
- Pathology Department, Mbeya Zonal Referral Hospital, Mbeya, Tanzania
| | - Liset Torres
- Pathology Department, Mbeya Zonal Referral Hospital, Mbeya, Tanzania
| | - Richard A Koup
- Vaccine Research Centre, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Arne Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Mkunde Chachage
- National Institute for Medical Research - Mbeya Medical Research Centre (NIMR-MMRC), Mbeya, Tanzania.,Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,Microbiology and Immunology Department, University of Dar es Salaam -Mbeya College of Health and Allied Sciences (UDSM-MCHAS), Mbeya, Tanzania
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
47
|
Vlasova VV, Saidakova EV, Korolevskaya LB, Shmagel NG, Chereshnev VA, Shmagel KV. Metabolic Features of Activated Memory CD4 + T-Cells Derived from HIV-Infected Immunological Non-responders to Highly Active Antiretroviral Therapy. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2021; 501:206-209. [PMID: 34962608 DOI: 10.1134/s0012496621060090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022]
Abstract
Immunological non-responders (INR) are HIV-infected subjects that fail to restore CD4+ T-cell counts despite undetectable HIV viral load, which is controlled by highly active antiretroviral therapy (HAART). In INR, impaired immune restoration is linked to low-productive proliferation of memory CD4+ T-lymphocytes. Taking into account that T-cell ability to divide depends on the activity of metabolic pathways, we aimed to determine rates of mitochondrial respiration and glycolysis in memory CD4+ T-cells of INR. Two groups of HIV-infected HAART-treated patients were studied: immunological non-responders and subjects with an adequate immunological response to therapy (immunological responders - IR). Control (C) group comprised uninfected volunteers. In both groups of HIV-infected patients glycolytic activity of memory CD4+ T-cells was lower than that in C. Mitochondrial respiration rate in memory CD4+ T-cells derived from IR was comparable to that of C at basal state, however, after stimulation IR failed to reach the values of uninfected subjects. INR had the lowest mitochondrial respiration rate both at basal state and after stimulation. Taken together, the data presented herein demonstrate that low regenerative potential of memory CD4+ T-cells derived from INR might be linked to diminished lymphocytes' metabolic activity.
Collapse
Affiliation(s)
- V V Vlasova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center UB RAS, 614081, Perm, Russia.
| | - E V Saidakova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center UB RAS, 614081, Perm, Russia
| | - L B Korolevskaya
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center UB RAS, 614081, Perm, Russia
| | - N G Shmagel
- Perm Regional Center for Prevention and Control of AIDS and Infectious Diseases, 614065, Perm, Russia
| | - V A Chereshnev
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center UB RAS, 614081, Perm, Russia.,Institute of Immunology and Physiology, 620219, Yekaterinburg, Russia
| | - K V Shmagel
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center UB RAS, 614081, Perm, Russia
| |
Collapse
|
48
|
Shen C, Li J, Lian Y, Lan H, Pu F, Zhang W, Kong L, Liu J. Chinese herbal medicine for incomplete immune reconstruction in patients with AIDS undergoing antiretroviral treatment: A systematic review of randomized trials. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [DOI: 10.1016/j.jtcms.2021.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
49
|
Anes E, Azevedo-Pereira JM, Pires D. Cathepsins and Their Endogenous Inhibitors in Host Defense During Mycobacterium tuberculosis and HIV Infection. Front Immunol 2021; 12:726984. [PMID: 34421929 PMCID: PMC8371317 DOI: 10.3389/fimmu.2021.726984] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/22/2021] [Indexed: 01/15/2023] Open
Abstract
The moment a very old bacterial pathogen met a young virus from the 80's defined the beginning of a tragic syndemic for humanity. Such is the case for the causative agent of tuberculosis and the human immunodeficiency virus (HIV). Syndemic is by definition a convergence of more than one disease resulting in magnification of their burden. Both pathogens work synergistically contributing to speed up the replication of each other. Mycobacterium tuberculosis (Mtb) and HIV infections are in the 21st century among the leaders of morbidity and mortality of humankind. There is an urgent need for development of new approaches for prevention, better diagnosis, and new therapies for both infections. Moreover, these approaches should consider Mtb and HIV as a co-infection, rather than just as separate problems, to prevent further aggravation of the HIV-TB syndemic. Both pathogens manipulate the host immune responses to establish chronic infections in intracellular niches of their host cells. This includes manipulation of host relevant antimicrobial proteases such as cathepsins or their endogenous inhibitors. Here we discuss recent understanding on how Mtb and HIV interact with cathepsins and their inhibitors in their multifactorial functions during the pathogenesis of both infections. Particularly we will address the role on pathogen transmission, during establishment of intracellular chronic niches and in granuloma clinical outcome and tuberculosis diagnosis. This area of research will open new avenues for the design of innovative therapies and diagnostic interventions so urgently needed to fight this threat to humanity.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
50
|
Ding C, Patel D, Ma Y, Mann JFS, Wu J, Gao Y. Employing Broadly Neutralizing Antibodies as a Human Immunodeficiency Virus Prophylactic & Therapeutic Application. Front Immunol 2021; 12:697683. [PMID: 34354709 PMCID: PMC8329590 DOI: 10.3389/fimmu.2021.697683] [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: 04/20/2021] [Accepted: 07/05/2021] [Indexed: 11/18/2022] Open
Abstract
Despite the discovery that the human immunodeficiency virus 1 (HIV-1) is the pathogen of acquired immunodeficiency syndrome (AIDS) in 1983, there is still no effective anti-HIV-1 vaccine. The major obstacle to the development of HIV-1 vaccine is the extreme diversity of viral genome sequences. Nonetheless, a number of broadly neutralizing antibodies (bNAbs) against HIV-1 have been made and identified in this area. Novel strategies based on using these bNAbs as an efficacious preventive and/or therapeutic intervention have been applied in clinical. In this review, we summarize the recent development of bNAbs and its application in HIV-1 acquisition prevention as well as discuss the innovative approaches being used to try to convey protection within individuals at risk and being treated for HIV-1 infection.
Collapse
Affiliation(s)
- Chengchao Ding
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Darshit Patel
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Yunjing Ma
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Jamie F S Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Jianjun Wu
- Department of AIDS Research, Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Yong Gao
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| |
Collapse
|