1
|
Zharikov Y, Shitova A, Melnikova P, Voloshin I, Orliuk M, Olsufieva A, Pontes-Silva A, Zharikova T. Autoantibody-mediated disorders of the central and peripheral nervous system: Overview Infection. J Neuroimmunol 2025; 403:578616. [PMID: 40245466 DOI: 10.1016/j.jneuroim.2025.578616] [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: 10/25/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
HIV-associated neurocognitive disorders are a common manifestation of HIV infection, affecting more than half of HIV-infected individuals, including those receiving targeted antiviral therapy. A common feature of the course of HIV infection during therapy is large-scale immune responses in the brain. Several pathways are involved in the neuropathogenesis of HIV infection: Cellular entry, inflammatory processes in microglia, activation of astroglia, myeloid cells, and damage to brain vessels leading to neurocirculatory disorders. Data on vascular diseases that influence the development of neurocognitive impairment in HIV-positive patients will also be examined, as well as better intervention strategies for complex neurocognitive disorders and neurodegenerative processes in HIV infection.
Collapse
Affiliation(s)
- Yury Zharikov
- FSAEI HE I.M. Sechenov First MSMU of MOH of Russia (Sechenovskiy University), Moscow, Russia
| | | | - Polina Melnikova
- FSAEI HE I.M. Sechenov First MSMU of MOH of Russia (Sechenovskiy University), Moscow, Russia
| | - Ilya Voloshin
- FSAEI HE I.M. Sechenov First MSMU of MOH of Russia (Sechenovskiy University), Moscow, Russia
| | | | | | - André Pontes-Silva
- Postgraduate Program in Physical Therapy, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil..
| | - Tatiana Zharikova
- FSAEI HE I.M. Sechenov First MSMU of MOH of Russia (Sechenovskiy University), Moscow, Russia
| |
Collapse
|
2
|
Tôrres PPBF, Meneses GC, Lima LAL, Lopes NC, de Araújo LM, Araújo KMDR, de Souza RN, Domingues-da-Silva RDO, Martins AMC, Daher EDF, da Silva Junior GB. Angiopoietin-2 and endothelial damage associated with viral load in untreated people living with HIV. Int J STD AIDS 2025; 36:498-505. [PMID: 40019463 DOI: 10.1177/09564624251323681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
BackgroundHIV infection is associated with various types of endothelial damage. Early and accurate diagnosis of endothelial dysfunction may help prevent adverse outcomes. For the first time, this cross-sectional study aims to investigate the effects of increased viral load on endothelial damage and nephrinuria in untreated people living with HIV.MethodsPeople living with HIV not on treatment were included. Laboratory parameters, such as viral load and CD4 count were collected. Participants were divided into three groups according to the tertiles of viral load: low, moderate and high viral load groups. Urinary nephrin, vascular cell adhesion molecule 1 (VCAM-1), angiopoietin-2 and syndecan-1 were quantified by sandwich ELISA assays kits.ResultsA total of 49 patients were examined. The high viral load group (3rd tertile) exhibited a greater frequency of nephrinuria and significantly higher levels of syndecan-1 (58 [48-93] vs 43 [38-65] ng/mL, p = .049) and angiopoietin-2 (2.58 [1.5-3.06] vs 1.34 [0.89-1.76] ng/mL, p = .035). Syndecan-1 levels positively correlated with viral load (rho = 0.452, p = .006), while angiopoietin-2 was positively correlated with viral load (rho = 0.529, p = .001) and inversely correlated with CD4 count (rho = -0.493, p = .003). No significant associations were found for VCAM-1 levels.ConclusionUntreated people living with HIV with elevated viral loads exhibited more pronounced endothelial damage and vascular inflammation, emphasizing the critical importance of early initiation of antiretroviral treatment to mitigate these complications. However, the study's findings should be interpreted with caution due to the small sample size, which may limit the generalizability of the results. Future studies with larger cohorts are needed to confirm these findings.
Collapse
Affiliation(s)
| | - Gdayllon Cavalcante Meneses
- Clinical and Toxicological Analysis Department, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Lana Andrade Lucena Lima
- Public Health and Medical Sciences Graduate Programs, Federal University of Fortaleza, Fortaleza, Brazil
| | - Nicole Coelho Lopes
- Pharmacology Post-Graduate Program, Federal University of Ceará, Fortaleza, Brazil
| | - Leticia Machado de Araújo
- Public Health and Medical Sciences Graduate Programs, Federal University of Fortaleza, Fortaleza, Brazil
| | | | - Rayane Nascimento de Souza
- Medical Sciences Post-Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará. Fortaleza, Ceará, Brazil
| | - Raoni de Oliveira Domingues-da-Silva
- Medical Sciences Post-Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará. Fortaleza, Ceará, Brazil
| | - Alice Maria Costa Martins
- Clinical and Toxicological Analysis Department, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Elizabeth De Francesco Daher
- Medical Sciences Post-Graduate Program, Department of Internal Medicine, School of Medicine, Federal University of Ceará. Fortaleza, Ceará, Brazil
| | - Geraldo Bezerra da Silva Junior
- Collective Health Post-Graduate Program, School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Brazil
| |
Collapse
|
3
|
Stephens VR, Ameli S, Major AS, Wanjalla CN. Mouse Models of HIV-Associated Atherosclerosis. Int J Mol Sci 2025; 26:3417. [PMID: 40244289 PMCID: PMC11989901 DOI: 10.3390/ijms26073417] [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: 03/04/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Several factors are implicated in the pathogenesis of CVD, and efforts have been made to reduce traditional risks, yet CVD remains a complex burden. Notably, people living with HIV (PLWH) are twice as likely to develop CVD compared to persons without HIV (PWoH). Intensive statin therapy, the first-line treatment to prevent cardiovascular events, is effective at reducing morbidity and mortality. However, statin therapy has not reduced the overall prevalence of CVD. Despite antiretroviral therapy (ART), and new guidelines for statin use, PLWH have persistent elevation of inflammatory markers, which is suggested to be a bigger driver of future cardiovascular events than low-density lipoprotein. Herein, we have summarized the development of atherosclerosis and highlighted mouse models of atherosclerosis in the presence and absence of HIV. Since most mouse strains have several mechanisms that are atheroprotective, researchers have developed mouse models to study CVD using dietary and genetic manipulations. In evaluating the current methodologies for studying HIV-associated atherosclerosis, we have detailed the benefits of integrating multi-omics analyses, genetic manipulations, and immune cell profiling within mouse models. These advanced approaches significantly enhance our capacity to address critical gaps in understanding the immune mechanisms driving CVD, including in the context of HIV.
Collapse
Affiliation(s)
- Victoria R. Stephens
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amy S. Major
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Tennessee Valley Health System, Department of Veterans Affairs, Nashville, TN 37212, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Shi M, Feinstein MJ. Immune Dysregulation in Ischemic Heart Disease Among Individuals with Human Immunodeficiency Virus. Heart Fail Clin 2025; 21:227-239. [PMID: 40107801 PMCID: PMC11960833 DOI: 10.1016/j.hfc.2024.12.005] [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] [Indexed: 03/22/2025]
Abstract
Individuals with human immunodeficiency virus (HIV) experience an elevated risk of ischemic heart disease and related cardiovascular sequelae. This is due to a combination of factors including traditional comorbidities, adverse effects of antiretroviral therapy, low-level viremia, viral coinfection, mucosal injury, and chronic immune activation and dysregulation. Understanding the underlying mechanisms, especially as they relate to inflammation, has implications for prevention, diagnostics, and therapeutics. This review highlights some of the foundational and recent literature on clinical phenotypes, diagnostic tools, and promising pharmacotherapies, along with future directions for translational research and clinical implementation.
Collapse
Affiliation(s)
- Meilynn Shi
- Division of Cardiology, Department of Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew J Feinstein
- Division of Cardiology, Department of Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
5
|
Chee YJ, Dalan R, Cheung C. The Interplay Between Immunity, Inflammation and Endothelial Dysfunction. Int J Mol Sci 2025; 26:1708. [PMID: 40004172 PMCID: PMC11855323 DOI: 10.3390/ijms26041708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The endothelium is pivotal in multiple physiological processes, such as maintaining vascular homeostasis, metabolism, platelet function, and oxidative stress. Emerging evidence in the past decade highlighted the immunomodulatory function of endothelium, serving as a link between innate, adaptive immunity and inflammation. This review examines the regulation of the immune-inflammatory axis by the endothelium, discusses physiological immune functions, and explores pathophysiological processes leading to endothelial dysfunction in various metabolic disturbances, including hyperglycemia, obesity, hypertension, and dyslipidaemia. The final section focuses on the novel, repurposed, and emerging therapeutic targets that address the immune-inflammatory axis in endothelial dysfunction.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| |
Collapse
|
6
|
Mielcarska MB, Rouse BT. Viruses and the Brain-A Relationship Prone to Trouble. Viruses 2025; 17:203. [PMID: 40006958 PMCID: PMC11860391 DOI: 10.3390/v17020203] [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: 01/06/2025] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Neurological disorders, some of which are associated with viral infections, are growing due to the aging and expanding population. Despite strong defenses of the central nervous system, some viruses have evolved ways to breach them, which often result in dire consequences. In this review, we recount the various ways by which different viruses can enter the CNS, and we describe the consequences of such invasions. Consequences may manifest as acute disease, such as encephalitis, meningitis, or result in long-term effects, such as neuromuscular dysfunction, as occurs in poliomyelitis. We discuss evidence for viral involvement in the causation of well-known chronic neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as vascular dementia in the elderly. We also describe the approaches currently available to control a few of the neural viral infections. These include antivirals that are effective against human immunodeficiency virus and herpes simplex virus, as well as vaccines valuable for controlling rabies virus, poliomyelitis virus, and some flavivirus infections. There is an urgent need to better understand, at a molecular level, how viruses contribute to acute and, especially, chronic neurological diseases and to develop more precise and effective vaccines and therapies.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
7
|
Jalilian S, Vasei M, Garshasbi A, Nabavi SS, Bastani MN. Viral intruders in the heart: A review of RNA viruses and their role in cardiac disorders. APMIS 2025; 133:e13500. [PMID: 39530180 DOI: 10.1111/apm.13500] [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: 01/10/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Viral cardiac diseases have a significant impact on global health, and RNA viruses play a crucial role in their pathogenesis. This literature review aims to provide a comprehensive understanding of the complex relationship between RNA viruses and cardiac diseases, focusing on the molecular processes and clinical implications of these interactions. The paper begins by discussing the various RNA viruses that have been linked to cardiac infections. Subsequently, the study explores the mechanisms through which RNA viruses can cause cardiac injury, including direct viral invasion, immune-mediated responses, and molecular mimicry. The review extensively examines the intricate interplay between the host immune system and RNA viruses, shedding light on both protective and harmful immune responses. Additionally, it investigates the role of viral persistence and chronic inflammation in the long-term effects on cardiac health. The thorough analysis presented not only enhances our scientific understanding of how RNA viruses contribute to the development of cardiac diseases but also highlights potential avenues for future research and breakthroughs in this field. Given the significant global health threat posed by viral cardiac disorders, unraveling the molecular foundations of these diseases is essential for advancing diagnostic capabilities and therapeutic interventions.
Collapse
Affiliation(s)
- Shahram Jalilian
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mona Vasei
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ashkan Garshasbi
- Immunology Department of Immunology and Microbiology, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Salaheddin Nabavi
- Department of General Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Navid Bastani
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Ladinsky MS, Zhu L, Ullah I, Uchil PD, Kumar P, Kay MS, Bjorkman PJ. Electron tomography visualization of HIV-1 virions trapped by fusion inhibitors to host cells in infected tissues. J Virol 2024; 98:e0143224. [PMID: 39475277 PMCID: PMC11575291 DOI: 10.1128/jvi.01432-24] [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: 08/16/2024] [Accepted: 09/26/2024] [Indexed: 11/06/2024] Open
Abstract
HIV-1 delivers its genetic material to infect a cell after fusion of the viral and host cell membranes, which takes place after the viral envelope (Env) binds host receptor and co-receptor proteins. Binding of host receptor CD4 to Env results in conformational changes that allow interaction with a host co-receptor (CCR5 or CXCR4). Further conformational rearrangements result in an elongated pre-hairpin intermediate structure in which Env is anchored to the viral membrane by its transmembrane region and to the host cell membrane by its fusion peptide. Although budding virions can be readily imaged by electron tomography (ET) of HIV-1-infected tissues and cultured cells, virions that are fusing (attached to host cells via pre-hairpin intermediates) are not normally visualized, perhaps because the process of membrane fusion is too fast to capture by ET. To image virions during fusion, we used fusion inhibitors to prevent downstream conformational changes in Env that lead to membrane fusion, thereby trapping HIV-1 virions linked to target cells by pre-hairpin intermediates. ET of HIV-1 pseudovirions bound to CD4+/CCR5+ TZM-bl cells revealed presumptive pre-hairpin intermediates as 2-4 narrow spokes linking a virion to the cell surface. To extend these results to a more physiological setting, we used ET to image tissues and organs derived from humanized bone marrow/liver/thymus mice infected with HIV-1 and then treated with CPT31, a high-affinity D-peptide fusion inhibitor linked to cholesterol. Trapped HIV-1 virions were found in all tissues studied (small intestine, mesenteric lymph nodes, spleen, and bone marrow), and spokes representing pre-hairpin intermediates linking trapped virions to cell surfaces were similar in structure and number to those seen in the previous pseudovirus and cultured cell ET study.IMPORTANCETrapped and untrapped HIV-1 virions, both mature and immature, were distinguished by localizing spokes via 3D tomographic reconstructions of HIV-1 infected and fusion-inhibitor-treated tissues of humanized mice. The findings of trapped HIV-1 virions in all tissues examined demonstrate a wide distribution of the CPT31 inhibitor, a desirable property for a potential therapeutic. In addition, the presence of virions trapped by spokes, particularly in vascular endothelial cells, demonstrates that the fusion inhibitors can be used as markers for potential HIV-1-target cells within tissues, facilitating the mapping of HIV-1 target cells within the complex cellular milieu of infected tissues.
Collapse
Affiliation(s)
- Mark S. Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Li Zhu
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Irfan Ullah
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Priti Kumar
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael S. Kay
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
9
|
Obare LM, Priest S, Ismail A, Mashayekhi M, Zhang X, Stolze LK, Sheng Q, Nthenge K, Vue Z, Neikirk K, Beasley HK, Gabriel C, Temu T, Gianella S, Mallal SA, Koethe JR, Hinton A, Bailin SS, Wanjalla CN. Cytokine and chemokine receptor profiles in adipose tissue vasculature unravel endothelial cell responses in HIV. J Cell Physiol 2024; 239:e31415. [PMID: 39263801 DOI: 10.1002/jcp.31415] [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: 03/10/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024]
Abstract
Chronic systemic inflammation significantly increases myocardial infarction risk in people living with HIV (PLWH). Endothelial cell dysfunction disrupts vascular homeostasis regulation, increasing the risk of vasoconstriction, inflammation, and thrombosis, contributing to cardiovascular disease. We aimed to characterize endothelial cell (EC) chemokines, cytokine, and chemokine receptors of PLWH, hypothesizing that in our cohort, glucose intolerance contributes to their differential expression implicated in endothelial dysfunction. Using single-cell transcriptomic analysis, we phenotyped chemokine and cytokine receptor expression on arterial ECs, capillary ECs, venous ECs, and vascular smooth muscle cells (VSMCs) in subcutaneous adipose tissue of 59 PLWH with and without glucose intolerance. Our results show that arterial and capillary ECs express significantly higher interferon and tumor necrosis factor (TNF) receptors than venous ECs and VSMCs. Venous ECs exhibited more interleukin (IL)1R1 and ACKR1 receptors, and VSMCs showed significant IL6R expression than arterial and capillary ECs. When stratified by group, arterial ECs from PLWH with glucose intolerance expressed significantly higher IL1R1, IL6R, CXCL12, CCL14, and ICAM2 transcripts than arterial ECs from PLWH without diabetes. Of the different vascular cell types studied, arterial ECs as a proportion of all ECs in adipose tissue were positively correlated with plasma fasting blood glucose. In contrast, venous ECs and VSMCs were positively correlated with plasma IL6. To directly assess the effect of plasma from PLWH on endothelial function, we cultured human arterial ECs (HAECs) in plasma-conditioned media from PLWH and performed bulk RNA sequencing. Plasma from PLWH stimulated ECs with the upregulation of genes that enrich for the oxidative phosphorylation and the TNF-α via NFK-β pathways. In conclusion, ECs in PLWH show heterogeneous cytokine and chemokine receptor expression, and arterial ECs were the most influenced by glucose intolerance. Further research must explicate cytokine and chemokine roles in EC dysfunction and identify biomarkers for disease progression and therapeutic response.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anas Ismail
- Department of Radiology, National Postgraduate Medical College of Nigeria, Lagos, Nigeria
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lindsey K Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kisyua Nthenge
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Curtis Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tecla Temu
- Division of Pathology, Harvard Medical College, Boston, Massachusetts, USA
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, California, USA
| | - Simon A Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - John R Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Samuel S Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Myburgh-Jacobsz CE, Botha-Le Roux S, Kotliar K, Wentzel A, Jacobs A, De Boever P, Goswami N, Strijdom H, Smith W. Retinal Vessel Functional Responses in South Africans Living With and Without HIV: The EndoAfrica-NWU Study. Microcirculation 2024; 31:e12878. [PMID: 39106121 DOI: 10.1111/micc.12878] [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: 08/27/2023] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVES The effects of HIV and antiretroviral therapy (ART) on microvascular function are poorly explored. We compared retinal vessel functional responses to flicker light-induced provocation (FLIP) in people living with HIV (PLWH) and people living without HIV (PLWoutH). METHODS We included 115 PLWH and 51 PLWoutH with a median age of 41 years. Treated PLWH received similar first-line fixed-dose combination ART. Clinical characteristics and retinal vessels functional responses to FLIP were compared in (a) PLWH and PLWoutH; and (b) PLWH groups stratified by the median of (i) CD4-count (511 cells/mm3), (ii) viral load (50 copies/mL), and (iii) ART duration (57.6 months). RESULTS PLWH were older, smoked more, and had a lower prevalence of hypertension than PLWoutH (p < 0.05). Almost 64% of PLWH were infected for more than 5 years. Retinal vessel responses to FLIP were similar between PLWH and PLWoutH after taking confounders into account. In addition, PLWH subgroups stratified according to immuno-virological status by CD4-count, viral load, and ART duration showed no differences in retinal vessel responses to FLIP. CONCLUSION Living with HIV and receiving ART were not associated with altered microvascular function as assessed with dynamic retinal vessel analysis in a South African case-control study.
Collapse
Affiliation(s)
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, Aachen University of Applied Sciences, Juelich, Germany
| | - Annemarie Wentzel
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Adriaan Jacobs
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Patrick De Boever
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Antwerp University Hospital (UZA), Edegem, Belgium
| | - Nandu Goswami
- Gravitational Physiology and Medicine Research Unit, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Center for Space and Aviation Health, College of Medicine, Mohammed bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Hans Strijdom
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
11
|
Ladinsky MS, Zhu L, Ullah I, Uchil PD, Kumar P, Kay MS, Bjorkman PJ. Electron tomography visualization of HIV-1 virions trapped by fusion inhibitors to host cells in infected tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608557. [PMID: 39229189 PMCID: PMC11370368 DOI: 10.1101/2024.08.19.608557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
HIV-1 delivers its genetic material to infect a cell after fusion of the viral and host cell membranes, which takes place after the viral envelope (Env) binds host receptor and co-receptor proteins. Binding of host receptor CD4 to Env results in conformational changes that allow interaction with a host co-receptor (CCR5 or CXCR4). Further conformational rearrangements result in an elongated pre-hairpin intermediate structure in which Env is anchored to the viral membrane by its transmembrane region and to the host cell membrane by its fusion peptide. Although budding virions can be readily imaged by electron tomography (ET) of HIV-1-infected tissues and cultured cells, virions that are fusing (attached to host cells via pre-hairpin intermediates) are not normally visualized, perhaps because the process of membrane fusion is too fast to capture by EM. To image virions during fusion, we used fusion inhibitors to prevent downstream conformational changes in Env that lead to membrane fusion, thereby trapping HIV-1 virions linked to target cells by prehairpin intermediates. ET of HIV-1 pseudovirions bound to CD4+/CCR5+ TZM-bl cells revealed presumptive pre-hairpin intermediates as 2-4 narrow spokes linking a virion to the cell surface. To extend these results to a more physiological setting, we used ET to image tissues and organs derived from humanized bone marrow, liver, thymus (BLT) mice infected with HIV-1 and then treated with CPT31, a high-affinity D-peptide fusion inhibitor linked to cholesterol. Trapped HIV-1 virions were found in all tissues studied (small intestine, mesenteric lymph nodes, spleen, and bone marrow), and spokes representing pre-hairpin intermediates linking trapped virions to cell surfaces were similar in structure and number to those seen in the previous pseudovirus and cultured cell ET study.
Collapse
Affiliation(s)
- Mark S. Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, 91125, USA
| | - Li Zhu
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510
| | - Irfan Ullah
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, 06510
| | - Priti Kumar
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510
| | - Michael S. Kay
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, 91125, USA
| |
Collapse
|
12
|
Wilcox CS, Herbert C, Wang C, Ma Y, Sun P, Li T, Verbesey J, Kumar P, Kassaye S, Welch WJ, Choi MJ, Pourafshar N, Wang D. Signals From Inflamed Perivascular Adipose Tissue Contribute to Small-Vessel Dysfunction in Women With Human Immunodeficiency Virus. J Infect Dis 2024; 230:67-77. [PMID: 39052698 PMCID: PMC11272057 DOI: 10.1093/infdis/jiae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND People with the human immunodeficiency virus (PWH) have microvascular disease. Because perivascular adipose tissue (PVAT) regulates microvascular function and adipose tissue is inflamed in PWH, we tested the hypothesis that PWH have inflamed PVAT that impairs the function of their small vessels. METHODS Subcutaneous small arteries were dissected with or without PVAT from a gluteal skin biopsy from 11 women with treated HIV (WWH) aged < 50 years and 10 matched women without HIV, and studied on isometric myographs. Nitric oxide (NO) and reactive oxygen species (ROS) were measured by fluorescence microscopy. Adipokines and markers of inflammation and ROS were assayed in PVAT. RESULTS PVAT surrounding the small arteries in control women significantly (P < .05) enhanced acetylcholine-induced endothelium-dependent relaxation and NO, and reduced contractions to thromboxane and endothelin-1. However, these effects of PVAT were reduced significantly (P < .05) in WWH whose PVAT released less adiponectin but more markers of ROS and inflammation. Moderation of contractions by PVAT were correlated positively with adipose adiponectin. CONCLUSIONS PVAT from WWH has oxidative stress, inflammation, and reduced release of adiponectin, which may contribute to enhanced contractions and therefore could promote small-artery dysfunction.
Collapse
Affiliation(s)
- Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Carly Herbert
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study Combined Cohort Study, Georgetown University, Washington, District of Columbia, USA
| | - Cheng Wang
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Division of Nephrology, The Fifth Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yuchi Ma
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Philena Sun
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Tian Li
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Jennifer Verbesey
- MedStar Georgetown Transplant Institute, Washington, District of Columbia, USA
| | - Princy Kumar
- Division of Infection Disease, Georgetown University, Washington, District of Columbia, USA
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study, Georgetown University, Washington, District of Columbia, USA
| | - Seble Kassaye
- Division of Infection Disease, Georgetown University, Washington, District of Columbia, USA
- Multicenter Aids Cohort Study and the Women's Interagency HIV Study, Georgetown University, Washington, District of Columbia, USA
| | - William J Welch
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| | - Michael J Choi
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Medstar Georgetown University Hospital, Department of Nephrology and Hypertension, Washington, District of Columbia, USA
| | - Negiin Pourafshar
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
- Medstar Georgetown University Hospital, Department of Nephrology and Hypertension, Washington, District of Columbia, USA
| | - Dan Wang
- Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
13
|
Sun Y, Xu M, Duan Q, Bryant JL, Xu X. The role of autophagy in the progression of HIV infected cardiomyopathy. Front Cell Dev Biol 2024; 12:1372573. [PMID: 39086659 PMCID: PMC11289186 DOI: 10.3389/fcell.2024.1372573] [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: 01/18/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024] Open
Abstract
Although highly active antiretroviral therapy (HAART) has changed infection with human immunodeficiency virus (HIV) from a diagnosis with imminent mortality to a chronic illness, HIV positive patients who do not develop acquired immunodeficiency syndrome (AIDs) still suffer from a high rate of cardiac dysfunction and fibrosis. Regardless of viral load and CD count, HIV-associated cardiomyopathy (HIVAC) still causes a high rate of mortality and morbidity amongst HIV patients. While this is a well characterized clinical phenomena, the molecular mechanism of HIVAC is not well understood. In this review, we consolidate, analyze, and discuss current research on the intersection between autophagy and HIVAC. Multiple studies have linked dysregulation in various regulators and functional components of autophagy to HIV infection regardless of mode of viral entry, i.e., coronary, cardiac chamber, or pericardial space. HIV proteins, including negative regulatory factor (Nef), glycoprotein 120 (gp120), and transactivator (Tat), have been shown to interact with type II microtubule-associated protein-1 β light chain (LC3-II), Rubiquitin, SQSTM1/p62, Rab7, autophagy-specific gene 7 (ATG7), and lysosomal-associated membrane protein 1 (LAMP1), all molecules critical to normal autophagy. HIV infection can also induce dysregulation of mitochondrial bioenergetics by altering production and equilibrium of adenosine triphosphate (ATP), mitochondrial reactive oxygen species (ROS), and calcium. These changes alter mitochondrial mass and morphology, which normally trigger autophagy to clear away dysfunctional organelles. However, with HIV infection also triggering autophagy dysfunction, these abnormal mitochondria accumulate and contribute to myocardial dysfunction. Likewise, use of HAART, azidothymidine and Abacavir, have been shown to induce cardiac dysfunction and fibrosis by inducing abnormal autophagy during antiretroviral therapy. Conversely, studies have shown that increasing autophagy can reduce the accumulation of dysfunctional mitochondria and restore cardiomyocyte function. Interestingly, Rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, has also been shown to reduce HIV-induced cytotoxicity by regulating autophagy-related proteins, making it a non-antiviral agent with the potential to treat HIVAC. In this review, we synthesize these findings to provide a better understanding of the role autophagy plays in HIVAC and discuss the potential pharmacologic targets unveiled by this research.
Collapse
Affiliation(s)
- Yuting Sun
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Mengmeng Xu
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University, New York, NY, United States
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
| | - Joseph L. Bryant
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
14
|
Masenga SK, Liweleya S, Kirabo A. High salt intake and HIV infection on endothelial glycocalyx shedding in salt-sensitive hypertension. Front Cell Dev Biol 2024; 12:1395885. [PMID: 39081863 PMCID: PMC11286502 DOI: 10.3389/fcell.2024.1395885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
The endothelial glycocalyx is closely associated with various physiological and pathophysiological events. Significant modification of the endothelial glycocalyx is an early process in the pathogenesis of cardiovascular disease. High dietary salt and HIV infection damages the endothelial glycocalyx causing endothelial dysfunction and increasing the risk for salt-sensitive hypertension and cardiovascular disease. The two factors, HIV infection and dietary salt are critical independent predictors of hypertension and cardiovascular disease and often synergize to exacerbate and accelerate disease pathogenesis. Salt-sensitive hypertension is more common among people living with HIV and is associated with risk for cardiovascular disease, stroke, heart attack and even death. However, the underlying mechanisms linking endothelial glycocalyx damage to dietary salt and HIV infection are lacking. Yet, both HIV infection/treatment and dietary salt are closely linked to endothelial glycocalyx damage and development of salt-sensitive hypertension. Moreover, the majority of individuals globally, consume more salt than is recommended and the burden of HIV especially in sub-Sahara Africa is disproportionately high. In this review, we have discussed the missing link between high salt and endothelial glycocalyx shedding in the pathogenesis of salt-sensitive hypertension. We have further elaborated the role played by HIV infection and treatment in modifying endothelial glycocalyx integrity to contribute to the development of hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Situmbeko Liweleya
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, United States
- Vanderbilt Institute for Global Health, Nashville, TN, United States
| |
Collapse
|
15
|
Seplowe M, Goyal A, Fowler T, Michaud L, Glassman R, Frishman WH, Aronow WS. Atherosclerosis and HIV: Exploring Cardiovascular Risks and Management. Cardiol Rev 2024:00045415-990000000-00296. [PMID: 38980010 DOI: 10.1097/crd.0000000000000747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The advent of antiretroviral therapy has markedly improved the life expectancy of individuals with HIV, leading to a shift in clinical focus from managing opportunistic infections to addressing chronic conditions, such as atherosclerotic cardiovascular disease (ASCVD). Emerging evidence highlights an elevated risk of ASCVD among people living with HIV, characterized by a higher incidence of acute myocardial infarction, ischemic stroke, and heart failure compared with the general population. This review examines the epidemiology, pathophysiology, and management of ASCVD in the context of HIV. It explores the interplay between HIV infection, antiretroviral therapy, and traditional cardiovascular risk factors, underscoring the need for comprehensive cardiovascular risk reduction strategies tailored to people living with HIV. Through synthesizing data from clinical trials, observational studies, and basic research, the review aims to enhance understanding of HIV-associated ASCVD and inform healthcare practices to improve the longevity and quality of life for this patient population.
Collapse
Affiliation(s)
- Matthew Seplowe
- From the Department of Hospital Medicine, Mount Sinai Morningside, New York, NY
| | - Anjali Goyal
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Tolison Fowler
- Department of General Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Liana Michaud
- Departments of Cardiology and Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | - Rebecca Glassman
- Department of General Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| | | | - Wilbert S Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center, New York Medical College, Valhalla, NY
| |
Collapse
|
16
|
Andric V, Boban J, Maric D, Kozic D, Brkic S, Bulovic A. Additive Effect of Metabolic Syndrome on Brain Atrophy in People Living with HIV-Magnetic Resonance Volumetry Study. Metabolites 2024; 14:331. [PMID: 38921466 PMCID: PMC11205900 DOI: 10.3390/metabo14060331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
With people living with HIV (PLWH) reaching the senium, the importance of aging-related comorbidities such as metabolic syndrome (MS) becomes increasingly important. This study aimed to determine the additive effect of MS on brain atrophy in PLWH. This prospective study included 43 PLWH, average age of 43.02 ± 10.93 years, and 24 healthy controls, average age of 36.87 ± 8.89 years. PLWH were divided into two subgroups: without MS and with MS, according to NCEP ATP III criteria. All patients underwent brain magnetic resonance imaging (MRI) on a 3T clinical scanner with MR volumetry, used for defining volumes of cerebrospinal fluid (CSF) spaces and white and grey matter structures, including basal ganglia. A Student's t-test was used to determine differences in brain volumes between subject subgroups. The binary classification was performed to determine the sensitivity and specificity of volumetry findings and cut-off values. Statistical significance was set at p < 0.05. PLWH presented with significantly lower volumes of gray matter, putamen, thalamus, globus pallidus, and nc. accumbens compared to healthy controls; cut-off values were: for gray matter 738.130 cm3, putamen 8.535 cm3, thalamus 11.895 cm3, globus pallidus 2.252 cm3, and nc. accumbens 0.715 cm3. The volumes of CSF and left lateral ventricles were found to be higher in PLWH with MS compared to those without MS, where, with a specificity of 0.310 and sensitivity of 0.714, it can be assumed that PLWH with a CSF volume exceeding 212.83 cm3 are likely to also have MS. This suggests that PLWH with metabolic syndrome may exhibit increased CSF volume above 212.83 cm3 as a consequence of brain atrophy. There seems to be an important connection between MS and brain volume reduction in PLWH with MS, which may add to the accurate identification of persons at risk of developing HIV-associated cognitive impairment.
Collapse
Affiliation(s)
- Vanja Andric
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Clinic for Infectious Diseases, Clinical Center of Vojvodina, Hajduk Veljkova 1, 21000 Novi Sad, Serbia
| | - Jasmina Boban
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Department for Radiology, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Daniela Maric
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Clinic for Infectious Diseases, Clinical Center of Vojvodina, Hajduk Veljkova 1, 21000 Novi Sad, Serbia
| | - Dusko Kozic
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Department for Radiology, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Snezana Brkic
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Clinic for Infectious Diseases, Clinical Center of Vojvodina, Hajduk Veljkova 1, 21000 Novi Sad, Serbia
| | - Aleksandra Bulovic
- Department for Infectious Diseases, Faculty of Medicine Novi Sad, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (J.B.); (D.M.); (D.K.); (S.B.); (A.B.)
- Clinic for Infectious Diseases, Clinical Center of Vojvodina, Hajduk Veljkova 1, 21000 Novi Sad, Serbia
| |
Collapse
|
17
|
Deshetty UM, Chatterjee N, Buch S, Periyasamy P. HIV-1 Tat-Mediated Human Müller Glial Cell Senescence Involves Endoplasmic Reticulum Stress and Dysregulated Autophagy. Viruses 2024; 16:903. [PMID: 38932195 PMCID: PMC11209317 DOI: 10.3390/v16060903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Antiretroviral treatments have notably extended the lives of individuals with HIV and reduced the occurrence of comorbidities, including ocular manifestations. The involvement of endoplasmic reticulum (ER) stress in HIV-1 pathogenesis raises questions about its correlation with cellular senescence or its role in initiating senescent traits. This study investigated how ER stress and dysregulated autophagy impact cellular senescence triggered by HIV-1 Tat in the MIO-M1 cell line (human Müller glial cells). Cells exposed to HIV-1 Tat exhibited increased vimentin expression combined with markers of ER stress (BiP, p-eIF2α), autophagy (LC3, Beclin-1, p62), and the senescence marker p21 compared to control cells. Western blotting and staining techniques like SA-β-gal were employed to examine these markers. Additionally, treatments with ER stress inhibitor 4-PBA before HIV-1 Tat exposure led to a decreased expression of ER stress, senescence, and autophagy markers. Conversely, pre-treatment with the autophagy inhibitor 3-MA resulted in reduced autophagy and senescence markers but did not alter ER stress markers compared to control cells. The findings suggest a link between ER stress, dysregulated autophagy, and the initiation of a senescence phenotype in MIO-M1 cells induced by HIV-1 Tat exposure.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Nivedita Chatterjee
- Vision Research Foundation, Sankara Netralaya, 18, College Road, Chennai 600006, India;
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| |
Collapse
|
18
|
Prakash P, Swami Vetha BS, Chakraborty R, Wenegieme TY, Masenga SK, Muthian G, Balasubramaniam M, Wanjalla CN, Hinton AO, Kirabo A, Williams CR, Aileru A, Dash C. HIV-Associated Hypertension: Risks, Mechanisms, and Knowledge Gaps. Circ Res 2024; 134:e150-e175. [PMID: 38781298 PMCID: PMC11126208 DOI: 10.1161/circresaha.124.323979] [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] [Indexed: 05/25/2024]
Abstract
HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.
Collapse
Affiliation(s)
- Prem Prakash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Berwin Singh Swami Vetha
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Rajasree Chakraborty
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Tara-Yesomi Wenegieme
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
| | - Gladson Muthian
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Muthukumar Balasubramaniam
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | | | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Azeez Aileru
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Chandravanu Dash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| |
Collapse
|
19
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [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] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
20
|
Jones R, Robinson AT, Beach LB, Lindsey ML, Kirabo A, Hinton A, Erlandson KM, Jenkins ND. Exercise to Prevent Accelerated Vascular Aging in People Living With HIV. Circ Res 2024; 134:1607-1635. [PMID: 38781293 PMCID: PMC11126195 DOI: 10.1161/circresaha.124.323975] [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] [Indexed: 05/25/2024]
Abstract
Given advances in antiretroviral therapy, the mortality rate for HIV infection has dropped considerably over recent decades. However, people living with HIV (PLWH) experience longer life spans coupled with persistent immune activation despite viral suppression and potential toxicity from long-term antiretroviral therapy use. Consequently, PLWH face a cardiovascular disease (CVD) risk more than twice that of the general population, making it the leading cause of death among this group. Here, we briefly review the epidemiology of CVD in PLWH highlighting disparities at the intersections of sex and gender, age, race/ethnicity, and the contributions of social determinants of health and psychosocial stress to increased CVD risk among individuals with marginalized identities. We then overview the pathophysiology of HIV and discuss the primary factors implicated as contributors to CVD risk among PLWH on antiretroviral therapy. Subsequently, we highlight the functional evidence of premature vascular dysfunction as an early pathophysiological determinant of CVD risk among PLWH, discuss several mechanisms underlying premature vascular dysfunction in PLWH, and synthesize current research on the pathophysiological mechanisms underlying accelerated vascular aging in PLWH, focusing on immune activation, chronic inflammation, and oxidative stress. We consider understudied aspects such as HIV-related changes to the gut microbiome and psychosocial stress, which may serve as mechanisms through which exercise can abrogate accelerated vascular aging. Emphasizing the significance of exercise, we review various modalities and their impacts on vascular health, proposing a holistic approach to managing CVD risks in PLWH. The discussion extends to critical future study areas related to vascular aging, CVD, and the efficacy of exercise interventions, with a call for more inclusive research that considers the diversity of the PLWH population.
Collapse
Affiliation(s)
- Raymond Jones
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Lauren B. Beach
- Department of Medical Social Sciences, Northwestern, Chicago, IL
- Department of Preventive Medicine, Northwestern, Chicago, IL
| | - Merry L. Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, TN
- Research Service, Nashville VA Medical Center, Nashville, TN
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Nashville, TN
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN
- Vanderbilt Institute for Global Health, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | | | - Nathaniel D.M. Jenkins
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| |
Collapse
|
21
|
Tan S, Li W, Yang C, Zhan Q, Lu K, Liu J, Jin YM, Bai JS, Wang L, Li J, Li Z, Yu F, Li YY, Duan YX, Lu L, Zhang T, Wei J, Li L, Zheng YT, Jiang S, Liu S. gp120-derived amyloidogenic peptides form amyloid fibrils that increase HIV-1 infectivity. Cell Mol Immunol 2024; 21:479-494. [PMID: 38443447 PMCID: PMC11061181 DOI: 10.1038/s41423-024-01144-y] [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: 07/13/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Apart from mediating viral entry, the function of the free HIV-1 envelope protein (gp120) has yet to be elucidated. Our group previously showed that EP2 derived from one β-strand in gp120 can form amyloid fibrils that increase HIV-1 infectivity. Importantly, gp120 contains ~30 β-strands. We examined whether gp120 might serve as a precursor protein for the proteolytic release of amyloidogenic fragments that form amyloid fibrils, thereby promoting viral infection. Peptide array scanning, enzyme degradation assays, and viral infection experiments in vitro confirmed that many β-stranded peptides derived from gp120 can indeed form amyloid fibrils that increase HIV-1 infectivity. These gp120-derived amyloidogenic peptides, or GAPs, which were confirmed to form amyloid fibrils, were termed gp120-derived enhancers of viral infection (GEVIs). GEVIs specifically capture HIV-1 virions and promote their attachment to target cells, thereby increasing HIV-1 infectivity. Different GAPs can cross-interact to form heterogeneous fibrils that retain the ability to increase HIV-1 infectivity. GEVIs even suppressed the antiviral activity of a panel of antiretroviral agents. Notably, endogenous GAPs and GEVIs were found in the lymphatic fluid, lymph nodes, and cerebrospinal fluid (CSF) of AIDS patients in vivo. Overall, gp120-derived amyloid fibrils might play a crucial role in the process of HIV-1 infectivity and thus represent novel targets for anti-HIV therapeutics.
Collapse
Affiliation(s)
- Suiyi Tan
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wenjuan Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chan Yang
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qingping Zhan
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Kunyu Lu
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jun Liu
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Yong-Mei Jin
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Jin-Song Bai
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Lin Wang
- Department of Pathology, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Jinqing Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhaofeng Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, College of Life Sciences, Hebei Agricultural University, Baoding, 071001, China
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yue-Xun Duan
- Yunnan Provincial Infectious Disease Hospital, Kunming, 650301, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jiaqi Wei
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Lin Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Shuwen Liu
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Johansson E, Nazziwa J, Freyhult E, Hong MG, Lindman J, Neptin M, Karlson S, Rezeli M, Biague AJ, Medstrand P, Månsson F, Norrgren H, Esbjörnsson J, Jansson M. HIV-2 mediated effects on target and bystander cells induce plasma proteome remodeling. iScience 2024; 27:109344. [PMID: 38500818 PMCID: PMC10945182 DOI: 10.1016/j.isci.2024.109344] [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: 10/18/2023] [Revised: 11/23/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Despite low or undetectable plasma viral load, people living with HIV-2 (PLWH2) typically progress toward AIDS. The driving forces behind HIV-2 disease progression and the role of viremia are still not known, but low-level replication in tissues is believed to play a role. To investigate the impact of viremic and aviremic HIV-2 infection on target and bystander cell pathology, we used data-independent acquisition mass spectrometry to determine plasma signatures of tissue and cell type engagement. Proteins derived from target and bystander cells in multiple tissues, such as the gastrointestinal tract and brain, were detected at elevated levels in plasma of PLWH2, compared with HIV negative controls. Moreover, viremic HIV-2 infection appeared to induce enhanced release of proteins from a broader range of tissues compared to aviremic HIV-2 infection. This study expands the knowledge on the link between plasma proteome remodeling and the pathological cell engagement in tissues during HIV-2 infection.
Collapse
Affiliation(s)
- Emil Johansson
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
| | - Jamirah Nazziwa
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
| | - Eva Freyhult
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mun-Gwan Hong
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Jacob Lindman
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Malin Neptin
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
| | - Sara Karlson
- Lund University Virus Centre, Lund, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Melinda Rezeli
- BioMS – Swedish National Infrastructure for Biological Mass Spectrometry, Lund University, Lund, Sweden
| | | | - Patrik Medstrand
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
| | - Fredrik Månsson
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Hans Norrgren
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Joakim Esbjörnsson
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marianne Jansson
- Lund University Virus Centre, Lund, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - for the SWEGUB CORE group
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund, Sweden
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- BioMS – Swedish National Infrastructure for Biological Mass Spectrometry, Lund University, Lund, Sweden
- National Public Health Laboratory, Bissau, Guinea-Bissau
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Mojahedi A. Evaluating the use of antiviral drugs in HIV patients with cardiovascular diseases and how to reduce the incidence of cardiac events in these patients. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2024; 14:70-80. [PMID: 38764550 PMCID: PMC11101959 DOI: 10.62347/obxq4787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/27/2024] [Indexed: 05/21/2024]
Abstract
Globally, the incidence of newly diagnosed human immunodeficiency virus (HIV) infections is concerning. Despite enhancing the quality of life for this patient population, antiretroviral therapy (ART) is linked to an increased risk of cardiovascular disease (CVD). In people living with HIV (PLWH) undergoing ART, recent research has demonstrated that the use of statins and aspirin (ASA) can reduce the incidence or progression of CVD. However, research has demonstrated that interactions may occur when these medications are used concurrently in the treatment regimen of PLWH. Therefore, we conclude this systematic review to evaluate the use of ART in HIV individuals with CVD and also the effect of adding ASA and statins to ART for reducing the cardiac adverse events.
Collapse
Affiliation(s)
- Azad Mojahedi
- Department of Internal Medicine, Stony Brook University Hospital Stony Brook, New York, The United States
| |
Collapse
|
24
|
Fisher SA, Jao JK, Yee LM, Serghides L, Chadwick EG, Jacobson DL. Association of Fatty Acid Signatures with HIV Viremia in Pregnancy. AIDS Res Hum Retroviruses 2024; 40:257-267. [PMID: 37772708 PMCID: PMC11040191 DOI: 10.1089/aid.2023.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFAs) are vital for fetal metabolic programming and immunomodulation. Higher n-6:n-3 ratios, reflecting a proinflammatory eicosanoid profile, are associated with adverse perinatal outcomes. Limited data exist, however, on n-6 and n-3 PUFAs specifically in the context of HIV and pregnancy. Our objective was to assess HIV clinical factors associated with PUFA signatures in pregnant persons with HIV (PWH). In this observational cohort, third trimester plasma PUFA concentrations (six n-6 PUFAs, four n-3 PUFAs) were measured, each as a percent of total fatty acid content, via esterification and gas chromatography in pregnant PWH enrolled from 2009 to 2011 in the Nutrition substudy of the Pediatric HIV/AIDS Cohort Study. PUFA ratios (n-6:n-3) were calculated. Exposures assessed were first/second trimester CD4 count (<200 vs. >200 cells/mm3), HIV RNA viral load (VL) (VL >400 vs. <400 copies/mL), and protease inhibitor (PI) versus non-PI antiretroviral therapy (ART). Linear regression models using generalized estimating equations were fit to assess mean differences and their 95% confidence intervals (CIs) in n-6:n-3 by each exposure, adjusted for potential confounders. Of 264 eligible pregnant PWH, the median age was 27 years, 12% had CD4 counts <200 cells/mm3, and 56% had VL ≥400 copies/mL in the first/second trimesters. PUFA concentrations and ratios were similar by CD4 count and PI exposure. n-3 concentrations were lower in PWH with VL ≥400 versus <400 copies/mL (median 2.8% vs. 3.0%, p < .01, respectively); no differences were observed for n-6 concentrations by VL. In models adjusted for age, education, tobacco use, body mass index, and PI-based ART, n-6:n-3 was higher in those with VL ≥400 copies/mL (mean difference: 1.6; 95% CI: 0.79-2.48, p = .0001). Therefore, PUFA signatures in viremic pregnant PWH reflect a proinflammatory eicosanoid milieu. Future studies should evaluate associations of proinflammatory PUFA signatures with adverse perinatal outcomes in PWH.
Collapse
Affiliation(s)
- Stephanie A. Fisher
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jennifer K. Jao
- Division of Infectious Diseases, Departments of Pediatrics and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Infectious Diseases, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Lynn M. Yee
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lena Serghides
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Immunology and Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - Ellen G. Chadwick
- Division of Infectious Diseases, Departments of Pediatrics and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Infectious Diseases, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Denise L. Jacobson
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Kamvuma K, Hamooya BM, Munsaka S, Masenga SK, Kirabo A. Mechanisms and Cardiorenal Complications of Chronic Anemia in People with HIV. Viruses 2024; 16:542. [PMID: 38675885 PMCID: PMC11053456 DOI: 10.3390/v16040542] [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: 03/12/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic anemia is more prevalent in people living with HIV (PLWH) compared to the general population. The mechanisms that drive chronic anemia in HIV are multifaceted and include functional impairment of hematopoietic stem cells, dysregulation of erythropoietin production, and persistent immune activation. Chronic inflammation from HIV infection adversely affects erythropoiesis, erythrocyte lifespan, and erythropoietin response, leading to a heightened risk of co-infections such as tuberculosis, persistent severe anemia, and increased mortality. Additionally, chronic anemia exacerbates the progression of HIV-associated nephrotoxicity and contributes to cardiovascular risk through immune activation and inflammation. This review highlights the cardinal role of chronic inflammation as a link connecting persistent anemia and cardiovascular complications in PLWH, emphasizing the need for a universal understanding of these interconnected pathways for targeted interventions.
Collapse
Affiliation(s)
- Kingsley Kamvuma
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (K.K.); (B.M.H.)
| | - Benson M. Hamooya
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (K.K.); (B.M.H.)
| | - Sody Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka P.O Box 50110, Zambia;
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (K.K.); (B.M.H.)
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Annet Kirabo
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
26
|
Obare LM, Priest S, Ismael A, Mashayekhi M, Zhang X, Stolze LK, Sheng Q, Vue Z, Neikirk K, Beasley H, Gabriel C, Temu T, Gianella S, Mallal S, Koethe JR, Hinton A, Bailin S, Wanjalla CN. Cytokine and Chemokine Receptor Profiles in Adipose Tissue Vasculature Unravel Endothelial Cell Responses in HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.10.584280. [PMID: 38559150 PMCID: PMC10979923 DOI: 10.1101/2024.03.10.584280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Chronic systemic inflammation contributes to a substantially elevated risk of myocardial infarction in people living with HIV (PLWH). Endothelial cell dysfunction disrupts vascular homeostasis regulation, increasing the risk of vasoconstriction, inflammation, and thrombosis that contribute to cardiovascular disease. Our objective was to study the effects of plasma from PLWH on endothelial cell (EC) function, with the hypothesis that cytokines and chemokines are major drivers of EC activation. We first broadly phenotyped chemokine and cytokine receptor expression on arterial ECs, capillary ECs, venous ECs, and vascular smooth muscle cells (VSMCs) in adipose tissue in the subcutaneous adipose tissue of 59 PLWH using single cell transcriptomic analysis. We used CellChat to predict cell-cell interactions between ECs and other cells in the adipose tissue and Spearman correlation to measure the association between ECs and plasma cytokines. Finally, we cultured human arterial ECs (HAECs) in plasma-conditioned media from PLWH and performed bulk sequencing to study the direct effects ex-vivo. We observed that arterial and capillary ECs expressed higher interferon and tumor necrosis factor (TNF) receptors. Venous ECs had more interleukin (IL)-1R1 and ACKR1 receptors, and VSMCs had high significant IL-6R expression. CellChat predicted ligand-receptor interactions between adipose tissue immune cells as senders and capillary ECs as recipients in TNF-TNFRSF1A/B interactions. Chemokines expressed largely by capillary ECs were predicted to bind ACKR1 receptors on venous ECs. Beyond the adipose tissue, the proportion of venous ECs and VSMCs were positively plasma IL-6. In ex-vivo experiments, HAECs cultured with plasma-conditioned media from PLWH expressed transcripts that enriched for the TNF-α and reactive oxidative phosphorylation pathways. In conclusion, ECs demonstrate heterogeneity in cytokine and chemokine receptor expression. Further research is needed to fully elucidate the role of cytokines and chemokines in EC dysfunction and to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anas Ismael
- Department of Radiology, National Postgraduate Medical College of Nigeria, Lagos, Nigeria
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lindsey K. Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Heather Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Curtis Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Division of Pathology, Harvard Medical College, Boston, MA, USA
| | - Sara Gianella
- Division of Infectious Diseases, University of California, San Diego, CA, USA
| | - Simon Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Samuel Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
27
|
Mokoena H, Mabhida SE, Choshi J, Dludla PV, Nkambule BB, Mchiza ZJ, Ndwandwe DE, Kengne AP, Hanser S. Endothelial dysfunction and cardiovascular diseases in people living with HIV on specific highly active antiretroviral therapy regimen: A systematic review of clinical studies. ATHEROSCLEROSIS PLUS 2024; 55:47-54. [PMID: 38379882 PMCID: PMC10876676 DOI: 10.1016/j.athplu.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024]
Abstract
Despite the improved efficacy of highly active antiretroviral therapy (HAART) in viral suppression, emerging evidence indicates an increased burden of noncommunicable diseases in people living with HIV (PLWH). Immune activation and persistently elevated levels of inflammation have been associated with endothelial dysfunction in PLWH, likely contributing to the development of cardiovascular diseases (CVDs). Here, electronic search databases including PubMed, Google Scholar, Cochrane Library, and Science Direct were used to retrieve scientific evidence reporting on any association between markers of endothelial function and CVD-related outcomes in PLWH on HAART. Extracted data was subjected to quality assessment using the Downs and Black checklist. Most (60 %) of the results indicated the presence of endothelial dysfunction in PLWH on HAART, and this was mainly through reduced flow mediated dilation and elevated serum makers of adhesion molecules like ICAM-1, VCAM-1, and P-selectin. The summarized evidence indicates an association between persistently elevated markers of endothelial dysfunction and a pro-inflammatory state in PLWH on HAART. Only a few studies reported on improved endothelial function markers in PLWH on HAART, while limited evidence is available to prove that endothelial dysfunction is associated with CVD-risk, which could be attributed to therapeutic effects of HAART. Limited studies with relatively high quality of evidence were included in this systematic review. In conclusion, results from this review lay an important foundation for future research, even a meta-analysis, that will improve the understanding of the contributing factors to the burden of CVDs in PLWH on HAART.
Collapse
Affiliation(s)
- Haskly Mokoena
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa
| | - Sihle E. Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Joel Choshi
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa
| | - Phiwayinkosi V. Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3880, South Africa
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Zandile J. Mchiza
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- School of Public Health, University of the Western Cape, Bellville, 7535, South Africa
| | - Duduzile E. Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - André P. Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- Department of Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Sidney Hanser
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, 0727, South Africa
| |
Collapse
|
28
|
Havlickova K, Snopkova S, Pohanka M, Svacinka R, Vydrar D, Husa P, Zavrelova J, Zlamal F, Fabianova L, Penka M, Husa P. Oxidative stress, microparticles, and E-selectin do not depend on HIV suppression. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024. [PMID: 38390755 DOI: 10.5507/bp.2024.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Oxidative stress and inflammation are considered predictors of diseases associated with aging. Markers of oxidative stress, inflammation, and endothelial activation were investigated in people with HIV on antiretroviral treatment to determine whether they had an immunosenescent phenotype that might predispose to the development of premature age-related diseases. PATIENTS AND METHODS This study was conducted on 213 subjects with HIV. The control groups consisted of healthy HIV-negative adults. The level of oxidative stress was measured by assessing the production of malondialdehyde levels, which were detected by thiobarbituric acid reactive substance (TBARS) assay. The level of microparticles indicated the presence of inflammation and endothelial activation was measured by E-selectin levels. Significant differences were determined by appropriate statistical tests, depending on the distribution of variables. Relationships between continuous variables were quantified using Spearman's rank correlation coefficient. RESULTS TBARS, and microparticle and E-selectin levels were significantly higher in untreated and treated subjects with HIV compared with HIV-negative controls (P<0.001). The levels of the investigated markers were not significantly different between untreated and treated patients and no significant correlation of these markers was found with CD4+ count, CD4+/CD8+ ratio, and the number of HIV-1 RNA copies. CONCLUSIONS Elevated markers of oxidative stress, inflammatory and endothelial activation were independent of the virologic and immunologic status of people with HIV. These results support the hypothesis that residual viremia in cellular reservoirs of various tissues is a key factor related to the premature aging of the immune system and predisposition to the premature development of diseases associated with aging.
Collapse
|
29
|
Frias MA, Pagano S, Bararpour N, Sidibé J, Kamau F, Fétaud-Lapierre V, Hudson P, Thomas A, Lecour S, Strijdom H, Vuilleumier N. People living with HIV display increased anti-apolipoprotein A1 auto-antibodies, inflammation, and kynurenine metabolites: a case-control study. Front Cardiovasc Med 2024; 11:1343361. [PMID: 38414919 PMCID: PMC10896987 DOI: 10.3389/fcvm.2024.1343361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/25/2024] [Indexed: 02/29/2024] Open
Abstract
Objective This study aimed to study the relationship between auto-antibodies against apolipoprotein A1 (anti-apoA1 IgG), human immunodeficiency virus (HIV) infection, anti-retroviral therapy (ART), and the tryptophan pathways in HIV-related cardiovascular disease. Design This case-control study conducted in South Africa consisted of control volunteers (n = 50), people living with HIV (PLWH) on ART (n = 50), and untreated PLWH (n = 44). Cardiovascular risk scores were determined, vascular measures were performed, and an extensive biochemical characterisation (routine, metabolomic, and inflammatory systemic profiles) was performed. Methods Anti-apoA1 IgG levels were assessed by an in-house ELISA. Inflammatory biomarkers were measured with the Meso Scale Discovery® platform, and kynurenine pathway metabolites were assessed using targeted metabolomic profiling conducted by liquid chromatography-multiple reaction monitoring/mass spectrometry (LC-MRM/MS). Results Cardiovascular risk scores and vascular measures exhibited similarities across the three groups, while important differences were observed in systemic inflammatory and tryptophan pathways. Anti-apoA1 IgG seropositivity rates were 15%, 40%, and 70% in control volunteers, PLWH ART-treated, and PLWH ART-naïve, respectively. Circulating anti-apoA1 IgG levels were significantly negatively associated with CD4+ cell counts and positively associated with viremia and pro-inflammatory biomarkers (IFNγ, TNFα, MIPα, ICAM-1, VCAM-1). While circulating anti-apoA1 IgG levels were associated with increased levels of kynurenine in both control volunteers and PLWH, the kynurenine/tryptophan ratio was significantly increased in PLWH ART-treated. Conclusion HIV infection increases the humoral response against apoA1, which is associated with established HIV severity criteria and kynurenine pathway activation.
Collapse
Affiliation(s)
- Miguel A. Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nasim Bararpour
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Genetics, Stanford University, Stanford, CA, United States
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, United States
| | - Jonathan Sidibé
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Festus Kamau
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vanessa Fétaud-Lapierre
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Hudson
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Aurélien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Sandrine Lecour
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hans Strijdom
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
30
|
Strauss KLE, Phoswa WN, Lebelo SL, Modjadji P, Mokgalaboni K. Endothelial dysfunction, a predictor of cardiovascular disease in HIV patients on antiretroviral therapy: A systematic review and meta-analysis. Thromb Res 2024; 234:101-112. [PMID: 38211378 DOI: 10.1016/j.thromres.2023.12.011] [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: 09/27/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Aim Although antiretroviral therapy (ART) is available, the rate of new HIV infections is alarming. With this trend, it is anticipated that the use of ART will continue to rise, potentially resulting in associated vascular disorders. Therefore, we aimed to examine the impact of ART on endothelial function in people living with HIV (PLHIV), a predictor of cardiovascular diseases. METHOD A comprehensive search for evidence was made on PubMed and Scopus on May 06, 2023, following the Preferred Reporting Items for Systematic Review and Meta-analysis. Cochrane and Newcastle-Ottawa quality assessment scales were used to evaluate quality, while the metaHun web tool and Review Manager version 5.4.1 were used for analysis. Subgroup, sensitivity, and publication bias were conducted for each outcome measure. RESULTS We identified 37 studies, including a sample size of 3700 with 2265 individuals on ART. The analyzed evidence showed a large significant effect of ART on vascular cell adhesion molecule-1, with a standardized mean difference (SMD) of -1.23 (95 % CI: -1.72, -0.74; p = 0.0013). Similarly, a significant medium effect of ART was observed on intercellular cell adhesion molecule-1 in PLHIV, with an SMD of -1.28 (95 % CI: -2.00, -0.56; p = 0.0231) compared to the control group. Furthermore, ART exhibited a significant but small effect on flow-mediated dilation (FMD) with an SMD of -0.40 (95 % CI: -0.62, -0.19, p = 0.0159). CONCLUSION Our findings show an improved endothelial function in PLHIV on ART, as demonstrated by reduced adhesion molecules; however, ART exhibited a small effect on FMD, thus suggesting PLHIV on ART may still be at risk of endothelial dysfunction and further cardiovascular diseases.
Collapse
Affiliation(s)
- Kay-Lee E Strauss
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa.
| | - Wendy N Phoswa
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa.
| | - Sogolo L Lebelo
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa.
| | - Perpetua Modjadji
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa; Non-Communicable Disease Research Unit, South African Medical Research Council, Cape Town 7505, South Africa; Department of Public Health, School of Health Care Sciences, Sefako Makgatho Health Sciences University, 1 Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa.
| | - Kabelo Mokgalaboni
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa.
| |
Collapse
|
31
|
Whiteson HZ, Drogy M, Eickel G, Frishman WH. Pitavastatin in the Prevention of Cardiovascular Disease in People Living with HIV: A Review. Cardiol Rev 2024:00045415-990000000-00191. [PMID: 38294226 DOI: 10.1097/crd.0000000000000646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
HIV is associated with a wide array of pathophysiologic mechanisms that ultimately contribute to mortality. While HIV is traditionally known as a disease that attacks the immune system, it is now established that infection with HIV can cause cardiovascular disease (CVD). Through inflammation, atherogenesis, interactions with antiretroviral therapy/highly-active antiretroviral therapy (ART/HAART), and other mechanisms, HIV is an independent risk factor for the development of CVD. The treatment of the CVD risks associated with HIV is complicated, especially due to interactions with hyperlipidemic drugs and ART/HAART. There is a prompt need for a drug (or drug class) that is known to reduce the risk of CVD, specifically in people living with HIV. Recently, the randomized trial to prevent vascular events in HIV trial evaluated the usage of pitavastatin in preventing major cardiac events in people with HIV, showing a significant reduction in cardiac events among those taking the therapeutic. In this review, we evaluate the mechanisms by which HIV contributes to CVD, and the randomized trial to prevent vascular events in HIV trial, and postulate about future directions of the drug in treating people living with HIV.
Collapse
Affiliation(s)
- Harris Z Whiteson
- From the Department of Medicine, New York Medical College, School of Medicine
| | - Maddison Drogy
- From the Department of Medicine, New York Medical College, School of Medicine
| | - Grant Eickel
- NYU Langone Transplant Institute, NYU Grossman School of Medicine, NY
| | - William H Frishman
- From the Department of Medicine, New York Medical College, School of Medicine
| |
Collapse
|
32
|
Basta D, Latinovic OS, Tagaya Y, Silvestri G. Potential Advantages of a Well-balanced Nutrition Regimen for People Living with Human Immunodeficiency Virus Type -1. JOURNAL OF AIDS AND HIV TREATMENT 2024; 6:11-27. [PMID: 38845818 PMCID: PMC11155617 DOI: 10.33696/aids.6.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
This review underscores the important role of nutrition in enhancing the management of Human Immunodeficiency Virus type 1 (HIV-1). Highlighting the efficacy of dietary interventions, including, the importance of omega-3 fatty acids, vitamins D and B-12, and the Mediterranean diet, we delineate how these beneficial nutritional strategies can improve the effectiveness of combined antiretroviral therapy (cART), mitigate its side effects, and ameliorate metabolic disorders in people living with HIV-1 (PLWH). Our review advocates for the integration and implementation of personalized nutritional assessments into the care plan for PLWH, proposing actionable strategies for healthcare providers in HIV-1 field. Summarizing the current standing of the relevance of the nutritional and well-planned diet recommended for the PLWH and emphasizing on the future research directions, this review establishes a foundation for nutrition as a cornerstone in comprehensive HIV-1 management. Our review aims to improve patients' health outcomes and overall quality of life for PLWH.
Collapse
Affiliation(s)
- Daniele Basta
- Green Home scarl, Scientific Committee, Arcavacata di Rende (CS), Italy
| | - Olga S. Latinovic
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Yutaka Tagaya
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, MD, 21201, USA
| | - Giovannino Silvestri
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, USA
| |
Collapse
|
33
|
Awamura T, Nakasone ES, Gangcuangco LM, Subia NT, Bali AJ, Chow DC, Shikuma CM, Park J. Platelet and HIV Interactions and Their Contribution to Non-AIDS Comorbidities. Biomolecules 2023; 13:1608. [PMID: 38002289 PMCID: PMC10669125 DOI: 10.3390/biom13111608] [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: 10/05/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Platelets are anucleate cytoplasmic cell fragments that circulate in the blood, where they are involved in regulating hemostasis. Beyond their normal physiologic role, platelets have emerged as versatile effectors of immune response. During an infection, cell surface receptors enable platelets to recognize viruses, resulting in their activation. Activated platelets release biologically active molecules that further trigger host immune responses to protect the body against infection. Their impact on the immune response is also associated with the recruitment of circulating leukocytes to the site of infection. They can also aggregate with leukocytes, including lymphocytes, monocytes, and neutrophils, to immobilize pathogens and prevent viral dissemination. Despite their host protective role, platelets have also been shown to be associated with various pathophysiological processes. In this review, we will summarize platelet and HIV interactions during infection. We will also highlight and discuss platelet and platelet-derived mediators, how they interact with immune cells, and the multifaceted responsibilities of platelets in HIV infection. Furthermore, we will give an overview of non-AIDS comorbidities linked to platelet dysfunction and the impact of antiretroviral therapy on platelet function.
Collapse
Affiliation(s)
- Thomas Awamura
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Elizabeth S. Nakasone
- University of Hawai‘i Cancer Center, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
| | - Louie Mar Gangcuangco
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Natalie T. Subia
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Aeron-Justin Bali
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Dominic C. Chow
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Cecilia M. Shikuma
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| |
Collapse
|
34
|
Ziogos E, Kwapong YA, Weiss RG, Schär M, Brown TT, Bagchi S, Soleimanifard A, Harb T, Piggott DA, Gerstenblith G, Leucker TM, Hays AG. Coronary artery endothelial function and aging in people with HIV and HIV-negative individuals. Am J Physiol Heart Circ Physiol 2023; 325:H1099-H1107. [PMID: 37682238 PMCID: PMC10907030 DOI: 10.1152/ajpheart.00143.2023] [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: 03/13/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Coronary artery disease (CAD) is a common comorbidity in people with human immunodeficiency virus (HIV) (PWH) and impaired coronary endothelial function (CEF) plays a central role in the pathogenesis of CAD. Age-related impaired CEF among PWH, however, is not well characterized. We investigated the association between CEF and age in males and females with and without HIV using 3-T magnetic resonance imaging (MRI). We measured the changes in coronary cross-sectional area (CSA) and coronary blood flow during isometric handgrip exercise (IHE), an established endothelial-dependent stressor with smaller increases in CSA and coronary blood flow indicative of impaired CEF. We included 106 PWH and 82 individuals without HIV. Differences in demographic and clinical characteristics between PWH and individuals without HIV were explored using Pearson's χ2 test for categorical variables and Welch's t test for continuous variables. Linear regression models were used to examine the association between CEF and age. CEF was significantly lower in PWH as compared with individuals without HIV. Coronary endothelial dysfunction was also present at younger ages in PWH than in the individuals without HIV and there were significant differences in CEF between the PWH and individuals without HIV across age groups. Among the individuals without HIV, the percent changes in CSA were inversely related to age in unadjusted and adjusted models. There was no significant association between CEF and age in PWH. To the best of our knowledge, this is the first study to examine the relationship between age and CEF in PWH, and our results suggest that factors other than age significantly impair CEF in PWH across the life span.NEW & NOTEWORTHY This is the first study to examine the relationship between age and coronary endothelial function (CEF) in people with human immunodeficiency virus (HIV) (PWH). CEF was assessed using magnetic resonance imaging (MRI) in people with and without HIV. Although age and CEF were significantly inversely related in individuals without HIV, there was no association between age and CEF in PWH.
Collapse
Affiliation(s)
- Efthymios Ziogos
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Yaa A Kwapong
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Robert G Weiss
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Michael Schär
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Todd T Brown
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shashwatee Bagchi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Alborz Soleimanifard
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Tarek Harb
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Damani A Piggott
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Gary Gerstenblith
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Thorsten M Leucker
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Allison G Hays
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
35
|
Avagimyan A, Pogosova N, Kakturskiy L, Sheibani M, Urazova O, Trofimenko A, Navarsdyan G, Jndoyan Z, Abgaryan K, Fogacci F, Galli M, Agati L, Kobalava Z, Shafie D, Marzilli M, Gogiashvili L, Sarrafzadegan N. HIV-Related Atherosclerosis: State-of-the-Art-Review. Curr Probl Cardiol 2023; 48:101783. [PMID: 37172874 DOI: 10.1016/j.cpcardiol.2023.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
The infection caused by the Human Immunodeficiency Virus (HIV) has spread rapidly across the globe, assuming the characteristics of an epidemic in some regions. Thanks to the introduction of antiretroviral therapy into routine clinical practice, there was a considerable breakthrough in the treatment of HIV, that is now HIV is potentially well-controlled even in low-income countries. To date, HIV infection has moved from the group of life-threatening conditions to the group of chronic and well controlled ones and the quality of life and life expectancy of HIV+ people, with an undetectable viral load is closer to that of an HIV- people. However, unsolved issues still persist. For example: people living with HIV are more prone to the age-related diseases, especially atherosclerosis. For this reason, a better understanding of the mechanisms of HIV-associated destabilization of vascular homeostasis seems to be an urgent duty, that may lead to the development of new protocols, bringing the possibilities of pathogenetic therapies to a new level. The purpose of the article was to evaluate the pathological aspects of HIV-induced atherosclerosis.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Assistant Professor, Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia.
| | - Nana Pogosova
- Professor, Deputy of General Director for Science and Preventive Cardiology, National Medical Research Centre of Cardiology after E. Chazov, Moscow, Russia
| | - Lev Kakturskiy
- Professor, Scientific Director, Research Institute of Human Morphology FSBI «Petrovskiy NRCS, Moscow, Russia
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Olga Urazova
- Professor, Head of Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Artem Trofimenko
- Associate Professor, Pathophysiology Department, Kuban State Medical University, Krasnodar, Russia
| | - Grizelda Navarsdyan
- Professor, Pathophysiology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Zinaida Jndoyan
- Professor, Head of Internal Diseases Propedeutics Department, Yerevan State Medical University after M. Heratsi, Armenia
| | - Kristina Abgaryan
- Associate Professor, Medical Microbiology Department, Yerevan State Medical University after M.Heratsi, Armenia
| | - Federica Fogacci
- Research Fellow, Atherosclerosis and Metabolic Disorders Research Unit, University of Bologna, Bologna, Italy
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Luciano Agati
- Professor of Cardiology Department, Head of Cardiology Unit Azienda Policlinico Umberto II, Sapienza University, Rome, Italy
| | - Zhanna Kobalava
- Professor, Head of Internal Disease, Cardiology and Clinical Pharmacology Department, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Davood Shafie
- Isfahan Cardiovascular Research Institute, Isfahan, Iran
| | - Mario Marzilli
- Professor, Head of Cardiovascular Medicine Division, University of Pisa, Pisa, Italy
| | - Liana Gogiashvili
- Professor, Head of Experimental and Clinical Pathology Department, Al. Natishvili Institute of Experimental Morphology, I. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Nizal Sarrafzadegan
- Professor, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Singh S, Moodley J, Naicker T. Differential expression of the angiotensin receptors (AT1, AT2, and AT4) in the placental bed of HIV-infected preeclamptic women of African ancestry. Hypertens Res 2023; 46:1970-1982. [PMID: 37308552 PMCID: PMC10404513 DOI: 10.1038/s41440-023-01314-x] [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: 11/25/2022] [Revised: 04/05/2023] [Accepted: 04/28/2023] [Indexed: 06/14/2023]
Abstract
The Renin-Angiotensin-Aldosterone System (RAAS) is implicated in the pathophysiology of preeclampsia (PE). There is a paucity of data on uteroplacental angiotensin receptors AT1-2 and 4. We evaluated the immunoexpression of AT1R, AT2R, and AT4R within the placental bed of PE vs. normotensive (N) pregnancies stratified by HIV status. Placental bed (PB) biopsies (n = 180) were obtained from N and PE women. Both groups were stratified by HIV status and gestational age into early-and late onset-PE. Immuno-labeling of AT1R, AT2R, and AT4R was quantified using morphometric image analysis. Immunostaining of PB endothelial cells (EC) and smooth muscle cells of spiral arteries (VSMC) displayed an upregulation of AT1R expression compared to the N group (p < 0.0001). Downregulation of AT2R and AT4R expression was observed in PE vs. N group (p = 0.0042 and p < 0.0001), respectively. AT2R immunoexpression declined between HIV+ve and HIV-ve groups, while AT1R and AT4R displayed an increase. An increase in AT1R expression was noted in the EOPE-ve/+ve and LOPE-ve/+ve compared to N-ve/N+ve. In contrast, AT2R and AT4R expression decreased in EOPE-ve/+ve and LOPE-ve/+ve compared to N-ve/N+ve. We demonstrate a significant downregulation of AT2R and AT4R with a concomitant elevated AT1R immunoexpression within PB of HIV-infected PE women. In addition, a decline in AT2R and AT4R with an increase in AT1R immunoexpression in PE, EOPE, and LOPE vs. normotensive pregnancies, irrespective of HIV status. Thus highlighting differential immunoexpression of uteroplacental RAAS receptors based on pregnancy type, HIV status, and gestational age.
Collapse
Affiliation(s)
- Shoohana Singh
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
37
|
Fragkou PC, Ikonomidis I, Benas D, Kavatha D, Moschopoulos CD, Protopapas K, Kostelli G, Thymis J, Mpirmpa D, Galani I, Tsakona M, Oikonomopoulou C, Theocharous G, Gorgoulis VG, Gallos P, Tsiodras S, Antoniadou A, Papadopoulos A, Triantafyllidi H. Endothelial Glycocalyx Integrity in Treatment-Naïve People Living with HIV before and One Year after Antiretroviral Treatment Initiation. Viruses 2023; 15:1505. [PMID: 37515191 PMCID: PMC10383742 DOI: 10.3390/v15071505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Endothelial glycocalyx (EG) derangement has been associated with cardiovascular disease (CVD). Studies on EG integrity among people living with HIV (PLWH), are lacking. We conducted a prospective cohort study among treatment-naïve PLWH who received emtricitabine/tenofovir alafenamide, combined with either an integrase strand transfer inhibitor (INSTI, dolutegravir, raltegravir or elvitegravir/cobicistat), or a protease inhibitor (PI, darunavir/cobicistat). We assessed EG at baseline, 24 (±4) and 48 (±4) weeks, by measuring the perfused boundary region (PBR, inversely proportional to EG thickness), in sublingual microvessels. In total, 66 consecutive PLWH (60 (90.9%) males) with a median age (interquartile range, IQR) of 37 (12) years, were enrolled. In total, 40(60.6%) received INSTI-based regimens. The mean (standard deviation) PBR decreased significantly from 2.17 (0.29) μm at baseline to 2.04 (0.26) μm (p = 0.019), and then to 1.93 (0.3) μm (p < 0.0001) at 24 (±4) and 48 (±4) weeks, respectively. PBR did not differ among treatment groups. PLWH on INSTIs had a significant PBR reduction at 48 (±4) weeks. Smokers and PLWH with low levels of viremia experienced the greatest PBR reduction. This study is the first to report the benefit of antiretroviral treatment on EG improvement in treatment-naïve PLWH and depicts a potential bedside biomarker and therapeutic target for CVD in PLWH.
Collapse
Affiliation(s)
- Paraskevi C Fragkou
- First Department of Critical Care and Pulmonary Services, Evangelismos Hospital, Athens Medical School, National and Kapodistrian University of Athens, 10676 Athens, Greece
| | - Ignatios Ikonomidis
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dimitrios Benas
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dimitra Kavatha
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Charalampos D Moschopoulos
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Protopapas
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Gavriella Kostelli
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - John Thymis
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dionysia Mpirmpa
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Irene Galani
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Maria Tsakona
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Chrysanthi Oikonomopoulou
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - George Theocharous
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Athens Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Athens Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Parisis Gallos
- Computational Biomedicine Laboratory, Department of Digital Systems, University of Piraeus, 18536 Piraeus, Greece
| | - Sotirios Tsiodras
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Anastasia Antoniadou
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Antonios Papadopoulos
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Helen Triantafyllidi
- Second Department of Cardiology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
38
|
Semulimi AW, Batte C, Bayiyana A, Nakabuye M, Mukisa J, Castelnuovo B, Ratanshi RP, Kirenga BJ, Ssinabulya I. Serum E-selectin and endothelial cell-specific Molecule-1 levels among people living with HIV on long term ART in Uganda: a pilot cross-sectional study. AIDS Res Ther 2023; 20:26. [PMID: 37161496 PMCID: PMC10169468 DOI: 10.1186/s12981-023-00519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Prolonged exposure to HIV and anti-retroviral therapy (ART) has been linked with endothelial cell activation which subsequently predisposes people living with HIV (PLWH) to cardiovascular diseases. Serum biomarkers of endothelial cell activation such as E-Selectin and endothelial cell-specific molecule-1 (ESM-1) could aid in early detection of PLWH at a risk of cardiovascular diseases. However, there is a paucity of data on these biomarkers like E-selectin and endothelial cell-specific molecule-1 (ESM-1) among PLWH on long term ART (≥ 10 years) in Uganda. The aim of this study is to determine the serum levels of these biomarkers in this population. METHODS This was a cross-sectional study where we randomly sampled 73 stored serum samples of PLWH who were enrolled in the Infectious Diseases Institute (IDI) ART long term (ALT cohort). We measured serum levels of E-selectin and ESM-1 by ELISA. Data was summarized using median and interquartile range. Inferential statistics were performed to determine predictors of elevated levels of E-selectin. RESULTS Of the 73 samples analyzed, 38 (52.1%) were from female participants. The mean age was 54 ± 9.0 years. Twenty participants (27.4%) had a history of smoking while 52 (71.2%) had a history of alcohol intake. Twenty-five (34.3%) of the participants were overweight whereas 4 (5.6%) were obese. Fifty-four (74%) had an undetectable viral load (≤ 0 copies/ml) and the mean duration of ART at the time of sampling (2014/2015) was 10.4 ± 0.4 years. While serum levels of ESM-1 were not detectable in any of our samples, the median E-selectin levels was 147.6 μm/L ranging from 8.44 μm/L and 1,979.36 μm/L. Sixty-seven participants (91.8%) had elevated levels of E-selectin (> 39 μm/L). CD4 count > 500 cells/µl compared to lower counts was a predictor of elevated levels of E-Selectin (adjusted Odd Ratio 12.5, 95% CI (1.03 - 149.95, p < 0.05). CONCLUSIONS The majority (91.8%) of PLWH on long term ART had elevated levels of E-selectin. Having high CD4 count (> 500 cells/µl) was predictive of elevated levels of E-Selectin. Future work should longitudinally assess the trend of levels of E-selectin and ESM-1 while assessing for cardiovascular diseases endpoint.
Collapse
Affiliation(s)
- Andrew Weil Semulimi
- Lung Institute, Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda.
| | - Charles Batte
- Lung Institute, Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Alice Bayiyana
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Mariam Nakabuye
- Lung Institute, Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - John Mukisa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Rosalind Parkes Ratanshi
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Bruce J Kirenga
- Lung Institute, Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Isaac Ssinabulya
- Department of Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW It is now recognized that SARS-CoV-2 infection can have a long-term impact on health. This review summarizes the current state of knowledge regarding Long COVID in people living with HIV (PLWH). RECENT FINDINGS PLWH may be at elevated risk of experiencing Long COVID. Although the mechanisms contributing to Long COVID are incompletely understood, there are several demographic and clinical factors that might make PLWH vulnerable to developing Long COVID. SUMMARY PLWH should be aware that new or worsening symptoms following SARS-CoV-2 infection might represent Long COVID. HIV providers should be aware of this clinical entity and be mindful that their patients recovering from SARS-CoV-2 infection may be at higher risk.
Collapse
Affiliation(s)
- Michael J. Peluso
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA 94110
| | - Annukka A. R. Antar
- Division of Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
40
|
Da Fonseca Ferreira A, Wei J, Zhang L, Macon CJ, Degnan B, Jayaweera D, Hare JM, Kolber MA, Bellio M, Khan A, Pan Y, Dykxhoorn DM, Wang L, Dong C. HIV Promotes Atherosclerosis via Circulating Extracellular Vesicle MicroRNAs. Int J Mol Sci 2023; 24:7567. [PMID: 37108729 PMCID: PMC10146407 DOI: 10.3390/ijms24087567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
People living with HIV (PLHIV) are at a higher risk of having cerebrocardiovascular diseases (CVD) compared to HIV negative (HIVneg) individuals. The mechanisms underlying this elevated risk remains elusive. We hypothesize that HIV infection results in modified microRNA (miR) content in plasma extracellular vesicles (EVs), which modulates the functionality of vascular repairing cells, i.e., endothelial colony-forming cells (ECFCs) in humans or lineage negative bone marrow cells (lin- BMCs) in mice, and vascular wall cells. PLHIV (N = 74) have increased atherosclerosis and fewer ECFCs than HIVneg individuals (N = 23). Plasma from PLHIV was fractionated into EVs (HIVposEVs) and plasma depleted of EVs (HIV PLdepEVs). HIVposEVs, but not HIV PLdepEVs or HIVnegEVs (EVs from HIVneg individuals), increased atherosclerosis in apoE-/- mice, which was accompanied by elevated senescence and impaired functionality of arterial cells and lin- BMCs. Small RNA-seq identified EV-miRs overrepresented in HIVposEVs, including let-7b-5p. MSC (mesenchymal stromal cell)-derived tailored EVs (TEVs) loaded with the antagomir for let-7b-5p (miRZip-let-7b) counteracted, while TEVs loaded with let-7b-5p recapitulated the effects of HIVposEVs in vivo. Lin- BMCs overexpressing Hmga2 (a let-7b-5p target gene) lacking the 3'UTR and as such is resistant to miR-mediated regulation showed protection against HIVposEVs-induced changes in lin- BMCs in vitro. Our data provide a mechanism to explain, at least in part, the increased CVD risk seen in PLHIV.
Collapse
Affiliation(s)
- Andrea Da Fonseca Ferreira
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jianqin Wei
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukun Zhang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Conrad J. Macon
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bernard Degnan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dushyantha Jayaweera
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael A. Kolber
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael Bellio
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yue Pan
- Biostatistics Division, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Derek M. Dykxhoorn
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Liyong Wang
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chunming Dong
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Section of Cardiology, Department of Medicine, Miami VA Health System, University of Miami, Miami, FL 33146, USA
| |
Collapse
|
41
|
Henning RJ, Greene JN. The epidemiology, mechanisms, diagnosis and treatment of cardiovascular disease in adult patients with HIV. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2023; 13:101-121. [PMID: 37213313 PMCID: PMC10193251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/22/2023] [Indexed: 05/23/2023]
Abstract
More than 1.2 million people in the United States have Human Immunodeficiency Virus (HIV) infections but 13% of these people are unaware of their HIV infection. Current combination antiretroviral therapy (ART) does not cure HIV infection but rather suppresses the infection with the virus persisting indefinitely in latent reservoirs in the body. As a consequence of ART, HIV infection has changed from a fatal disease in the past to a chronic disease today. Currently in the United States, more than 45% of HIV+ individuals are greater than 50 years of age and 25% will be greater than 65 years of age by 2030. Atherosclerotic cardiovascular disease (CVD), including myocardial infarction, stroke, and cardiomyopathy, is now the major cause of death in HIV+ individuals. Novel risk factors, including chronic immune activation and inflammation in the body, antiretroviral therapy, and traditional CVD risk factors, such as tobacco and illicit drug use, hyperlipidemia, the metabolic syndrome, diabetes mellitus, hypertension, and chronic renal disease, contribute to cardiovascular atherosclerosis. This article discusses the complex interactions involving HIV infection, the novel and traditional risk factors for CVD, and the antiretroviral HIV therapies which can contribute to CVD in HIV-infected people. In addition, the treatment of HIV+ patients with acute myocardial infarction, stroke, and cardiomyopathy/heart failure are discussed. Current recommended ART and their major side effects are summarized in table format. All medical personnel must be aware of the increasing incidence of CVD on the morbidity and mortality in HIV infected patients and must be watchful for the presence of CVD in their patients with HIV.
Collapse
Affiliation(s)
- Robert J Henning
- University of South Florida and The Moffitt Cancer Center Tampa, Florida, USA
| | - John N Greene
- University of South Florida and The Moffitt Cancer Center Tampa, Florida, USA
| |
Collapse
|
42
|
Karpova NS, Dmitrenko OP, Budykina TS. Literature Review: The sFlt1/PlGF Ratio and Pregestational Maternal Comorbidities: New Risk Factors to Predict Pre-Eclampsia. Int J Mol Sci 2023; 24:ijms24076744. [PMID: 37047717 PMCID: PMC10095124 DOI: 10.3390/ijms24076744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
One of the main causes of maternal and neonatal morbidity and mortality is pre-eclampsia. It is characterized by a high sFlt1/PlGF ratio, according to prior research. Pregestational diseases in mothers may increase the risk of developing pre-eclampsia. Only a few studies have looked at the connection between maternal comorbidities before conception and the sFlt1/PlGF ratio. The most recent information regarding the association between maternal pregestational diseases and the ratio of sFlt1/PlGF is described in this review. The paper also examines current research suggesting that changes in pregnancy hormones and metabolites are related to a high sFlt1/PlGF ratio. Certain maternal disorders have been found to dramatically raise sFlt-1 and sFlt1/PlGF levels, according to an analysis of the literature. There is still debate about the data on the association between the sFlt1/PlGF ratio and maternal disorders such as HIV, acute coronary syndromes, cardiovascular function in the mother between 19 and 23 weeks of pregnancy, thyroid hormones, diabetes, and cancer. Additional research is needed to confirm these findings.
Collapse
Affiliation(s)
- Nataliia Sergeevna Karpova
- Federal State Budgetary Institution “Research Institute of Pathology and Pathophysiology”, St. Baltiyskaya, House 8, Moscow 125315, Russia
| | - Olga Pavlovna Dmitrenko
- Federal State Budgetary Institution “Research Institute of Pathology and Pathophysiology”, St. Baltiyskaya, House 8, Moscow 125315, Russia
| | - Tatyana Sergeevna Budykina
- State Budgetary Health Institution of the Moscow Region “Moscow Regional Research Institute of Obstetrics and Gynecology”, St. Pokrovka, d.22a, Moscow 101000, Russia
| |
Collapse
|
43
|
Mouchati C, Durieux JC, Zisis SN, McComsey GA. HIV and race are independently associated with endothelial dysfunction. AIDS 2023; 37:271-277. [PMID: 36541639 PMCID: PMC9794140 DOI: 10.1097/qad.0000000000003421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Evaluating the vascular function in HIV-infected compared with HIV uninfected with assessment of body composition, inflammation, and gut integrity markers. DESIGN A noninvasive test that measures the endothelial function. METHODS We included participants at least 18 years old, with peripheral arterial tonometry testing (EndoPAT2000) between 2014 and 2022. Persons with HIV (PWH) had documented infection, a stable ART regimen, and a viral load less than 400 copies/ml. We measured the vessel's function with the reactive hyperemia index (RHI) (normal >1.67) and Augmentation Index. Lower Augmentation Index reflect better arterial elasticity. We assessed markers of systemic inflammation, immune activation, and gut integrity. We used linear mixed models to estimate endothelial dysfunction with a significant P value less than 0.05. RESULTS Overall, 511 participants (296 HIV-infected; 215 HIV-uninfected controls) were included. Estimated RHI among PWH was 13% lower (P = 0.01) compared with persons without HIV. In nonwhite race, the estimated RHI was 9% lower (P = 0.001) than white race. For every 1% increase in BMI, we would expect RHI to increase 0.17% (P = 0.01). At the time of EndoPAT, the estimated RHI was 8% lower (P = 0.04) among protease inhibitor users compared with PWH who were not taking protease inhibitors. The estimated odds of abnormal RHI ≤1.67) is 1.56 times greater [95% confidence interval (CI) 1.05-2.31] in nonwhite race compared with white race, independent of HIV status [OR = 1.4 (95% CI 0.94-2.13)]. There was not enough evidence to suggest that inflammation, gut, or monocyte markers, current or nadir CD4+ cell count, or duration of HIV were associated with endothelial dysfunction. CONCLUSION HIV, nonwhite race, and protease inhibitor use are independently associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Christian Mouchati
- School of Medicine, Case Western Reserve University
- Center for Clinical Research, University Hospitals Cleveland Medical Center
| | - Jared C. Durieux
- Center for Clinical Research, University Hospitals Cleveland Medical Center
| | - Sokratis N. Zisis
- School of Medicine, Case Western Reserve University
- Center for Clinical Research, University Hospitals Cleveland Medical Center
| | - Grace A. McComsey
- School of Medicine, Case Western Reserve University
- Center for Clinical Research, University Hospitals Cleveland Medical Center
- Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| |
Collapse
|
44
|
Luo P, Li L, Huang J, Mao D, Lou S, Ruan J, Chen J, Tang R, Shi Y, Zhou S, Yang H. The role of SUMOylation in the neurovascular dysfunction after acquired brain injury. Front Pharmacol 2023; 14:1125662. [PMID: 37033632 PMCID: PMC10073463 DOI: 10.3389/fphar.2023.1125662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Acquired brain injury (ABI) is the most common disease of the nervous system, involving complex pathological processes, which often leads to a series of nervous system disorders. The structural destruction and dysfunction of the Neurovascular Unit (NVU) are prominent features of ABI. Therefore, understanding the molecular mechanism underlying NVU destruction and its reconstruction is the key to the treatment of ABI. SUMOylation is a protein post-translational modification (PTM), which can degrade and stabilize the substrate dynamically, thus playing an important role in regulating protein expression and biological signal transduction. Understanding the regulatory mechanism of SUMOylation can clarify the molecular mechanism of the occurrence and development of neurovascular dysfunction after ABI and is expected to provide a theoretical basis for the development of potential treatment strategies. This article reviews the role of SUMOylation in vascular events related to ABI, including NVU dysfunction and vascular remodeling, and puts forward therapeutic prospects.
Collapse
Affiliation(s)
- Pengren Luo
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Li
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiashang Huang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Deqiang Mao
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Silong Lou
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jian Ruan
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jie Chen
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Ronghua Tang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - You Shi
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Shuai Zhou
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| | - Haifeng Yang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| |
Collapse
|
45
|
Porcelain Aorta in a Young Person Living with HIV Who Presented with Angina. Diagnostics (Basel) 2022; 12:diagnostics12123147. [PMID: 36553155 PMCID: PMC9776980 DOI: 10.3390/diagnostics12123147] [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: 10/28/2022] [Revised: 11/29/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
People living with human immunodeficiency virus have an increased cardiovascular risk due to higher prevalence of traditional risk factors, such as smoking, dyslipidemia, hypertension, diabetes, or obesity, and particular risk factors, such as inflammation, endothelial dysfunction, and antiretroviral therapy. Thus, people living with human immunodeficiency virus can develop accelerated atherosclerosis. The incidence of coronary artery disease in these patients may be twice as high compared with that of HIV-negative individuals with similar characteristics. "Porcelain aorta" is a term used to describe extensive circumferential calcification of the thoracic aorta. The pathophysiology of porcelain aorta is not fully understood. We present a case of a young man who was a smoker and living with HIV since childhood, without other traditional cardiovascular risk factors, who presented to the emergency room with a positive stress test for myocardial ischemia. Transthoracic echocardiography revealed normal regional and global myocardial wall motion, ascending aorta ectasia, and moderate aortic regurgitation. Coronary angiography showed a critical calcified proximal left anterior descending artery stenosis and an important calcification of the thoracic aorta. Therefore, the most important challenge was the management of coronary syndrome in a young person living with HIV, with associated porcelain aorta and aortic regurgitation.
Collapse
|
46
|
Cardiovascular Risk Factors in Patients with Congenital Hemophilia: A Focus on Hypertension. Diagnostics (Basel) 2022; 12:diagnostics12122937. [PMID: 36552943 PMCID: PMC9776547 DOI: 10.3390/diagnostics12122937] [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: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
Aging hemophiliacs face cardiovascular disease. Lots of evidence has been gathered that hemophiliacs have a more unfavorable cardiovascular profile than the general population does, especially due to the increased prevalence of hypertension (HTN). Among the existing scattered evidence, our study provides the most comprehensive and systematized analysis of the determinants of HTN in hemophiliacs. We discussed the contribution to the HTN substrate of hemophilia-specific factors, such as type, severity and the presence of inhibitors. The complex mechanism of kidney dysfunction in relation to hematuria and viral infections was meticulously addressed. Furthermore, we highlighted the new pathogenic concepts of endothelial dysfunction and the association between HTN and hemophilic arthropathy. The clustering of cardiovascular risk factors is common in hemophiliacs, and it enhances the negative vascular effect of HTN and aggravates HTN. It usually leads to an increased risk for coronary and cerebrovascular events. Our work provides reliable evidence to guide and improve the management of HTN in hemophiliacs.
Collapse
|
47
|
Zhang J, Chen S, Chen X. Fatal pulmonary embolism due to invasion of the hepatic inferior vena cava by HIV secondary hepatic abscess: A case report. Asian J Surg 2022; 46:2019-2020. [PMID: 36369138 DOI: 10.1016/j.asjsur.2022.10.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
|
48
|
Medrano-Garcia S, Morales-Cano D, Barreira B, Vera-Zambrano A, Kumar R, Kosanovic D, Schermuly RT, Graham BB, Perez-Vizcaino F, Mathie A, Savai R, Pullamseti S, Butrous G, Fernández-Malavé E, Cogolludo A. HIV and Schistosoma Co-Exposure Leads to Exacerbated Pulmonary Endothelial Remodeling and Dysfunction Associated with Altered Cytokine Landscape. Cells 2022; 11:cells11152414. [PMID: 35954255 PMCID: PMC9368261 DOI: 10.3390/cells11152414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/27/2022] Open
Abstract
HIV and Schistosoma infections have been individually associated with pulmonary vascular disease. Co-infection with these pathogens is very common in tropical areas, with an estimate of six million people co-infected worldwide. However, the effects of HIV and Schistosoma co-exposure on the pulmonary vasculature and its impact on the development of pulmonary vascular disease are largely unknown. Here, we have approached these questions by using a non-infectious animal model based on lung embolization of Schistosoma mansoni eggs in HIV-1 transgenic (HIV) mice. Schistosome-exposed HIV mice but not wild-type (Wt) counterparts showed augmented pulmonary arterial pressure associated with markedly suppressed endothelial-dependent vasodilation, increased endothelial remodeling and vessel obliterations, formation of plexiform-like lesions and a higher degree of perivascular fibrosis. In contrast, medial wall muscularization was similarly increased in both types of mice. Moreover, HIV mice displayed an impaired immune response to parasite eggs in the lung, as suggested by decreased pulmonary leukocyte infiltration, small-sized granulomas, and augmented residual egg burden. Notably, vascular changes in co-exposed mice were associated with increased expression of proinflammatory and profibrotic cytokines, including IFN-γ and IL-17A in CD4+ and γδ T cells and IL-13 in myeloid cells. Collectively, our study shows for the first time that combined pulmonary persistence of HIV proteins and Schistosoma eggs, as it may occur in co-infected people, alters the cytokine landscape and targets the vascular endothelium for aggravated pulmonary vascular pathology. Furthermore, it provides an experimental model for the understanding of pulmonary vascular disease associated with HIV and Schistosoma co-morbidity.
Collapse
Affiliation(s)
- Sandra Medrano-Garcia
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35305 Giessen, Germany
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, 28040 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28040 Madrid, Spain
- Correspondence: (D.M.-C.); (A.C.); Tel.: +34-913947120 (A.C.)
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, 28040 Madrid, Spain
| | - Alba Vera-Zambrano
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, 28040 Madrid, Spain
| | - Rahul Kumar
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Djuro Kosanovic
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Ralph Theo Schermuly
- Department of internal Medicine, Justus-Liebig University, Member of the German Center for Lung Research (DZL), 35305 Giessen, Germany
| | - Brian B. Graham
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, 28040 Madrid, Spain
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham ME4 4BF, UK
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35305 Giessen, Germany
- Department of internal Medicine, Justus-Liebig University, Member of the German Center for Lung Research (DZL), 35305 Giessen, Germany
| | - Soni Pullamseti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35305 Giessen, Germany
- Department of internal Medicine, Justus-Liebig University, Member of the German Center for Lung Research (DZL), 35305 Giessen, Germany
| | - Ghazwan Butrous
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham ME4 4BF, UK
| | - Edgar Fernández-Malavé
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid and Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red Enfermedades Respiratorias, 28040 Madrid, Spain
- Correspondence: (D.M.-C.); (A.C.); Tel.: +34-913947120 (A.C.)
| |
Collapse
|
49
|
Sharma AL, Wang H, Zhang Z, Millien G, Tyagi M, Hongpaisan J. HIV Promotes Neurocognitive Impairment by Damaging the Hippocampal Microvessels. Mol Neurobiol 2022; 59:4966-4986. [PMID: 35665894 PMCID: PMC10071835 DOI: 10.1007/s12035-022-02890-8] [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] [Received: 03/09/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Current evidence suggests that mild cerebrovascular changes could induce neurodegeneration and contribute to HIV-associated neurocognitive disease (HAND) in HIV patients. We investigated both the quantitative and qualitative impact of HIV infection on brain microvessels, especially on hippocampal microvessels, which are crucial for optimal O2 supply, and thus for maintaining memory and cognitive abilities. The results obtained using cultured human brain microvascular endothelial cells (HBMEC) were reproduced using a suitable mouse model and autopsied human HIV hippocampus. In HBMEC, we found significantly higher oxidative stress-dependent apoptotic cell loss following 5 h of treatment of GST-Tat (1 µg/ml) compared to GST (1 µg/ml) control. We noticed complete recovery of HBMEC cells after 24 h of GST-Tat treatment, due to temporal degradation or inactivation of GST-Tat. Interestingly, we found a sustained increase in mitochondrial oxidative DNA damage marker 8-OHdG, as well as an increase in hypoxia-inducible factor hypoxia-inducible factor-1α (HIF-1α). In our mouse studies, upon short-term injection of GST-Tat, we found the loss of small microvessels (mostly capillaries) and vascular endothelial growth factor (VEGF), but not large microvessels (arterioles and venules) in the hippocampus. In addition to capillary loss, in the post-mortem HIV-infected human hippocampus, we observed large microvessels with increased wall cells and perivascular tissue degeneration. Together, our data show a crucial role of Tat in inducing HIF-1α-dependent inhibition of mitochondrial transcriptional factor A (TFAM) and dilated perivascular space. Thus, our results further define the underlying molecular mechanism promoting mild cerebrovascular disease, neuropathy, and HAND pathogenesis in HIV patients.
Collapse
Affiliation(s)
- Adhikarimayum Lakhikumar Sharma
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Huaixing Wang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Zongxiu Zhang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Guetchyn Millien
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Mudit Tyagi
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| |
Collapse
|
50
|
Palakeel JJ, Ali M, Chaduvula P, Chhabra S, Lamsal Lamichhane S, Ramesh V, Opara CO, Khan FY, Kabiraj G, Kauser H, Mostafa JA. An Outlook on the Etiopathogenesis of Pulmonary Hypertension in HIV. Cureus 2022; 14:e27390. [PMID: 36046315 PMCID: PMC9418639 DOI: 10.7759/cureus.27390] [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: 06/15/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
Although overall survival rates of patients infected with human immunodeficiency virus (HIV) have been significantly improved by antiretroviral therapy (ART), chronic comorbidities associated with HIV result in a worsening quality of life. Pulmonary arterial hypertension (PAH) is the most prevalent comorbidity associated with HIV infection. Despite low viremia and a non-replicative state maintained by ART, few people develop PAH. Previous data from animal models and human pulmonary microvascular endothelial cells (HPMVECs) suggests a constellation of events occurring during the propagation of HIV-associated PAH (HIV-PAH). However, these studies have not successfully isolated HIV virions, HIV-DNA, protein 24 antigen (p24), or HIV-RNA from the pulmonary endothelial cells (ECs). It provides an insight into an ongoing inflammatory process that could be attributed to viral proteins. Several studies have demonstrated the role of viral proteins on vascular remodeling. A composite of chronic inflammatory changes mediated by cytokines and growth factors along with several inciting risk factors such as Hepatitis C virus (HCV) co-infection, genetic factors, male predominance, illegal drug usage, and duration of HIV infection have led to molecular changes that result in an initial phase of apoptosis followed by the formation of apoptotic resistant hyperproliferative ECs with altered phenotype. This study aims to identify the risk factors and mechanisms behind HIV-PAH pathobiology at the host-pathogen interface at the intracellular level.
Collapse
|