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
|
Iskhakov D, Deleger JN, Plutzky J, Triant VA, Hyle EP. Cardiovascular Outcomes After Acute Coronary Syndrome in Persons Living with HIV: A Scoping Review. Curr Cardiol Rep 2025; 27:84. [PMID: 40227544 PMCID: PMC11999250 DOI: 10.1007/s11886-024-02186-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 04/15/2025]
Abstract
PURPOSE OF REVIEW Over the past twenty-five years, a growing body of research has investigated outcomes after an acute coronary syndrome (ACS) event in persons with HIV (PWH). In this scoping review, we assessed the published literature on outcomes after a prior ACS event in PWH compared to persons without HIV (PWoH) in the US and Europe. RECENT FINDINGS Of 27 studies of secondary prevention of cardiovascular disease, 15 studies found a greater incidence of recurrent ACS, heart failure, in-stent thrombosis, revascularization, restenosis, and mortality in PWH after a prior ACS event compared to PWoH. PWH were more likely to present with STEMI, less likely to receive percutaneous coronary intervention, and more likely to exhibit an active inflammatory state. The remaining 12 studies found no significant difference in outcomes after a prior ACS event. PWH may face an increased risk of adverse outcomes after ACS that differ in presentation and management compared with PWoH. Larger observational studies are needed to assess outcomes after a prior ACS event in PWH compared to PWoH and improve clinical management.
Collapse
Affiliation(s)
- David Iskhakov
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Julie N Deleger
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jorge Plutzky
- Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Virginia A Triant
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Emily P Hyle
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Harvard University, Boston, MA, USA.
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Zheng Q, Wu Y, Zhang X, Zhang Y, Zhu Z, Luan B, Zang P, Sun D. Analysis and validation of hub genes for atherosclerosis and AIDS and immune infiltration characteristics based on bioinformatics and machine learning. Sci Rep 2025; 15:12316. [PMID: 40210656 PMCID: PMC11985999 DOI: 10.1038/s41598-025-96907-6] [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: 08/19/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Atherosclerosis is the major cause of cardiovascular diseases worldwide, and AIDS linked with chronic inflammation and immune activation, increases atherosclerosis risk. The application of bioinformatics and machine learning to identify hub genes for atherosclerosis and AIDS has yet to be reported. Thus, this study aims to identify the hub genes for atherosclerosis and AIDS. Gene expression profiles were downloaded from the Gene Expression Omnibus database. The Robust Multichip Average was performed for data preprocessing, and the limma package was used for screening differentially expressed genes. Enrichment analysis employed GO and KEGG, protein-protein interaction network was constructed. Hub genes were filtered using topological and machine learning algorithms and validated in external cohorts. Then immune infiltration and correlation analysis of hub genes were constructed. Nomogram, receiver operating curve, and single-sample gene set enrichment analysis were applied to evaluate hub genes. This study identified 48 intersecting genes. Enrichment analyses indicated that these genes are significantly enriched in viral response, inflammatory response, and cytokine signaling pathways. CCR5 and OAS1 were identified as common hub genes in atherosclerosis and AIDS for the first time, highlighting their roles in antiviral immunity, inflammation and immune infiltration. These findings contributed to understanding the shared pathogenesis of Atherosclerosis and AIDS and provided possible potential therapeutic targets for immunomodulatory therapy.
Collapse
Affiliation(s)
- Qirui Zheng
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Yupeng Wu
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Pan-Vascular Management Center, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Xiaojiao Zhang
- Department of Cardiology, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Yuzhu Zhang
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Zaihan Zhu
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Bo Luan
- Department of Cardiology, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Peizhuo Zang
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China.
- Pan-Vascular Management Center, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
- Liaoning Provincial Key Laboratory of Neurointerventional Therapy and Biomaterials Research and Development, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
| | - Dandan Sun
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China.
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
- Liaoning Provincial Key Laboratory of Neurointerventional Therapy and Biomaterials Research and Development, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
| |
Collapse
|
4
|
Sheets K, Baker JV. HIV and Inflamm-Aging: How Do We Reach the Summit of Healthy Aging? TOPICS IN ANTIVIRAL MEDICINE 2024; 32:589-596. [PMID: 39765238 PMCID: PMC11737810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
People with HIV (PWH) are living longer and experiencing a greater burden of morbidity from non-AIDS-defining conditions. Chronically treated HIV disease is associated with ongoing systemic inflammation that contributes to the development of chronic conditions (eg, cardiovascular disease) and geriatric syndromes (eg, frailty). Apart from HIV disease, a progressive increase in systemic inflammation is a characteristic feature of biologic aging, a process described as "inflammaging." Inflamm-aging is driven by persistent antigen stimulation and stress, leading to an immune profile characterized by elevated levels of blood inflammatory markers and cellular activation and senescence. Chronic HIV disease is hypothesized to accentuate the immune profile of inflamm-aging, in part through viral persistence in lymphatic tissues, permanent injury impairing immune recovery, the presence of copathogens, gut dysbiosis and microbial translocation, and chromosomal and genetic alterations associated with immune activation. Few strategies exist for safe and effective modulation of systemic inflammation among older PWH. The strongest current evidence supports aggressive management of modifiable risk factors such as lipids, blood pressure, and levels of physical activity. Future inflamm-aging research should be directed toward advancing the implementation of proven approaches, such as physical activity, as well as studying novel mechanisms of, and treatments for, inflamm-aging among PWH.
Collapse
Affiliation(s)
- Kerry Sheets
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| | - Jason V. Baker
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| |
Collapse
|
5
|
Ding AK, Wallis ZK, White KS, Sumer CE, Kim WK, Ardeshir A, Williams KC. Galectin-3, Galectin-9, and Interleukin-18 Are Associated with Monocyte/Macrophage Activation and Turnover More so than Simian Immunodeficiency Virus-Associated Cardiac Pathology or Encephalitis. AIDS Res Hum Retroviruses 2024; 40:531-542. [PMID: 38787309 PMCID: PMC11905219 DOI: 10.1089/aid.2024.0008] [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: 05/25/2024] Open
Abstract
Despite antiretroviral therapy (ART), people living with HIV (PLWH) are at increased risk of developing cardiovascular disease (CVD) and HIV-associated neurocognitive disorder (HAND), among other comorbidities. Studies from ART-treated individuals identified galectin-3 (gal-3) and interleukin (IL)-18 as CVD biomarkers, galectin-9 (gal-9) as a HAND biomarker, and sCD163, a marker of monocyte/macrophage activation, as a biomarker of both. We asked if plasma gal-3, gal-9, and IL-18 are associated with an individual comorbidity or increase in both with animals that develop AIDS with both pathologies versus (CVD-path) alone or simian immunodeficiency virus encephalitis (SIVE) alone. We found that no biomarkers were selective between individual pathologies, and all biomarkers increased with co-development of CVD-path and SIVE (gal-3, p = 0.11; gal-9, p = 0.001; IL-18, p = 0.007; sCD163, p < 0.001; %BrdU p = 0.02). Although gal-3, gal-9, and IL-18 did not distinguish between pathologies, they correlated strongly with one another, with sCD163, a marker of monocyte/macrophage activation, and the %BrdU monocytes, a marker of monocyte turnover. Compared to animals with CVD-path or SIVE alone, animals that co-developed both pathologies had consistently elevated IL-18 throughout infection (p = 0.02) and increased sCD163 in late infection (p = 0.01). These data indicate that gal-3, gal-9, and IL-18 are associated with monocyte/macrophage activation by sCD163 and monocyte turnover by the %BrdU+ monocytes more so than CVD-path or SIVE.
Collapse
Affiliation(s)
- Andrew K Ding
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Zoey K Wallis
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Kevin S White
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Cinar Efe Sumer
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Amir Ardeshir
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kenneth C Williams
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
6
|
Peters BA, Hanna DB, Xue X, Weber K, Appleton AA, Kassaye SG, Topper E, Tracy RP, Guillemette C, Caron P, Tien PC, Qi Q, Burk RD, Sharma A, Anastos K, Kaplan RC. Menopause and Estrogen Associations With Gut Barrier, Microbial Translocation, and Immune Activation Biomarkers in Women With and Without HIV. J Acquir Immune Defic Syndr 2024; 96:214-222. [PMID: 38905473 PMCID: PMC11196004 DOI: 10.1097/qai.0000000000003419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/29/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVES Estrogens may protect the gut barrier and reduce microbial translocation and immune activation, which are prevalent in HIV infection. We investigated relationships of the menopausal transition and estrogens with gut barrier, microbial translocation, and immune activation biomarkers in women with and without HIV. DESIGN Longitudinal and cross-sectional studies nested in the Women's Interagency HIV Study. METHODS Intestinal fatty acid binding protein, lipopolysaccharide binding protein, and soluble CD14 (sCD14) levels were measured in serum from 77 women (43 with HIV) before, during, and after the menopausal transition (∼6 measures per woman over ∼13 years). A separate cross-sectional analysis was conducted among 72 postmenopausal women with HIV with these biomarkers and serum estrogens. RESULTS Women in the longitudinal analysis were a median age of 43 years at baseline. In piecewise, linear, mixed-effects models with cutpoints 2 years before and after the final menstrual period to delineate the menopausal transition, sCD14 levels increased over time during the menopausal transition (Beta [95% CI]: 38 [12 to 64] ng/mL/yr, P = 0.004), followed by a decrease posttransition (-46 [-75 to -18], P = 0.001), with the piecewise model providing a better fit than a linear model (P = 0.0006). In stratified analyses, these results were only apparent in women with HIV. In cross-sectional analyses, among women with HIV, free estradiol inversely correlated with sCD14 levels (r = -0.26, P = 0.03). Lipopolysaccharide binding protein and intestinal fatty acid binding protein levels did not appear related to the menopausal transition and estrogen levels. CONCLUSIONS Women with HIV may experience heightened innate immune activation during menopause, possibly related to the depletion of estrogens.
Collapse
Affiliation(s)
- Brandilyn A. Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathleen Weber
- Cook County Health/Hektoen Institute of Medicine, Chicago, IL, USA
| | - Allison A. Appleton
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, Rensselaer, NY, United States
| | | | - Elizabeth Topper
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec - Université Laval Research Center, Cancer research center (CRC) and Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Patrick Caron
- Centre Hospitalier Universitaire (CHU) de Québec - Université Laval Research Center, Cancer research center (CRC) and Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Phyllis C. Tien
- Department of Veterans Affairs Medical Center, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert D. Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Microbiology and Immunology and Obstetrics & Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
7
|
Hmiel L, Zhang S, Obare LM, Santana MADO, Wanjalla CN, Titanji BK, Hileman CO, Bagchi S. Inflammatory and Immune Mechanisms for Atherosclerotic Cardiovascular Disease in HIV. Int J Mol Sci 2024; 25:7266. [PMID: 39000373 PMCID: PMC11242562 DOI: 10.3390/ijms25137266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Atherosclerotic vascular disease disproportionately affects persons living with HIV (PLWH) compared to those without. The reasons for the excess risk include dysregulated immune response and inflammation related to HIV infection itself, comorbid conditions, and co-infections. Here, we review an updated understanding of immune and inflammatory pathways underlying atherosclerosis in PLWH, including effects of viral products, soluble mediators and chemokines, innate and adaptive immune cells, and important co-infections. We also present potential therapeutic targets which may reduce cardiovascular risk in PLWH.
Collapse
Affiliation(s)
- Laura Hmiel
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Suyu Zhang
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Boghuma K. Titanji
- Division of Infectious Diseases, Emory University, Atlanta, GA 30322, USA
| | - Corrilynn O. Hileman
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Shashwatee Bagchi
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
8
|
Dix C, Dolan G, Hunt BJ. Reducing the risk of atherosclerotic cardiovascular disease in people with hemophilia: the importance of primary prevention. J Thromb Haemost 2024; 22:1304-1312. [PMID: 38309435 DOI: 10.1016/j.jtha.2024.01.017] [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: 11/27/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Revolutionary advances in the treatment of hemophilia has led to a significant improvement in life expectancy. Associated with this has been an increase in age-related diseases especially atherosclerotic cardiovascular disease (CVD). While people with hemophilia (PWH) develop atherosclerosis at rates similar to those of the general population, rates of atherothrombosis and mortality related to CVD have been much lower, due to their hypocoagulable state. Changing treatment paradigms, aimed at reducing the risk of bleeding by improving hemostasis to levels approaching normality, has meant that the protection they are thought to have had may be lost. CVD risk factors are just as common in PWH as in the general population, but appear to be undertreated. In particular, primary prevention of CVD is vital in all individuals, but particularly in PWH as treatment of established CVD can be difficult. Active identification and management of CVD risk factors, such as obesity, physical inactivity, hypertension, and hypercholesterolemia, is required. In particular, statins have been shown to significantly reduce cardiovascular and all-cause mortality with few adverse events and no increased risk of bleeding in the general population, and their use needs urgent assessment in PWH. Further longitudinal research into preventing CVD in PWH, including accurate CVD risk assessment, is required to optimize prevention and management.
Collapse
Affiliation(s)
- Caroline Dix
- Haemophilia & Thrombosis Centre, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Gerry Dolan
- Haemophilia & Thrombosis Centre, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Beverley J Hunt
- Haemophilia & Thrombosis Centre, Guy's & St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
9
|
Nazari I, Feinstein MJ. Evolving mechanisms and presentations of cardiovascular disease in people with HIV: implications for management. Clin Microbiol Rev 2024; 37:e0009822. [PMID: 38299802 PMCID: PMC10938901 DOI: 10.1128/cmr.00098-22] [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: 02/02/2024] Open
Abstract
People with HIV (PWH) are at elevated risk for cardiovascular diseases (CVDs), including myocardial infarction, heart failure, and sudden cardiac death, among other CVD manifestations. Chronic immune dysregulation resulting in persistent inflammation is common among PWH, particularly those with sustained viremia and impaired CD4+ T cell recovery. This inflammatory milieu is a major contributor to CVDs among PWH, in concert with common comorbidities (such as dyslipidemia and smoking) and, to a lesser extent, off-target effects of antiretroviral therapy. In this review, we discuss the clinical and mechanistic evidence surrounding heightened CVD risks among PWH, implications for specific CVD manifestations, and practical guidance for management in the setting of evolving data.
Collapse
Affiliation(s)
- Ilana Nazari
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew J. Feinstein
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
10
|
Borjabad B, Inciarte A, Chivite I, Gonzalez-Cordon A, Mosquera M, Hurtado C, Rovira C, Gonzalez T, Sempere A, Torres B, Calvo J, De La Mora L, Martinez-Rebollar M, Laguno M, Foncillas A, Ambrosioni J, Blanch J, Rodriguez A, Solbes E, Llobet R, Berrocal L, Mallolas J, Miro JM, Alcami J, Blanco JL, Sanchez-Palomino S, De Lazzari E, Martinez E. Efficacy and safety of raltegravir plus lamivudine maintenance therapy. J Antimicrob Chemother 2024; 79:255-261. [PMID: 38039097 DOI: 10.1093/jac/dkad364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Decreasing medication burden with raltegravir plus lamivudine in virologically suppressed persons with HIV (PWH) maintained efficacy and was well tolerated at 24 weeks, but more comprehensive data over longer follow-up are required. METHODS Prospective 48 week extension phase of the raltegravir plus lamivudine arm from a previous 24 week pilot randomized clinical trial in which virologically suppressed PWH were randomized 2:1 to switch to fixed-dose combination 150 mg lamivudine/300 mg raltegravir twice daily or to continue therapy. In this 48 week extension phase, raltegravir was dosed at 1200 mg/day and lamivudine 300 mg/day. Primary outcome was the proportion of PWH with treatment failure at Week 48. Secondary outcomes were changes in ultrasensitive plasma HIV RNA, HIV DNA in CD4 cells, serum IL-6, ultrasensitive C-reactive protein and sCD14, body composition, sleep quality, quality of life and adverse effects. RESULTS Between May 2018 and June 2019, 33 PWH were enrolled. One participant experienced virological failure without resistance mutations and re-achieved sustained virological suppression without therapy discontinuation, and two others discontinued therapy due to adverse effects. Treatment failure was 9% (95% CI 2%-24%) and 3% (95% CI 0%-17%) in the ITT and on-treatment populations. There were significant changes between baseline and Week 48 in serum cytokines but not in other secondary outcomes. CONCLUSIONS Switching to raltegravir and lamivudine in PWH with virological suppression maintains efficacy and is well tolerated. This maintenance regimen might be a cost-effective option for PWH at risk of drug-drug interactions or needing to avoid specific toxicities of certain antiretroviral drugs or their negative impact on comorbidities.
Collapse
Affiliation(s)
- Beatriz Borjabad
- Internal Medicine Service, Hospital Moises Broggi, Sant Joan Despí, Spain
| | - Alexy Inciarte
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Ivan Chivite
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Ana Gonzalez-Cordon
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Mosquera
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Hurtado
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Cristina Rovira
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Tania Gonzalez
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Abiu Sempere
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Berta Torres
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julia Calvo
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Lorena De La Mora
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Martinez-Rebollar
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Laguno
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Foncillas
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Juan Ambrosioni
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Blanch
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- Internal Medicine Service, Hospital Universitari de Santa Maria, Lleida, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Rodriguez
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Estela Solbes
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Roger Llobet
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Leire Berrocal
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
| | - Josep Mallolas
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M Miro
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Alcami
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Acquired Immunodeficiency Syndrome (AIDS) Immunopathology Unit, National Center for Microbiology, Institute of Health Carlos III, Majadahonda, Spain
| | - Jose L Blanco
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Sonsoles Sanchez-Palomino
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisa De Lazzari
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Esteban Martinez
- Infectious Diseases Unit, Hospital Clínic, University of Barcelona 08036, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Wiche Salinas TR, Zhang Y, Gosselin A, Rosario NF, El-Far M, Filali-Mouhim A, Routy JP, Chartrand-Lefebvre C, Landay AL, Durand M, Tremblay CL, Ancuta P. Alterations in Th17 Cells and Non-Classical Monocytes as a Signature of Subclinical Coronary Artery Atherosclerosis during ART-Treated HIV-1 Infection. Cells 2024; 13:157. [PMID: 38247848 PMCID: PMC10813976 DOI: 10.3390/cells13020157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Cardiovascular disease (CVD) remains an important comorbidity in people living with HIV-1 (PLWH) receiving antiretroviral therapy (ART). Our previous studies performed in the Canadian HIV/Aging Cohort Study (CHACS) (>40 years-old; Framingham Risk Score (FRS) > 5%) revealed a 2-3-fold increase in non-calcified coronary artery atherosclerosis (CAA) plaque burden, measured by computed tomography angiography scan (CTAScan) as the total (TPV) and low attenuated plaque volume (LAPV), in ART-treated PLWH (HIV+) versus uninfected controls (HIV-). In an effort to identify novel correlates of subclinical CAA, markers of intestinal damage (sCD14, LBP, FABP2); cell trafficking/inflammation (CCL20, CX3CL1, MIF, CCL25); subsets of Th17-polarized and regulatory (Tregs) CD4+ T-cells, classical/intermediate/non-classical monocytes, and myeloid/plasmacytoid dendritic cells were studied in relationship with HIV and TPV/LAPV status. The TPV detection/values coincided with higher plasma sCD14, FABP2, CCL20, MIF, CX3CL1, and triglyceride levels; lower Th17/Treg ratios; and classical monocyte expansion. Among HIV+, TPV+ versus TPV- exhibited lower Th17 frequencies, reduced Th17/Treg ratios, higher frequencies of non-classical CCR9lowHLADRhigh monocytes, and increased plasma fibrinogen levels. Finally, Th17/Treg ratios and non-classical CCR9lowHLADRhigh monocyte frequencies remained associated with TPV/LAPV after adjusting for FRS and HIV/ART duration in a logistic regression model. These findings point to Th17 paucity and non-classical monocyte abundance as novel immunological correlates of subclinical CAA that may fuel the CVD risk in ART-treated PLWH.
Collapse
Affiliation(s)
- Tomas Raul Wiche Salinas
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada; (T.R.W.S.); (Y.Z.); (C.L.T.)
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Yuwei Zhang
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada; (T.R.W.S.); (Y.Z.); (C.L.T.)
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Annie Gosselin
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Natalia Fonseca Rosario
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Mohamed El-Far
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Ali Filali-Mouhim
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Carl Chartrand-Lefebvre
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada
| | | | - Madeleine Durand
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
- Département de Médecine, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada
| | - Cécile L. Tremblay
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada; (T.R.W.S.); (Y.Z.); (C.L.T.)
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| | - Petronela Ancuta
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal (UdeM), Montreal, QC H2X 0A9, Canada; (T.R.W.S.); (Y.Z.); (C.L.T.)
- CRCHUM, Montreal, QC H2X 0A2, Canada; (A.G.); (N.F.R.); (M.E.-F.); (A.F.-M.); (C.C.-L.); (M.D.)
| |
Collapse
|
12
|
Wiche Salinas TR, Zhang Y, Gosselin A, Do Rosario NF, El-Far M, Filali-Mouhim A, Routy JP, Chartrand-Lefebvre C, Landay AL, Durand M, Tremblay CL, Ancuta P. A Blood Immunological Signature of Subclinical Coronary Artery Atherosclerosis in People Living with HIV-1 Receiving Antiretroviral Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571922. [PMID: 38187644 PMCID: PMC10769180 DOI: 10.1101/2023.12.15.571922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Cardiovascular disease (CVD) remains an important co-morbidity in people living with HIV-1 (PLWH) receiving antiretroviral therapy (ART). Our previous studies performed on the Canadian HIV/Aging Cohort Study (CHACS) (>40 years-old; Framingham Risk Score (FRS) >5%), revealed a 2-3-fold increase in non-calcified coronary artery atherosclerosis (CAA) plaque burden, measured by Computed tomography angiography scan (CTAScan) as total (TPV) and low attenuated plaque volume (LAPV) in ART-treated PLWH (HIV+) versus uninfected controls (HIV-). In an effort to identify novel correlates of subclinical CAA, markers of intestinal damage (sCD14, LBP, FABP2); cell trafficking/inflammation (CCL20, CX3CL1, MIF, CCL25); subsets of Th17-polarized and regulatory (Tregs) CD4 + T-cells, classical/intermediate/non-classical monocytes, and myeloid/plasmacytoid dendritic cells, were studied in relationship with HIV and TPV/LAPV status. The TPV detection/values coincided with higher plasma sCD14, FABP2, CCL20, MIF, CX3CL1 and triglyceride levels, lower Th17/Treg ratios, and classical monocyte expansion. Among HIV + , TPV + versus TPV - exhibited lower Th17 frequencies, reduced Th17/Treg ratios, higher frequencies of non-classical CCR9 low HLADR high monocyte, and increased plasma fibrinogen levels. Finally, Th17/Treg ratios and non-classical CCR9 low HLADR high monocyte frequencies remained associated with TPV/LAPV after adjusting for FRS and HIV/ART duration in a logistic regression model. These findings point to Th17 paucity and non-classical monocyte abundance as novel immunological correlates of subclinical CAA that may fuel the CVD risk in ART-treated PLWH.
Collapse
|
13
|
White KS, Walker JA, Wang J, Autissier P, Miller AD, Abuelezan NN, Burrack R, Li Q, Kim WK, Williams KC. Simian immunodeficiency virus-infected rhesus macaques with AIDS co-develop cardiovascular pathology and encephalitis. Front Immunol 2023; 14:1240946. [PMID: 37965349 PMCID: PMC10641955 DOI: 10.3389/fimmu.2023.1240946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/03/2023] [Indexed: 11/16/2023] Open
Abstract
Despite effective antiretroviral therapy, HIV co-morbidities remain where central nervous system (CNS) neurocognitive disorders and cardiovascular disease (CVD)-pathology that are linked with myeloid activation are most prevalent. Comorbidities such as neurocogntive dysfunction and cardiovascular disease (CVD) remain prevalent among people living with HIV. We sought to investigate if cardiac pathology (inflammation, fibrosis, cardiomyocyte damage) and CNS pathology (encephalitis) develop together during simian immunodeficiency virus (SIV) infection and if their co-development is linked with monocyte/macrophage activation. We used a cohort of SIV-infected rhesus macaques with rapid AIDS and demonstrated that SIV encephalitis (SIVE) and CVD pathology occur together more frequently than SIVE or CVD pathology alone. Their co-development correlated more strongly with activated myeloid cells, increased numbers of CD14+CD16+ monocytes, plasma CD163 and interleukin-18 (IL-18) than did SIVE or CVD pathology alone, or no pathology. Animals with both SIVE and CVD pathology had greater numbers of cardiac macrophages and increased collagen and monocyte/macrophage accumulation, which were better correlates of CVD-pathology than SIV-RNA. Animals with SIVE alone had higher levels of activated macrophage biomarkers and cardiac macrophage accumulation than SIVnoE animals. These observations were confirmed in HIV infected individuals with HIV encephalitis (HIVE) that had greater numbers of cardiac macrophages and fibrosis than HIV-infected controls without HIVE. These results underscore the notion that CNS and CVD pathologies frequently occur together in HIV and SIV infection, and demonstrate an unmet need for adjunctive therapies targeting macrophages.
Collapse
Affiliation(s)
- Kevin S. White
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Joshua A. Walker
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - John Wang
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Patrick Autissier
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Andrew D. Miller
- Department of Biomedical Sciences, Section of Anatomic Physiology, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| | - Nadia N. Abuelezan
- Connel School of Nursing, Boston College, Chestnut Hill, MA, United States
| | - Rachel Burrack
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | | |
Collapse
|
14
|
Remch M, Franceschini N, Davy-Mendez T, Floris-Moore M, Napravnik S. Hypertension burden, treatment, and control among people with HIV at a clinical care center in the Southeastern US, 2014-2019. AIDS Care 2023; 35:1594-1603. [PMID: 36524873 PMCID: PMC10272289 DOI: 10.1080/09540121.2022.2148626] [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: 08/16/2021] [Accepted: 11/14/2022] [Indexed: 12/23/2022]
Abstract
Hypertension management outcomes in people with HIV (PWH) are not well characterized, despite high hypertension burden. We assessed hypertension prevalence, incidence, treatment, and outcomes among patients with HIV at a clinical center in the southeastern US, from 2014 to 2019. To identify characteristics associated with treatment and outcomes, we estimated adjusted risk ratios (aRR) and 95% confidence intervals (CI). Among 2274 patients, 72% were cisgender men, 56% non-Hispanic Black, median age 47 years, 48% MSM, 12% had CD4 cell count <200 cells/μl, 72% HIV RNA level <400 copies/mL and 39% prevalent hypertension. Hypertension incidence rate was 6.3/100 person-years (95% CI, 5.6-7.0). Among incident hypertension cases (n = 275), 16% (95% CI, 11-20) initiated an antihypertensive within one year. Compared to non-Hispanic white patients, Hispanic (aRR, 6.68; 95% CI, 1.50-29.74) and non-Hispanic Black patients (aRR, 2.18; 95% CI, 0.91-5.24) were more likely to initiate an antihypertensive. Among patients initiating an antihypertensive (n = 178), 63% (95% CI 56-70) experienced blood pressure control within one year. Patients with HIV experienced a high burden of hypertension with notable delays in antihypertensive initiation, as well as gaps in achieving blood pressure control, highlighting opportunities for interventions designed to minimize delays in controlling hypertension in this vulnerable population.
Collapse
Affiliation(s)
- Molly Remch
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Thibaut Davy-Mendez
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, CA, USA
| | - Michelle Floris-Moore
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Sonia Napravnik
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
15
|
Wang Z, Peters BA, Bryant M, Hanna DB, Schwartz T, Wang T, Sollecito CC, Usyk M, Grassi E, Wiek F, Peter LS, Post WS, Landay AL, Hodis HN, Weber KM, French A, Golub ET, Lazar J, Gustafson D, Sharma A, Anastos K, Clish CB, Burk RD, Kaplan RC, Knight R, Qi Q. Gut microbiota, circulating inflammatory markers and metabolites, and carotid artery atherosclerosis in HIV infection. MICROBIOME 2023; 11:119. [PMID: 37237391 PMCID: PMC10224225 DOI: 10.1186/s40168-023-01566-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/05/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Alterations in gut microbiota have been implicated in HIV infection and cardiovascular disease. However, how gut microbial alterations relate to host inflammation and metabolite profiles, and their relationships with atherosclerosis, have not been well-studied, especially in the context of HIV infection. Here, we examined associations of gut microbial species and functional components measured by shotgun metagenomics with carotid artery plaque assessed by B-mode carotid artery ultrasound in 320 women with or at high risk of HIV (65% HIV +) from the Women's Interagency HIV Study. We further integrated plaque-associated microbial features with serum proteomics (74 inflammatory markers measured by the proximity extension assay) and plasma metabolomics (378 metabolites measured by liquid chromatography tandem mass spectrometry) in relation to carotid artery plaque in up to 433 women. RESULTS Fusobacterium nucleatum, a potentially pathogenic bacteria, was positively associated with carotid artery plaque, while five microbial species (Roseburia hominis, Roseburia inulinivorans, Johnsonella ignava, Odoribacter splanchnicus, Clostridium saccharolyticum) were inversely associated with plaque. Results were consistent between women with and without HIV. Fusobacterium nucleatum was positively associated with several serum proteomic inflammatory markers (e.g., CXCL9), and the other plaque-related species were inversely associated with proteomic inflammatory markers (e.g., CX3CL1). These microbial-associated proteomic inflammatory markers were also positively associated with plaque. Associations between bacterial species (especially Fusobacterium nucleatum) and plaque were attenuated after further adjustment for proteomic inflammatory markers. Plaque-associated species were correlated with several plasma metabolites, including the microbial metabolite imidazole-propionate (ImP), which was positively associated with plaque and several pro-inflammatory markers. Further analysis identified additional bacterial species and bacterial hutH gene (encoding enzyme histidine ammonia-lyase in ImP production) associated with plasma ImP levels. A gut microbiota score based on these ImP-associated species was positively associated with plaque and several pro-inflammatory markers. CONCLUSION Among women living with or at risk of HIV, we identified several gut bacterial species and a microbial metabolite ImP associated with carotid artery atherosclerosis, which might be related to host immune activation and inflammation. Video Abstract.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - MacKenzie Bryant
- Department of Pediatrics, University of California, La Jolla, San Diego, CA, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tara Schwartz
- Department of Pediatrics, University of California, La Jolla, San Diego, CA, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Mykhaylo Usyk
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evan Grassi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Fanua Wiek
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lauren St Peter
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wendy S Post
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Howard N Hodis
- Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Audrey French
- Department of Internal Medicine, Stroger Hospital of Cook County, Chicago, IL, USA
| | - Elizabeth T Golub
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jason Lazar
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Deborah Gustafson
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, La Jolla, San Diego, CA, USA
- Department of Bioengineering, University of California, La Jolla, San Diego, CA, USA
- Department of Computer Science and Engineering, University of California, La Jolla, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, La Jolla, San Diego, CA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
16
|
Lin J, Ehinger E, Hanna DB, Qi Q, Wang T, Ghosheh Y, Mueller K, Anastos K, Lazar JM, Mack WJ, Tien PC, Berman JW, Cohen MH, Ofotokun I, Gange S, Liu C, Heath SL, Tracy RP, Hodis HN, Landay AL, Ley K, Kaplan RC. HIV infection and cardiovascular disease have both shared and distinct monocyte gene expression features: Women's Interagency HIV study. PLoS One 2023; 18:e0285926. [PMID: 37205656 PMCID: PMC10198505 DOI: 10.1371/journal.pone.0285926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023] Open
Abstract
Persistent inflammation contributes to the development of cardiovascular disease (CVD) as an HIV-associated comorbidity. Innate immune cells such as monocytes are major drivers of inflammation in men and women with HIV. The study objectives are to examine the contribution of circulating non-classical monocytes (NCM, CD14dimCD16+) and intermediate monocytes (IM, CD14+CD16+) to the host response to long-term HIV infection and HIV-associated CVD. Women with and without chronic HIV infection (H) were studied. Subclinical CVD (C) was detected as plaques imaged by B-mode carotid artery ultrasound. The study included H-C-, H+C-, H-C+, and H+C+ participants (23 of each, matched on race/ethnicity, age and smoking status), selected from among enrollees in the Women's Interagency HIV Study. We assessed transcriptomic features associated with HIV or CVD alone or comorbid HIV/CVD comparing to healthy (H-C-) participants in IM and NCM isolated from peripheral blood mononuclear cells. IM gene expression was little affected by HIV alone or CVD alone. In IM, coexisting HIV and CVD produced a measurable gene transcription signature, which was abolished by lipid-lowering treatment. In NCM, versus non-HIV controls, women with HIV had altered gene expression, irrespective of whether or not they had comorbid CVD. The largest set of differentially expressed genes was found in NCM among women with both HIV and CVD. Genes upregulated in association with HIV included several potential targets of drug therapies, including LAG3 (CD223). In conclusion, circulating monocytes from patients with well controlled HIV infection demonstrate an extensive gene expression signature which may be consistent with the ability of these cells to serve as potential viral reservoirs. Gene transcriptional changes in HIV patients were further magnified in the presence of subclinical CVD.
Collapse
Affiliation(s)
- Juan Lin
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Erik Ehinger
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Yanal Ghosheh
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Karin Mueller
- Department of Cardiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jason M. Lazar
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - Wendy J. Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Phyllis C. Tien
- Department of Medicine, and Department of Veterans Affairs, Medical Center, University of California, San Francisco, San Francisco, CA, United States of America
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Mardge H. Cohen
- Department of Medicine, John Stroger Hospital and Rush University, Chicago, IL, United States of America
| | - Igho Ofotokun
- Department of Medicine, Infectious Disease Division and Grady Health Care System, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Stephen Gange
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Chenglong Liu
- Department of Medicine, Georgetown University Medical Center, Washington, DC, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine and Biochemistry, University of Vermont Larner College of Medicine, Colchester, VT, United States of America
| | - Howard N. Hodis
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States of America
| | - Klaus Ley
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
- Department of Bioengineering, University of California San Diego, San Diego, CA, United States of America
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, United States of America
| |
Collapse
|
17
|
Pérez-López FR, Fernández-Alonso AM, Mezones-Holguín E, Vieira-Baptista P. Low genitourinary tract risks in women living with the human immunodeficiency virus. Climacteric 2023:1-7. [PMID: 37054721 DOI: 10.1080/13697137.2023.2194528] [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: 04/15/2023]
Abstract
This review analyzes the clinical associations between specific low genitourinary tract clinical circumstances in perimenopausal and postmenopausal women living with human immunodeficiency virus (WLHIV). Modern antiretroviral therapy (ART) improves survival and reduces opportunistic infections and HIV transmission. Despite appropriate ART, WLHIV may display menstrual dysfunction, risk of early menopause, vaginal microbiome alterations, vaginal dryness, dyspareunia, vasomotor symptoms and low sexual function as compared to women without the infection. They have increased risks of intraepithelial and invasive cervical, vaginal and vulvar cancers. The reduced immunity capacity may also increase the risk of urinary tract infections, side-effects or toxicity of ARTs, and opportunistic infections. Menstrual dysfunction and early menopause may contribute to the early onset of vascular atherosclerosis and plaque formation, and increased osteoporosis risks requiring specific early interventions. On the other hand, the association between being postmenopausal and having a low sexual function is significant and related to low adherence to ART. WLHIV deserve a specific approach to manage different low genitourinary risks and complications related to hormone dysfunction and early menopause.
Collapse
Affiliation(s)
- F R Pérez-López
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain
- Obstetrics and Reproduction, Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
| | | | - E Mezones-Holguín
- Centro de Estudios Económicos y Sociales en Salud, Universidad San Ignacio de Loyola, Lima, Perú
| | - P Vieira-Baptista
- Department of Gynecology-Obstetrics and Pediatrics, Hospital Lusíadas Porto, Porto, Portugal
- Lower Genital Tract Unit, Centro Hospitalar de São João, Porto, Portugal
- Department of Gynecology-Obstetrics and Pediatrics, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| |
Collapse
|
18
|
Yap YJ, Wong PF, AbuBakar S, Sam SS, Shunmugarajoo A, Soh YH, Misbah S, Ab Rahman AK. The clinical utility of CD163 in viral diseases. Clin Chim Acta 2023; 541:117243. [PMID: 36740088 DOI: 10.1016/j.cca.2023.117243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Macrophage activation and hypercytokinemia are notable presentations in certain viral infections leading to severe disease and poor prognosis. Viral infections can cause macrophage polarization into the pro-inflammatory M1 or anti-inflammatory M2 phenotype. Activated M1 macrophages usually restrict viral replication whereas activated M2 macrophages suppress inflammation and promote tissue repair. In response to inflammatory stimuli, macrophages polarize to the M2 phenotype expressing hemoglobin scavenger CD163 surface receptor. The CD163 receptor is shed as the soluble form, sCD163, into plasma or tissue fluids. sCD163 causes detoxification of pro-oxidative hemoglobin which produces anti-inflammatory metabolites that promote the resolution of inflammation. Hence, increased CD163 expression in tissues and elevated circulatory levels of sCD163 have been associated with acute and chronic inflammatory diseases. CD163 and other macrophage activation markers have been commonly included in the investigation of disease pathogenesis and progression. This review provides an overview of the involvement of CD163 in viral diseases. The clinical utility of CD163 in viral disease diagnosis, progression, prognosis and treatment evaluation is discussed.
Collapse
Affiliation(s)
- Yi-Jing Yap
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Universiti Malaya, 50603 Kuala Lumpur, Malaysia; World Health Organization Collaborating Centre for Arbovirus Reference and Research (Dengue and Severe Dengue) MAA-12, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Sing-Sin Sam
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Anusha Shunmugarajoo
- Medical Department, Tengku Ampuan Rahimah Hospital, 41200 Klang, Selangor, Malaysia
| | - Yih-Harng Soh
- Centers for Disease Control and Prevention Unit, Central Melaka District Health Office, Jalan Bukit Baru, 75150 Melaka, Malaysia
| | - Suzana Misbah
- Biological Security and Sustainability Research Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Ahmad Kashfi Ab Rahman
- Department of Medicine (Infectious Disease Unit), Sultanah Nur Zahirah Hospital, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| |
Collapse
|
19
|
Peters BA, Hanna DB, Sharma A, Anastos K, Hoover DR, Shi Q, Moran CA, Jackson EA, Alcaide ML, Ofotokun I, Adimora AA, Haberlen SA, Cohen M, Tien PC, Michel KG, Levine SR, Hodis HN, Kaplan RC, Yin MT. Menopausal Hormone Therapy and Subclinical Cardiovascular Disease in Women With and Without Human Immunodeficiency Virus. Clin Infect Dis 2023; 76:e661-e670. [PMID: 35903868 PMCID: PMC10169435 DOI: 10.1093/cid/ciac620] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/20/2022] [Accepted: 07/26/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Estrogen-based hormone therapy (HT) may have beneficial cardiovascular effects when initiated in early menopause. This has not been examined in women with human immunodeficiency virus (HIV), who have heightened immune activation and cardiovascular risks. METHODS Among 609 postmenopausal women (1234 person-visits) in the Women's Interagency HIV Study, we examined the relationship of ever HT use (oral, patch, or vaginal) with subclinical atherosclerosis: carotid artery intima-media thickness (CIMT), distensibility, and plaque assessed via repeated B-mode ultrasound imaging (2004-2013). We also examined associations of HT with cross-sectional biomarkers of immune activation and D-dimer. Statistical models were adjusted for sociodemographic, behavioral, and cardiometabolic factors. RESULTS Women (mean age, 51 years; 80% HIV positive) who ever used HT at baseline were older, and more likely to be non-Hispanic White and report higher income, than never-users. Women who ever used HT had 43% lower prevalence of plaque (prevalence ratio, 0.57 [95% confidence interval {CI}, .40-.80]; P < .01), 2.51 µm less progression of CIMT per year (95% CI, -4.60, to -.41; P = .02), and marginally lower incidence of plaque over approximately 7 years (risk ratio, 0.38 [95% CI, .14-1.03; P = .06), compared with never-users, adjusting for covariates; ever HT use was not associated with distensibility. These findings were similar for women with and without HIV. Ever HT use was associated with lower serum D-dimer, but not with biomarkers of immune activation after covariate adjustment. CONCLUSIONS HT may confer a subclinical cardiovascular benefit in women with HIV. These results begin to fill a knowledge gap in menopausal care for women with HIV, in whom uptake of HT is very low.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathryn Anastos
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Donald R Hoover
- Department of Statistics and Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, New Jersey, USA
| | - Qiuhu Shi
- School of Health Sciences and Practice, New York Medical College, Valhalla, New York, USA
| | - Caitlin A Moran
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Elizabeth A Jackson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Maria L Alcaide
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Igho Ofotokun
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Adaora A Adimora
- Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sabina A Haberlen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mardge Cohen
- Department of Medicine, Stroger Hospital, Cook County Health and Hospital System, Chicago, Illinois, USA
| | - Phyllis C Tien
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Veterans Affairs Medical Center, San Francisco, California, USA
| | - Katherine G Michel
- Department of Medicine, Georgetown University, Washington, District of Columbia, USA
| | - Steven R Levine
- Departments of Neurology and Emergency Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Howard N Hodis
- Departments of Medicine and Population and Public Health Sciences, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, Washington, USA
| | - Michael T Yin
- Department of Medicine, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
20
|
The Gut Microbiome, Microbial Metabolites, and Cardiovascular Disease in People Living with HIV. Curr HIV/AIDS Rep 2023; 20:86-99. [PMID: 36708497 DOI: 10.1007/s11904-023-00648-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW To synthesize recent evidence relating the gut microbiome and microbial metabolites to cardiovascular disease (CVD) in people living with HIV (PLWH). RECENT FINDINGS A few cross-sectional studies have reported on the gut microbiome and cardiovascular outcomes in the context of HIV, with no consistent patterns emerging. The largest such study found that gut Fusobacterium was associated with carotid artery plaque. More studies have evaluated microbial metabolite trimethylamine N-oxide with CVD risk in PLWH, but results were inconsistent, with recent prospective analyses showing null effects. Studies of other microbial metabolites are scarce. Microbial translocation biomarkers (e.g., lipopolysaccharide binding protein) have been related to incident CVD in PLWH. Microbial translocation may increase CVD risk in PLWH, but there is insufficient and/or inconsistent evidence regarding specific microbial species and microbial metabolites associated with cardiovascular outcomes in PLWH. Further research is needed in large prospective studies integrating the gut microbiome, microbial translocation, and microbial metabolites with cardiovascular outcomes in PLWH.
Collapse
|
21
|
Ntsekhe M, Baker JV. Cardiovascular Disease Among Persons Living With HIV: New Insights Into Pathogenesis and Clinical Manifestations in a Global Context. Circulation 2023; 147:83-100. [PMID: 36576956 DOI: 10.1161/circulationaha.122.057443] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Widespread use of contemporary antiretroviral therapy globally has transformed HIV disease into a chronic illness associated with excess risk for disorders of the heart and circulatory system. Current clinical care and research has focused on improving HIV-related cardiovascular disease outcomes, survival, and quality of life. In high-income countries, emphasis on prevention of atherosclerotic coronary artery disease over the past decade, including aggressive management of traditional risk factors and earlier initiation of antiretroviral therapy, has reduced risk for myocardial infarction among persons living with human immunodeficiency virus-1 infection. Still, across the globe, persons living with human immunodeficiency virus-1 infection on effective antiretroviral therapy treatment remain at increased risk for ischemic outcomes such as myocardial infarction and stroke relative to the persons without HIV. Unique features of HIV-related cardiovascular disease, in part, include the pathogenesis of coronary disease characterized by remodeling ectasia and unusual plaque morphology, the relative high proportion of type 2 myocardial infarction events, abnormalities of the aorta such as aneurysms and diffuse aortic inflammation, and HIV cerebrovasculopathy as a contributor to stroke risk. Literature over the past decade has also reflected a shift in the profile and prevalence of HIV-associated heart failure, with a reduced but persistent risk of heart failure with reduced ejection fraction and a growing risk of heart failure with preserved ejection fraction. Cardiac magnetic resonance imaging and autopsy data have emphasized the central importance of intramyocardial fibrosis for the pathogenesis of both heart failure with preserved ejection fraction and the increase in risk of sudden cardiac death. Still, more research is needed to better characterize the underlying mechanisms and clinical phenotype of HIV-associated myocardial disease in the current era. Across the different cardiovascular disease manifestations, a common pathogenic feature is that HIV-associated inflammation working through different mechanisms may amplify underlying pathology because of traditional risk and other host factors. The prevalence and phenotype of individual cardiovascular disease manifestations is ultimately influenced by the degree of injury from HIV disease combined with the profile of underlying cardiometabolic factors, both of which may differ substantially by region globally.
Collapse
Affiliation(s)
- Mpiko Ntsekhe
- Division of Cardiology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa (M.N.)
| | - Jason V Baker
- Division of Infectious Diseases, Hennepin Healthcare Research Institute, Minneapolis, MN (J.V.B.).,Department of Medicine, University of Minnesota, Minneapolis (J.V.B.)
| |
Collapse
|
22
|
Boczar KE, Faller E, Zeng W, Wang J, Small GR, Corrales-Medina VF, deKemp RA, Ward NC, Beanlands RSB, MacPherson P, Dwivedi G. Anti-inflammatory effect of rosuvastatin in patients with HIV infection: An FDG-PET pilot study. J Nucl Cardiol 2022; 29:3057-3068. [PMID: 34820771 DOI: 10.1007/s12350-021-02830-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/22/2021] [Indexed: 01/29/2023]
Abstract
AIMS This study aimed to evaluate markers of systemic as well as imaging markers of inflammation in the ascending aorta, bone marrow, and spleen measured by 18F-FDG PET/CT, in HIV+ patients at baseline and following therapy with rosuvastatin. METHODS AND RESULTS Of the 35 HIV+ patients enrolled, 17 were randomized to treatment with 10 mg/day rosuvastatin and 18 to usual care for 6 months. An HIV- control cohort was selected for baseline comparison of serum inflammatory markers and monocyte markers of inflammation. 18F-FDG-PET/CT imaging of bone marrow, spleen, and thoracic aorta was performed in the HIV+ cohort at baseline and 6 months. While CD14++CD16- and CCR2 expressions were reduced, serum levels of IL-7, IL-8, and MCP-1 were elevated in the HIV+ population compared to the controls. There was a significant drop in FDG uptake in the bone marrow (TBRmax), spleen (SUVmax) and thoracic aortic (TBRmax) in the statin-treated group compared to the control group (bone marrow: - 10.3 ± 16.9% versus 5.0 ± 18.9%, p = .0262; spleen: - 9.8 ± 20.3% versus 11.3 ± 28.8%, p = .0497; thoracic aorta: - 19.1 ± 24.2% versus 4.3 ± 15.4%, p = .003). CONCLUSIONS HIV+ patients had significantly markers of systemic inflammation including monocyte activation. Treatment with low-dose rosuvastatin in the HIV+ cohort significantly reduced bone marrow, spleen and thoracic aortic FDG uptake.
Collapse
Affiliation(s)
- Kevin E Boczar
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Elliot Faller
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wanzhen Zeng
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jerry Wang
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Gary R Small
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Vicente F Corrales-Medina
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Robert A deKemp
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Natalie C Ward
- School of Public Health, Curtin University, Perth, Australia
- School of Medicine, University of Western Australia, Perth, Australia
| | - Rob S B Beanlands
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Paul MacPherson
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Girish Dwivedi
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada.
- School of Medicine, University of Western Australia, Perth, Australia.
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, The University of Western Australia, Murdoch, Australia.
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, WA, 6009, Australia.
- School of Biomedical Sciences at Curtin University, Perth, WA, Australia.
| |
Collapse
|
23
|
Peterson TE, Landon C, Haberlen SA, Bhondoekhan F, Plankey MW, Palella FJ, Piggott DA, Margolick JB, Brown TT, Post WS, Wu KC. Circulating biomarker correlates of left atrial size and myocardial extracellular volume fraction among persons living with and without HIV. BMC Cardiovasc Disord 2022; 22:393. [PMID: 36057773 PMCID: PMC9441072 DOI: 10.1186/s12872-022-02835-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/26/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Infection with human immunodeficiency virus (HIV) is associated with higher risk for myocardial disease despite modern combination antiretroviral therapy (cART). Factors contributing to this excess risk, however, remain poorly characterized. We aimed to assess cross-sectional relationships between elevations of left atrial volume index (LAVI) and myocardial extracellular volume (ECV) fraction that have been reported in persons living with HIV and levels of circulating biomarkers of inflammation, fibrosis, and myocyte stretch among persons living with and without HIV (PLWH, PLWOH). METHODS Participants from three cohorts of PLWH and PLWOH underwent cardiovascular magnetic resonance imaging for measurement of LAVI and ECV. Levels of circulating proteins (IL-6, sCD14, galectin-3, NT-proBNP, GDF-15, TIMP-2, MMP-2, and hsTnI) were measured using immunoassays. Associations were assessed using logistic and linear regression, adjusting for demographics, substance use, and clinical characteristics. RESULTS Among 381 participants with and without HIV, median age (IQR) was 55.1 (51.2, 58.4) years, 28% were female, 69% were Black, and 46% were current smokers. Sixty-two percent were PLWH (n = 235), of whom 88% were receiving cART and 72% were virally suppressed. PLWH had higher levels of sCD14 (p = < 0.001), GDF-15 (p = < 0.001), and NT-proBNP (p = 0.03) compared to PLWOH, while levels of other biomarkers did not differ by HIV serostatus, including IL-6 (p = 0.84). Among PLWH, higher sCD14, GDF-15, and NT-proBNP were also associated with lower CD4 + cell count, and higher NT-proBNP was associated with detectable HIV viral load. NT-proBNP was associated with elevated LAVI (OR: 1.79 [95% CI: 1.31, 2.44]; p < 0.001) with no evidence of effect measure modification by HIV serostatus. Other associations between HIV-associated biomarkers and LAVI or ECV were small or imprecise. CONCLUSIONS Our findings suggest that elevated levels of sCD14, GDF-15, and NT-proBNP among PLWH compared to PLWOH observed in the current cART era may only minimally reflect HIV-associated elevations in LAVI and ECV. Future studies of excess risk of myocardial disease among contemporary cohorts of PLWH should investigate mechanisms other than those connoted by the studied biomarkers.
Collapse
Affiliation(s)
- Tess E. Peterson
- grid.21107.350000 0001 2171 9311Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Christian Landon
- grid.21107.350000 0001 2171 9311Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Sabina A. Haberlen
- grid.21107.350000 0001 2171 9311Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Fiona Bhondoekhan
- grid.40263.330000 0004 1936 9094Department of Epidemiology, Brown University School of Public Health, Providence, RI USA
| | - Michael W. Plankey
- grid.411667.30000 0001 2186 0438Department of Medicine, Georgetown University Medical Center, Washington, DC USA
| | - Frank J. Palella
- grid.16753.360000 0001 2299 3507Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Damani A. Piggott
- grid.21107.350000 0001 2171 9311Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Joseph B. Margolick
- grid.21107.350000 0001 2171 9311Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Todd T. Brown
- grid.21107.350000 0001 2171 9311Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Wendy S. Post
- grid.21107.350000 0001 2171 9311Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Katherine C. Wu
- grid.21107.350000 0001 2171 9311Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
24
|
Abstract
OBJECTIVES People with HIV (PWH) are at increased risk for premature cardiovascular disease (CVD). Clonal hematopoiesis is a common age-related condition that may be associated with increased CVD risk. The goal of this study was to determine the prevalence of clonal hematopoiesis and its association with chronic inflammation and CVD in PWH. DESIGN Cross-sectional study utilizing archived specimens and data from 118 men (86 PWH and 32 HIV-uninfected) from the Baltimore-Washington DC center of the Multicenter AIDS Cohort Study (MACS) who had had coronary computed tomography angiography (CTA) and measurement of 34 serologic inflammatory biomarkers. METHODS Clonal hematopoiesis was assessed on peripheral blood mononuclear cells utilizing targeted error-corrected next generation sequencing (NGS) focused on 92 genes frequently mutated in hematologic malignancies. Clinical and laboratory data were obtained from the MACS database. RESULTS Clonal hematopoiesis with a variant allele frequency (VAF) greater than 1% was significantly more common in PWH [20/86 (23.3%)] than in HIV-uninfected men [2/32 (6.3%)] ( P = 0.035). PWH with clonal hematopoiesis (VAF > 1%) were more likely to have coronary artery stenosis of at least 50% than those without clonal hematopoiesis [6/20 (30%) vs. 6/64 (9%); P = 0.021]. Presence of clonal hematopoiesis was not significantly associated with serological inflammatory markers, except for significantly lower serum leptin levels; this was not significant after adjustment for abdominal or thigh subcutaneous fat area. CONCLUSION Clonal hematopoiesis was more common in PWH and among PWH was associated with the extent of coronary artery disease. Larger studies are needed to further examine the biological and clinical consequences of clonal hematopoiesis in PWH.
Collapse
|
25
|
Kentoffio K, Temu TM, Shakil SS, Zanni MV, Longenecker CT. Cardiovascular disease risk in women living with HIV. Curr Opin HIV AIDS 2022; 17:270-278. [PMID: 35938460 PMCID: PMC9370828 DOI: 10.1097/coh.0000000000000756] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To synthesize current evidence on the impact of cardiovascular disease among women living with HIV (WLWH) with a particular focus on disease prevalence, mechanisms and prevention. RECENT FINDINGS HIV-related cardiovascular disease risk is 1.5-fold to 2-fold higher for women than for men. Mechanisms of enhanced risk are multifactorial and include reinforcing pathways between traditional risk factors, metabolic dysregulation, early reproductive aging and chronic immune activation. These pathways influence both the presentation of overt syndromes of myocardial infarction, stroke and heart failure, as well as subclinical disease, such as microvascular dysfunction and cardiac fibrosis. Cardiovascular disease, therefore, remains a consistent threat to healthy aging among WLWH. SUMMARY Although no specific prevention strategies exist, patient-centered risk mitigation approaches that are adaptable to the needs of aging individuals are essential to combat disparities in cardiovascular outcomes among WLWH. Further research into the optimal prevention approach for CVD among WLWH, particularly for women living in under-resourced health systems, is needed.
Collapse
Affiliation(s)
- Katherine Kentoffio
- Department of Medicine, Division of Cardiology, University of California, San Francisco, CA, USA
| | - Tecla M Temu
- Department of Global Health, University of Washington School of Medicine, Seattle, WA, USA
| | - Saate S Shakil
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Markella V. Zanni
- Department of Medicine, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
| | - Chris T. Longenecker
- Department of Global Health, University of Washington School of Medicine, Seattle, WA, USA
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Fitch KV, Fulda ES, Grinspoon SK. Statins for primary cardiovascular disease prevention among people with HIV: emergent directions. Curr Opin HIV AIDS 2022; 17:293-300. [PMID: 35938463 PMCID: PMC9415230 DOI: 10.1097/coh.0000000000000752] [Citation(s) in RCA: 3] [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] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW While people with HIV (PWH) are living longer due to advances in antiretroviral therapy, recent data have demonstrated an increased risk of cardiovascular disease (CVD) among this population. This increased risk is thought to be due to both traditional (for example, smoking, diabetes) and HIV-specific (for example, inflammation, persistent immune activation) risk factors. This review focuses on the potential for statin therapy to mitigate this increased risk. RECENT FINDINGS Several randomized clinical trials have demonstrated that statins, a class of lipid-lowering medications, are effective as a primary CVD prevention strategy among people without HIV. Among PWH, statins have been shown to lower cholesterol, exert immunomodulatory effects, stabilize coronary atherosclerotic plaque, and even induce plaque regression. SUMMARY Prevention of CVD among the aging population of people with controlled, but chronic, HIV is vital. Data exploring primary prevention in this context are thus far limited. The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) is ongoing; this trial will inform the field by investigating the effects of pitavastatin calcium as a primary prevention strategy for major adverse cardiovascular events among PWH on antiretroviral therapy (ART) at low-to-moderate traditional CVD risk.
Collapse
Affiliation(s)
- Kathleen V Fitch
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
27
|
Wang Z, Peters BA, Usyk M, Xing J, Hanna DB, Wang T, Post WS, Landay AL, Hodis HN, Weber K, French A, Golub ET, Lazar J, Gustafson D, Kassaye S, Aouizerat B, Haberlen S, Malvestutto C, Budoff M, Wolinsky SM, Sharma A, Anastos K, Clish CB, Kaplan RC, Burk RD, Qi Q. Gut Microbiota, Plasma Metabolomic Profiles, and Carotid Artery Atherosclerosis in HIV Infection. Arterioscler Thromb Vasc Biol 2022; 42:1081-1093. [PMID: 35678187 PMCID: PMC9339474 DOI: 10.1161/atvbaha.121.317276] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Alterations in gut microbiota and blood metabolomic profiles have been implicated in HIV infection and cardiovascular disease. However, it remains unclear whether alterations in gut microbiota may contribute to disrupted host blood metabolomic profiles in relation to atherosclerosis, especially in the context of HIV infection. METHODS We analyzed cross-sectional associations between gut microbiota features and carotid artery plaque in 361 women with or at high risk of HIV (67% HIV+), and further integrated plaque-associated microbial features with plasma lipidomic/metabolomic profiles. Furthermore, in 737 women and men, we examined prospective associations of baseline gut bacteria-associated lipidomic and metabolomic profiles with incident carotid artery plaque over 7-year follow-up. RESULTS We found 2 potentially pathogenic bacteria, Fusobacterium and Proteus, were associated with carotid artery plaque; while the beneficial butyrate producer Odoribacter was inversely associated with plaque. Fusobacterium and Proteus were associated with multiple lipids/metabolites which were clustered into 8 modules in network. A module comprised of 9 lysophosphatidylcholines and lysophosphatidylethanolamines and a module comprised of 9 diglycerides were associated with increased risk of carotid artery plaque (risk ratio [95% CI], 1.34 [1.09-1.64] and 1.24 [1.02-1.51] per SD increment, respectively). Functional analyses identified bacterial enzymes in lipid metabolism associated with these plasma lipids. In particular, phospholipase A1 and A2 are the key enzymes in the reactions producing lysophosphatidylcholines and lysophosphatidylethanolamines. CONCLUSIONS Among individuals with or at high risk of HIV infection, we identified altered gut microbiota and related functional capacities in the lipid metabolism associated with disrupted plasma lipidomic profiles and carotid artery atherosclerosis.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York
| | - Brandilyn A Peters
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York
| | - Mykhaylo Usyk
- Department of Pediatrics (M.U., R.D.B.), Albert Einstein College of Medicine, Bronx, New York
| | - Jiaqian Xing
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York
| | - David B Hanna
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York
| | - Wendy S Post
- Department of Medicine, Johns Hopkins University, Baltimore, MD (W.S.P.)
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL (A.L.L)
| | - Howard N Hodis
- Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles (H.N.H.)
| | | | - Audrey French
- Department of Internal Medicine, John H. Stroger Jr Hospital of Cook County, Chicago, IL (A.F.)
| | - Elizabeth T Golub
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (E.T.G., S.H.)
| | - Jason Lazar
- Department of Medicine (J.L.), State University of New York Downstate Medical Center, Brooklyn
| | - Deborah Gustafson
- Department of Neurology (D.G.), State University of New York Downstate Medical Center, Brooklyn
| | - Seble Kassaye
- Department of Medicine, Georgetown University, Washington DC (S.K.)
| | | | - Sabina Haberlen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (E.T.G., S.H.)
| | | | - Matthew Budoff
- David Geffen School of Medicine, University of California, Los Angeles (M.B.)
| | - Steven M Wolinsky
- Department of Medicine, Feinberg School of Medicine, Northwestern University Chicago, IL (S.M.W.)
| | - Anjali Sharma
- Department of Medicine (A.S., K.A.), Albert Einstein College of Medicine, Bronx, New York
| | - Kathryn Anastos
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York.,Department of Medicine (A.S., K.A.), Albert Einstein College of Medicine, Bronx, New York
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA (C.B.C.)
| | - Robert C Kaplan
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA (R.C.K.)
| | - Robert D Burk
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York.,Department of Pediatrics (M.U., R.D.B.), Albert Einstein College of Medicine, Bronx, New York.,Department of Microbiology & Immunology (R.D.B.), Albert Einstein College of Medicine, Bronx, New York
| | - Qibin Qi
- Department of Epidemiology and Population Health (Z.W., B.A.P., J.X., D.B.H., T.W., K.A., R.C.K., R.D.B., Q.Q.), Albert Einstein College of Medicine, Bronx, New York.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (Q.Q.)
| |
Collapse
|
28
|
Vanpouille C, Wells A, Wilkin T, Mathad JS, Morris S, Margolis L, Gianella S. Sex differences in cytokine profiles during suppressive antiretroviral therapy. AIDS 2022; 36:1215-1222. [PMID: 35608113 PMCID: PMC9283283 DOI: 10.1097/qad.0000000000003265] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Despite lower plasma HIV RNA levels, women progress faster to AIDS than men. The reasons for these differences are not clear but might be a consequence of an elevated inflammatory response in women. METHODS We investigated sex differences in cytokine profiles by measuring the concentrations of 36 cytokines/chemokines by Luminex in blood of women and men (sex at birth) with chronic HIV infection under suppressive therapy. We initially performed a principal component analysis to see if participants clustered by sex, and then fit a partial least squares discriminant analysis (PLS-DA) model where we used cytokines to predict sex at birth. The significance of the difference in nine cytokines with VIP greater than 1 was tested using Wilcoxon test-rank. Further, potential confounding factors were tested by multivariate linear regression models. RESULTS Overall, we predicted sex at birth in the PLS-DA model with an error rate of approximately 13%. We identified five cytokines, which were significantly higher in women compared with men, namely the pro-inflammatory chemokines CXCL1 (Gro-α), CCL5 (RANTES), CCL3 (MIP-1α), CCL4 (MIP-1β), as well as the T-cell homeostatic factor IL-7. The effect of sex remained significant after adjusting for CD4 + , age, ethnicity, and race for all cytokines, except for CCL3 and race. CONCLUSION The observed sex-based differences in cytokines might contribute to higher immune activation in women compared with men despite suppressive therapy. Increased levels of IL-7 in women suggest that homeostatic proliferation may have a differential contribution to HIV reservoir maintenance in female and male individuals. Our study emphasizes the importance of sex-specific studies of viral pathogenesis.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Alan Wells
- Department of Medicine, University of California San Diego, La Jolla, CA
| | | | | | - Sheldon Morris
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Leonid Margolis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Sara Gianella
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
29
|
Kettelhut A, Bowman E, Gabriel J, Hand B, Liyanage NPM, Kulkarni M, Avila-Soto F, Lake JE, Funderburg NT. Estrogen May Enhance Toll-Like Receptor 4-Induced Inflammatory Pathways in People With HIV: Implications for Transgender Women on Hormone Therapy. Front Immunol 2022; 13:879600. [PMID: 35720418 PMCID: PMC9205606 DOI: 10.3389/fimmu.2022.879600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Background Transgender women (TW) are at increased risk for both human immunodeficiency virus (HIV) and cardiovascular disease (CVD). Antiretroviral therapy-treated HIV has been associated with a two-fold increased risk of CVD, potentially due to dysregulated Toll-like receptor (TLR)-induced immune activation. Use of estrogens in feminizing hormone therapy (FHT) may enhance inflammatory responses and the risk of cardiovascular mortality in TW. Despite this, the immunomodulatory effects of estrogen use in TW with HIV have been inadequately explored. Methods As an in vitro model for FHT, cryopreserved PBMCs (cryoPBMCs) from HIV negative (HIV-), HIV+ ART-suppressed (HIV+SP), and HIV+ ART-unsuppressed (HIV+USP) cisgender men were cultured overnight in the presence of 17-β estradiol or 17-α ethinylestradiol with and without the TLR4 agonist LPS or the TLR8 agonist ssPolyU. Monocyte activation (CD69, HLA-DR, CD38) was assessed by flow cytometry. Cytokine levels (IL-6, TNF-α, IL-1β, and IL-10) were measured in cell culture supernatants by Legendplex. Levels of phosphorylated TLR signaling molecules (JNK, MAPK p38) were assessed by Phosflow. Plasma levels of immune activation biomarkers (LPS-binding protein, monocyte activation markers sCD14 and sCD163, and inflammatory molecules IL-6 and TNF-α receptor I) were measured by ELISA. Results PBMCs from people with HIV (PWH) produced greater levels of inflammatory cytokines following exposure to LPS or ssPolyU compared to levels from cells of HIV- individuals. While estrogen exposure alone induced mild changes in immune activation, LPS-induced TLR4 activation was elevated with estrogen in cisgender men (CM) with HIV, increasing monocyte activation and inflammatory cytokine production (IL-6, TNF-α). Interestingly, testosterone inhibited LPS-induced cytokine production in CM regardless of HIV status. Plasma markers of immune activation and microbial translocation (e.g., sCD14, sCD163, LPS-binding protein) were generally higher in PWH compared to HIV- CM, and these markers were positively associated with in vitro responsiveness to estrogen and LPS in CM with HIV. Conclusions Our in vitro data suggest that estrogen exposure may enhance innate immune activation in PWH. Further examination is needed to fully understand the complex interactions of FHT, HIV, and CVD in TW, and determine optimal FHT regimens or supplementary treatments aimed at reducing excess immune activation.
Collapse
Affiliation(s)
- Aaren Kettelhut
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Emily Bowman
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Janelle Gabriel
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Brittany Hand
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Namal P. M. Liyanage
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Manjusha Kulkarni
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Frances Avila-Soto
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| | - Jordan E. Lake
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nicholas T. Funderburg
- Department of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
30
|
Knudsen AD, Bouazzi R, Afzal S, Gelpi M, Benfield T, Høgh J, Thomsen MT, Trøseid M, Nordestgaard BG, Nielsen SD. Monocyte count and soluble markers of monocyte activation in people living with HIV and uninfected controls. BMC Infect Dis 2022; 22:451. [PMID: 35546661 PMCID: PMC9097376 DOI: 10.1186/s12879-022-07450-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 05/04/2022] [Indexed: 11/10/2022] Open
Abstract
Background Monocytes play an important role in inflammation, and monocytosis and monocyte activation are features of chronic inflammation. We aimed to investigate if HIV status was associated with monocyte count and monocyte activation and to assess the relationship between monocyte count and monocyte activation markers and HIV-related factors. Methods Persons living with HIV (PLWH) with measured monocyte count and sCD14 and sCD163 were included from the Copenhagen Comorbidity in HIV infection (COCOMO) study and matched 1:5 on sex and age with uninfected controls. In addition, 74 uninfected individuals from COCOMO with measured sCD14 and sCD163 were included. Identical protocols and equipment were used to determine monocyte counts and monocyte activation in PLWH and uninfected controls. Linear regression adjusted for age, sex, smoking and waist-to-hip-ratio was used to analyze the association between possible risk factors and monocyte outcomes. Results We included 871 PLWH and 4355 uninfected controls. PLWH had − 0.021 [− 0.031 − 0.011] × 109/L) lower monocyte count than uninfected controls, and in adjusted analyses HIV status was independently associated with − 0.035 [− 0.045, − 0.025] × 109/L lower monocyte count. In contrast, PLWH had higher sCD163 and sCD14 concentrations than uninfected controls. After adjustment, HIV-status was associated with higher sCD14 and sCD163 concentrations (588 [325, 851] ng/ml, and 194 [57, 330] ng/ml, respectively). Conclusion PLWH had lower monocyte counts than controls, but the absolute difference was small, and any clinical impact is likely limited. In contrast, concentrations of monocyte activation markers, previously implicated as drivers of non-AIDS comorbidity, were higher in PLWH than in controls. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07450-y.
Collapse
Affiliation(s)
- Andreas D Knudsen
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark.,Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Randa Bouazzi
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark
| | - Shoaib Afzal
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Gelpi
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Julie Høgh
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark
| | - Magda Teresa Thomsen
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark
| | - Marius Trøseid
- Section of Clinical Immunology and Infectious Disease, Oslo University Hospital, Oslo, Norway
| | - Børge G Nordestgaard
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Susanne D Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Blegdamsvej 9B, DK-2100, Copenhagen Ø, Denmark.
| |
Collapse
|
31
|
Martinez CA, Rikhi R, Pester MS, Parker M, Gonzalez A, Larson M, Chavez J, Mendez A, Raines JK, Kolber MA, Schulman IH, Alcaide ML, Hurwitz BE. Abacavir antiretroviral therapy and indices of subclinical vascular disease in persons with HIV. PLoS One 2022; 17:e0264445. [PMID: 35271614 PMCID: PMC8912137 DOI: 10.1371/journal.pone.0264445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 02/10/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Indices of cardiovascular disease (CVD) risk, vascular endothelial dilation, arterial stiffness and endothelial repair were examined in persons with HIV (PWH) on an antiretroviral therapy (ART) that included abacavir (ABC+) in comparison with PWH on ART without abacavir (ABC-), and with HIV seronegative (HIV-) individuals. Approach The 115 participants (63% men), aged 30–50 years, did not have CVD, metabolic, endocrine, or chronic renal conditions. PWH were on stable ART for six-months or more. Vascular assessments included flow-mediated dilation (FMD), aortic, radial and femoral arterial stiffness (cAIx, crPWV, cfPWV), and thigh and calf arterial compliance (Vmax50). Endothelial repair was indexed by endothelial progenitor cell colony forming units (EPC-CFU). Traditional CVD risk measures included blood pressure, central adiposity, lipids, insulin resistance (HOMA-IR), CRP and ASCVD score. Analyses controlled for demographics (age, sex, education), medications (antihypertensive, statin/fibrate, antipsychotic), and substance abuse (ASSIST). Results No group differences were observed in central adiposity, HOMA-IR, CRP, or ASCVD risk score. However, the ABC- group displayed greater dyslipidemia. The ABC+ group displayed no difference on FMD, cAIx, cfPWV or calf Vmax50 compared with other groups. When CD4 count and viral load were controlled, no additional differences between the ABC+ and ABC- groups emerged. Analyses of crPWV and thigh Vmax50 suggested supported by a trend toward lower EPC-CFU in the HIV+ groups than the HIV- group. Conclusions Findings indicate that ABC treatment of 30–50 year-old PWH on stable ART is not likely to contribute in a robust way to higher CVD risk.
Collapse
Affiliation(s)
- Claudia A. Martinez
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| | - Rishi Rikhi
- Division of Cardiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Mollie S. Pester
- Behavioral Medicine Research Center, University of Miami, Miami, Florida, United States of America
- Department of Psychology, University of Miami, Coral Gables, Florida, United States of America
| | - Meela Parker
- Behavioral Medicine Research Center, University of Miami, Miami, Florida, United States of America
| | - Alex Gonzalez
- Behavioral Medicine Research Center, University of Miami, Miami, Florida, United States of America
| | - Michaela Larson
- Department of Public Health Science, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jennifer Chavez
- Department of Public Health Science, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Armando Mendez
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Jeffrey K. Raines
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Michael A. Kolber
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ivonne H. Schulman
- Katz Family Division of Nephrology & Hypertension, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Maria L. Alcaide
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Barry E. Hurwitz
- Behavioral Medicine Research Center, University of Miami, Miami, Florida, United States of America
- Department of Psychology, University of Miami, Coral Gables, Florida, United States of America
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
32
|
Hughey CM, Vuong BW, Ribaudo HB, Mitchell CCK, Korcarz CE, Hodis HN, Currier JS, Stein JH. Grayscale Ultrasound Texture Features of Carotid and Brachial Arteries in People With HIV Infection Before and After Antiretroviral Therapy. J Am Heart Assoc 2022; 11:e024142. [PMID: 35179037 PMCID: PMC9075086 DOI: 10.1161/jaha.121.024142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background We aimed to investigate novel grayscale ultrasound characteristics of the carotid and brachial arteries in people with HIV infection before and after starting initial antiretroviral therapy (ART). Methods and Results We performed grayscale ultrasound image analyses of the common carotid artery (CCA) and brachial artery before and after receipt of 1 of 3 randomly allocated ART regimens. We measured arterial wall echogenicity (grayscale median), contrast (gray-level difference statistic method), and entropy. These measures and their changes were compared with atherosclerotic cardiovascular disease risk factors, measures of HIV disease severity, and inflammatory biomarkers before and after ART. Changes in the grayscale measures were evaluated within and between ART arms. Among 201 ART-naïve people with HIV, higher systolic blood pressure, higher body mass index, lower CD4+ T cells, and non-Hispanic White race and ethnicity were associated independently with lower CCA grayscale median. Changes in each CCA grayscale measure from baseline to 144 weeks correlated with changes in soluble CD163: grayscale median (ρ=-0.17; P=0.044), gray-level difference statistic-contrast (ρ=-0.19; P=0.024), and entropy (ρ=-0.21; P=0.016). Within the atazanavir/ritonavir arm, CCA entropy increased (adjusted β=0.023 [95% CI, 0.001-0.045]; P=0.04), but no other within-arm changes in grayscale measures were seen. Correlations of brachial artery grayscale measures were weaker. Conclusions In ART-naïve people with HIV, CCA grayscale ultrasound measures were associated with atherosclerotic cardiovascular disease risk factors and lower grayscale median was associated with lower CD4+ T cells. Reductions in soluble CD163 with initial ART were associated with improvements in all 3 CCA grayscale measures, suggesting that reductions in macrophage activation with ART initiation may lead to less arterial injury. Registration URL: https://clinicaltrials.gov/; Unique identifiers: NCT00811954; NCT00851799.
Collapse
Affiliation(s)
| | - Belinda W Vuong
- University of Wisconsin School of Medicine and Public Health Madison WI
| | | | | | - Claudia E Korcarz
- University of Wisconsin School of Medicine and Public Health Madison WI
| | - Howard N Hodis
- Keck School of Medicine of University of Southern California Los Angeles CA
| | - Judith S Currier
- David Geffen School of Medicine at University of California-Los Angeles Los Angeles CA
| | - James H Stein
- University of Wisconsin School of Medicine and Public Health Madison WI
| |
Collapse
|
33
|
Moran CA, Collins LF, Beydoun N, Mehta PK, Fatade Y, Isiadinso I, Lewis TT, Weber B, Goldstein J, Ofotokun I, Quyyumi A, Choi MY, Titanji K, Lahiri CD. Cardiovascular Implications of Immune Disorders in Women. Circ Res 2022; 130:593-610. [PMID: 35175848 PMCID: PMC8869407 DOI: 10.1161/circresaha.121.319877] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune responses differ between men and women, with women at higher risk of developing chronic autoimmune diseases and having more robust immune responses to many viruses, including HIV and hepatitis C virus. Although immune dysregulation plays a prominent role in chronic systemic inflammation, a key driver in the development of atherosclerotic cardiovascular disease (ASCVD), standard ASCVD risk prediction scores underestimate risk in populations with immune disorders, particularly women. This review focuses on the ASCVD implications of immune dysregulation due to disorders with varying global prevalence by sex: autoimmune disorders (female predominant), HIV (male-female equivalent), and hepatitis C virus (male predominant). Factors contributing to ASCVD in women with immune disorders, including traditional risk factors, dysregulated innate and adaptive immunity, sex hormones, and treatment modalities, are discussed. Finally, the need to develop new ASCVD risk stratification tools that incorporate variables specific to populations with chronic immune disorders, particularly in women, is emphasized.
Collapse
Affiliation(s)
- Caitlin A. Moran
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Lauren F. Collins
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Nour Beydoun
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Puja K. Mehta
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Yetunde Fatade
- Emory University School of Medicine, Department of Medicine, Atlanta, GA, USA
| | - Ijeoma Isiadinso
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Tené T Lewis
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, GA, USA
| | - Brittany Weber
- Harvard Medical School, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jill Goldstein
- Massachusetts General Hospital, Department of Psychiatry, and Harvard Medical School, Departments of Psychiatry and Medicine, Boston, MA, USA
| | - Igho Ofotokun
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Arshed Quyyumi
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - May Y. Choi
- Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Kehmia Titanji
- Emory University, Department of Medicine, Division of Endocrinology, Metabolism, and Lipids, Atlanta, GA, USA
| | - Cecile D. Lahiri
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| |
Collapse
|
34
|
McGettrick P, Mallon PWG. Biomarkers to predict cardiovascular disease in people living with HIV. Curr Opin Infect Dis 2022; 35:15-20. [PMID: 34873078 DOI: 10.1097/qco.0000000000000802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Cardiovascular diseases (CVD) is one of the leading cause of morbidity and mortality in antiretroviral treated people living with HIV (PWH) with risk score algorithms based on traditional risk factors being shown to be consistently unreliable in estimating risk in this population. This review aims to examine recent data published in last 18-24 months exploring biomarkers that may be useful in identifying PWH at risk of developing CVD. RECENT FINDINGS Ongoing research explores the association of inflammatory biomarkers with subclinical CVD with few studies examining their clinical utility in improving CVD risk prediction. Further mechanistic studies explore the role of monocyte/macrophages in CVD pathogenesis with some studies examining functional assays as better predictors of CVD risk. SUMMARY Although persistent associations with inflammatory markers and CVD are demonstrated, few biomarkers have emerged as being clinically useful. Large population studies are needed to assess their utility in improving CVD risk prediction in PWH.
Collapse
Affiliation(s)
- Padraig McGettrick
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin
| | - Patrick W G Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin
- Department of Infectious Diseases, St Vincents University Hospital, Dublin, Ireland
| |
Collapse
|
35
|
Peters BA, Xue X, Sheira LA, Qi Q, Sharma A, Santoro N, Alcaide ML, Ofotokun I, Adimora AA, McKay HS, Tien PC, Michel KG, Gustafson D, Turan B, Landay AL, Kaplan RC, Weiser SD. Menopause Is Associated With Immune Activation in Women With HIV. J Infect Dis 2022; 225:295-305. [PMID: 34174074 PMCID: PMC8763955 DOI: 10.1093/infdis/jiab341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/24/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Persistent immune activation due to gut barrier dysfunction is a suspected cause of morbidity in HIV, but the impact of menopause on this pathway is unknown. METHODS In 350 women with HIV from the Women's Interagency HIV Study, plasma biomarkers of gut barrier dysfunction (intestinal fatty acid binding protein; IFAB), innate immune activation (soluble CD14 and CD163; sCD14, sCD163), and systemic inflammation (interleukin-6 and tumor necrosis factor receptor 1; IL-6, TNFR1) were measured at 674 person-visits spanning ≤2 years. RESULTS Menopause (post- vs premenopausal status) was associated with higher plasma sCD14 and sCD163 in linear mixed-effects regression adjusting for age and other covariates (β = 161.89 ng/mL; 95% confidence interval [CI], 18.37-305.41 and 65.48 ng/mL, 95% CI, 6.64-124.33, respectively); but not with plasma IFAB, IL-6, or TNFR1. In piece-wise linear mixed-effects regression of biomarkers on years before/after the final menstrual period, sCD14 increased during the menopausal transition by 250.71 ng/mL per year (95% CI, 16.63-484.79; P = .04), but not in premenopausal or postmenopausal periods. CONCLUSIONS In women with HIV, menopause may increase innate immune activation, but data did not support an influence on the gut barrier or inflammation. Clinical implications of immune activation during menopausal transition warrant further investigation.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lila A Sheira
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco, San Francisco, California, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Maria L Alcaide
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Igho Ofotokun
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Adaora A Adimora
- School of Medicine and Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Heather S McKay
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Phyllis C Tien
- Department of Veterans Affairs Medical Center, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Katherine G Michel
- Department of Medicine, Georgetown University Medical Center, Washington, District of Colombia, USA
| | - Deborah Gustafson
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Bulent Turan
- Department of Psychology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sheri D Weiser
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
36
|
El-Far M, Hanna DB, Durand M, Larouche-Anctil E, Sylla M, Chartrand-Lefebvre C, Cloutier G, Goulet JP, Kassaye S, Karim R, Kizer JR, French AL, Gange SJ, Lazar JM, Hodis HN, Routy JP, Ancuta P, Chomont N, Landay AL, Kaplan RC, Tremblay CL. Brief Report: Subclinical Carotid Artery Atherosclerosis Is Associated With Increased Expression of Peripheral Blood IL-32 Isoforms Among Women Living With HIV. J Acquir Immune Defic Syndr 2021; 88:186-191. [PMID: 34138771 PMCID: PMC8434945 DOI: 10.1097/qai.0000000000002746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Persistent inflammation in HIV infection is associated with elevated cardiovascular disease (CVD) risk, even with viral suppression. Identification of novel surrogate biomarkers can enhance CVD risk stratification and suggest novel therapies. We investigated the potential of interleukin 32 (IL-32), a proinflammatory multi-isoform cytokine, as a biomarker for subclinical carotid artery atherosclerosis in virologically suppressed women living with HIV (WLWH). METHODS AND RESULTS Nested within the Women's Interagency HIV Study, we conducted a cross-sectional comparison of IL-32 between 399 WLWH and 100 women without HIV, followed by a case-control study of 72 WLWH (36 carotid artery plaque cases vs. 36 age-matched controls without plaque). Plasma IL-32 protein was measured by ELISA, and mRNA of IL-32 isoforms (IL-32α, β, γ, D, ε, and θ) was quantified by reverse transcription polymerase chain reaction from peripheral blood mononuclear cells. Plasma IL-32 protein levels were higher in WLWH compared with women without HIV (P = 0.02). Among WLWH, although plasma IL-32 levels did not differ significantly between plaque cases and controls, expression of IL-32 isoforms α, β, and ε mRNA was significantly higher in peripheral blood mononuclear cells from cases (P = 0.01, P = 0.005, and P = 0.018, respectively). Upregulation of IL-32β and IL-32ε among WLWH with carotid artery plaque persisted after adjustment for age, race/ethnicity, smoking, systolic blood pressure, body mass index, and history of hepatitis C virus (P = 0.04 and P = 0.045); the adjusted association for IL-32α was marginally significant (P = 0.07). CONCLUSIONS IL-32 isoforms should be studied further as potential CVD biomarkers. This is of particular interest in WLWH by virtue of altered IL-32 levels in this population.
Collapse
Affiliation(s)
| | | | - Madeleine Durand
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | | | | | - Carl Chartrand-Lefebvre
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Radiologie, Radio-oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal
| | - Guy Cloutier
- CHUM-Research Centre, Montréal, QC, Canada
- Département de radiologie et Institut de génie biomedical, Université de Montréal, Montréal, QC, Canada
| | | | - Seble Kassaye
- Department of medicine, Georgetown University, Washington, DC, USA
| | - Roksana Karim
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jorge R. Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Audrey L. French
- Division of Infectious Diseases, Stroger Hospital of Cook County, Chicago IL, USA
| | - Stephen J. Gange
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jason M. Lazar
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Howard N. Hodis
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA, USA
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | - Nicolas Chomont
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | | | - Robert C. Kaplan
- Albert Einstein College of Medicine, Bronx, NY, USA
- Divsion of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle WA, USA
| | - Cécile L. Tremblay
- CHUM-Research Centre, Montréal, QC, Canada
- Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
37
|
Furch BD, Mwakamui S, Sianongo S, Zyambo K, Heimburger DC, Koethe JR, Kelly P. Contribution of Schistosoma mansoni to systemic inflammation and microbial translocation among people with HIV in Zambia. Trans R Soc Trop Med Hyg 2021; 116:179-186. [PMID: 34263318 DOI: 10.1093/trstmh/trab103] [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: 02/02/2021] [Revised: 03/31/2021] [Accepted: 06/30/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Schistosoma mansoni is hyperendemic in many rural areas of Zambia where up to 77% of people are positive for infection via serologic evaluation. Zambia also has a high prevalence of HIV infection. Individually, S. mansoni and HIV infection impair gastrointestinal barrier integrity and induce inflammation, but the effects of coinfection are not well understood. We set out to test the hypothesis that HIV would exacerbate intestinal barrier failure in patients with S. mansoni infection. METHODS Adults attending medical outpatient clinics in Kaoma, Western Province, Zambia, were enrolled in a case-control study to determine the relative contributions of schistosomiasis and HIV to microbial translocation (measured as soluble CD14 [sCD14] and lipopolysaccharide binding protein [LBP]) and inflammation (measured as CRP). RESULTS Among 152 adults evaluated, 74 (49%) were HIV-seropositive, 45 (29%) were shedding schistosome ova (Kato-Katz), 120 (81%) were seropositive for schistosome antibodies (i.e. prior or current infection, with or without egg shedding) and 16 (11%) were HIV/schistosome coinfected (defined by Kato-Katz). HIV infection was associated with higher circulating sCD14 concentrations (p=0.003 by Kruskal-Wallis test), but schistosomiasis was not. HIV infection was associated with greater exposure to schistosomes assessed serologically (OR=2.48, 95% CI 1.05 to 5.86; p=0.03), but reduced likelihood of egg shedding (OR 0.47, 95% CI 0.21 to 1.01; p=0.03). CONCLUSIONS There was no evidence for a compounding or synergistic effect of coinfection on microbial translocation that appeared to be correlated with HIV infection. Further studies are needed to understand how the increase in LBP secondary to HIV infection may decrease schistosome egg excretion in coinfected individuals.
Collapse
Affiliation(s)
- Briana D Furch
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Nationalist Road, Lusaka, Zambia.,Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Simutanyi Mwakamui
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Nationalist Road, Lusaka, Zambia
| | - Sandie Sianongo
- University Teaching Hospital, Nationalist Rd, Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Nationalist Road, Lusaka, Zambia
| | - Douglas C Heimburger
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA.,Department of Internal Medicine, University of Zambia School of Medicine Nationalist Rd, Lusaka, Zambia
| | - John R Koethe
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Nationalist Road, Lusaka, Zambia.,Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, Turner Street, London E1 2AD, UK
| |
Collapse
|
38
|
Spagnolo-Allende A, Gutierrez J. Role of Brain Arterial Remodeling in HIV-Associated Cerebrovascular Outcomes. Front Neurol 2021; 12:593605. [PMID: 34239489 PMCID: PMC8258100 DOI: 10.3389/fneur.2021.593605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 05/07/2021] [Indexed: 01/11/2023] Open
Abstract
As the life expectancy of people living with HIV (PLWH) on combination antiretroviral therapy (cART) increases, so does morbidity from cerebrovascular disease and neurocognitive disorders. Brain arterial remodeling stands out as a novel investigational target to understand the role of HIV in cerebrovascular and neurocognitive outcomes. We therefore conducted a review of publications in PubMed, EMBASE, Web of Science and Wiley Online Library, from inception to April 2021. We included search terms such as HIV, cART, brain, neuroimmunity, arterial remodeling, cerebrovascular disease, and neurocognitive disorders. The literature shows that, in the post-cART era, PLWH continue to experience an increased risk of stroke and neurocognitive disorders (albeit milder forms) compared to uninfected populations. PLWH who are immunosuppressed have a higher proportion of hemorrhagic strokes and strokes caused by opportunistic infection and HIV vasculopathy, while PLWH on long-term cART have higher rates of ischemic strokes, compared to HIV-seronegative controls. Brain large artery atherosclerosis in PLWH is associated with lower CD4 nadir and higher CD4 count during the stroke event. HIV vasculopathy, a form of non-atherosclerotic outward remodeling, on the other hand, is associated with protracted immunosuppression. HIV vasculopathy was also linked to a thinner media layer and increased adventitial macrophages, suggestive of non-atherosclerotic degeneration of the brain arterial wall in the setting of chronic central nervous system inflammation. Cerebrovascular architecture seems to be differentially affected by HIV infection in successfully treated versus immunosuppressed PLWH. Brain large artery atherosclerosis is prevalent even with long-term immune reconstitution post-cART. HIV-associated changes in brain arterial walls may also relate to higher rates of HIV-associated neurocognitive disorders, although milder forms are more prevalent in the post-cART era. The underlying mechanisms of HIV-associated pathological arterial remodeling remain poorly understood, but a role has been proposed for chronic HIV-associated inflammation with increased burden on the vasculature. Neuroimaging may come to play a role in assessing brain arterial remodeling and stratifying cerebrovascular risk, but the data remains inconclusive. An improved understanding of the different phenotypes of brain arterial remodeling associated with HIV may reveal opportunities to reduce rates of cerebrovascular disease in the aging population of PLWH on cART.
Collapse
Affiliation(s)
| | - Jose Gutierrez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
39
|
González-Cordón A, Assoumou L, Moyle G, Waters L, Johnson M, Domingo P, Fox J, Stellbrink HJ, Guaraldi G, Masiá M, Gompels M, De Wit S, Florence E, Esser S, Raffi F, Behrens G, Pozniak A, Gatell JM, Martínez E. Switching from boosted PIs to dolutegravir decreases soluble CD14 and adiponectin in high cardiovascular risk people living with HIV. J Antimicrob Chemother 2021; 76:2380-2393. [PMID: 34120186 DOI: 10.1093/jac/dkab158] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Switching from boosted PIs to dolutegravir in people living with HIV (PLWH) with high cardiovascular risk improved plasma lipids at 48 weeks in the NEAT022 trial. Whether this strategy may have an impact on cardiovascular biomarkers is unknown. METHODS We assessed 48 week changes in biomarkers associated with inflammation, endothelial dysfunction, monocyte immune activation, oxidation, insulin resistance, hypercoagulability, heart failure, myocardial injury, and glomerular and tubular kidney injury. RESULTS Of 415 PLWH randomized in the NEAT022 study, 313 (75.4%) remained on allocated therapy and had paired samples available. Soluble CD14 (-11%, P < 0.001) and adiponectin (-11%, P < 0.001) significantly declined and high-sensitive C-reactive protein (-13%, P = 0.069) and oxidized LDL (-13%, P = 0.084) tended to decrease with dolutegravir. Switching to dolutegravir remained significantly associated with soluble CD14 and adiponectin reductions after adjustment for baseline variables. There were inverse correlations between soluble CD14 and CD4 count changes (P = 0.05), and between adiponectin and BMI changes (P < 0.001). CONCLUSIONS Switching from boosted PIs to dolutegravir in PLWH with high cardiovascular risk led to soluble CD14 and adiponectin reductions at 48 weeks. While decreasing soluble CD14 may entail favourable health effects in PLWH, adiponectin reduction may reflect less insulin sensitivity associated with weight gain.
Collapse
Affiliation(s)
| | - Lambert Assoumou
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Paris, France
| | - Graeme Moyle
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Laura Waters
- Mortimer Market Centre, Central & North West London NHS Foundation Trust, London, UK
| | | | | | - Julie Fox
- Guy's & St Thomas' NHS Foundation Trust, London, UK
| | | | | | - Mar Masiá
- Hospital General Universitario de Elche, Elche, Spain
| | | | - Stephane De Wit
- Centre Hospitalier Universitaire Saint-Pierre, Brussels, Belgium
| | | | | | | | | | - Anton Pozniak
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - José M Gatell
- Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain.,ViiV Healthcare, Brentford, UK
| | - Esteban Martínez
- Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Siedner MJ, Bibangambah P, Kim J, Lankowski A, Chang JL, Yang IT, Kwon DS, North CM, Triant VA, Longenecker C, Ghoshhajra B, Peck RN, Sentongo RN, Gilbert R, Kakuhikire B, Boum Y, Haberer JE, Martin JN, Tracy R, Hunt PW, Bangsberg DR, Tsai AC, Hemphill LC, Okello S. Treated HIV Infection and Progression of Carotid Atherosclerosis in Rural Uganda: A Prospective Observational Cohort Study. J Am Heart Assoc 2021; 10:e019994. [PMID: 34096320 PMCID: PMC8477876 DOI: 10.1161/jaha.120.019994] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Although ≈70% of the world's population of people living with HIV reside in sub-Saharan Africa, there are minimal prospective data on the contributions of HIV infection to atherosclerosis in the region. Methods and Results We conducted a prospective observational cohort study of people living with HIV on antiretroviral therapy >40 years of age in rural Uganda, along with population-based comparators not infected with HIV. We collected data on cardiovascular disease risk factors and carotid ultrasound measurements annually. We fitted linear mixed effects models, adjusted for cardiovascular disease risk factors, to estimate the association between HIV serostatus and progression of carotid intima media thickness (cIMT). We enrolled 155 people living with HIV and 154 individuals not infected with HIV and collected cIMT images at 1045 visits during a median of 4 annual visits per participant (interquartile range 3-4, range 1-5). Age (median 50.9 years) and sex (49% female) were similar by HIV serostatus. At enrollment, there was no difference in mean cIMT by HIV serostatus (0.665 versus 0.680 mm, P=0.15). In multivariable models, increasing age, blood pressure, and non-high-density lipoprotein cholesterol were associated with greater cIMT (P<0.05), however change in cIMT per year was also no different by HIV serostatus (0.004 mm/year for HIV negative [95% CI, 0.001-0.007 mm], 0.006 mm/year for people living with HIV [95% CI, 0.003-0.008 mm], HIV×time interaction P=0.25). Conclusions In rural Uganda, treated HIV infection was not associated with faster cIMT progression. These results do not support classification of treated HIV infection as a risk factor for subclinical atherosclerosis progression in rural sub-Saharan Africa. Registration URL: https://www.ClinicalTrials.gov; Unique identifier: NCT02445079.
Collapse
Affiliation(s)
- Mark J. Siedner
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Prossy Bibangambah
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - June‐Ho Kim
- Department of MedicineHarvard Medical SchoolBostonMA,Department of MedicineBrigham and Women's HospitalBostonMA
| | - Alexander Lankowski
- Department of MedicineUniversity of WashingtonSeattleWA,Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWA
| | - Jonathan L. Chang
- Department of MedicineHarvard Medical SchoolBostonMA,Department of MedicineBrigham and Women's HospitalBostonMA
| | - Isabelle T. Yang
- Department of MedicineGeisel School of Medicine at DartmouthHanoverNH
| | - Douglas S. Kwon
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and HarvardCambridgeMA
| | - Crystal M. North
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Virginia A. Triant
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | | | - Brian Ghoshhajra
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Robert N. Peck
- Center for Global HealthWeill Cornell Medical CollegeNew YorkNY
| | - Ruth N. Sentongo
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Rebecca Gilbert
- Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Bernard Kakuhikire
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Yap Boum
- Epicentre Research BaseMbararaUganda
| | - Jessica E. Haberer
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | | | - Russell Tracy
- Department of Pathology and Laboratory MedicineUniversity of VermontBurlingtonVT
| | - Peter W. Hunt
- Department of MedicineUniversity of CaliforniaSan FranciscoCA
| | | | - Alexander C. Tsai
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Linda C. Hemphill
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Samson Okello
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| |
Collapse
|
41
|
Stanislawski MA, Lange LA, Raffield LM, Zakai NA, Meyer M, Ferrier K, Szeto MD, Leavitt C, Shortt JA, Thornton TA, Tracy RP, Auer PL, Reiner AP, Lange EM, Olson NC. Soluble CD14 Levels in the Jackson Heart Study: Associations With Cardiovascular Disease Risk and Genetic Variants. Arterioscler Thromb Vasc Biol 2021; 41:e369-e378. [PMID: 33910371 PMCID: PMC8159903 DOI: 10.1161/atvbaha.121.316035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/13/2021] [Indexed: 01/20/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Maggie A Stanislawski
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | - Leslie A Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
- Department of Epidemiology (L.A.L.), University of Colorado School of Public Health, Aurora
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill (L.M.R.)
| | - Neil A Zakai
- Department of Pathology and Laboratory Medicine (N.A.Z., R.P.T., N.C.O.), Larner College of Medicine, University of Vermont, Burlington
- Department of Medicine (N.A.Z.), Larner College of Medicine, University of Vermont, Burlington
| | - Mariah Meyer
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | - Kendra Ferrier
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | - Mindy D Szeto
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | - Colton Leavitt
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | - Jonathan A Shortt
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
| | | | - Russell P Tracy
- Department of Pathology and Laboratory Medicine (N.A.Z., R.P.T., N.C.O.), Larner College of Medicine, University of Vermont, Burlington
- Department of Biochemistry (R.P.T.), Larner College of Medicine, University of Vermont, Burlington
| | - Paul L Auer
- Department of Biostatistics, School of Public Health, University of Wisconsin, Milwaukee (P.L.A.)
| | - Alex P Reiner
- Department of Epidemiology (A.P.R.), University of Washington, Seattle
| | - Ethan M Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora (M.A.S., L.A.L., M.M., K.K., M.D.S., C.L., J.A.S., E.M.L.)
- Department of Biostatistics and Informatics (E.M.L.), University of Colorado School of Public Health, Aurora
| | - Nels C Olson
- Department of Pathology and Laboratory Medicine (N.A.Z., R.P.T., N.C.O.), Larner College of Medicine, University of Vermont, Burlington
| |
Collapse
|
42
|
Kipke J, Margevicius S, Kityo C, Mirembe G, Buggey J, Yun C, Hung C, McComsey GA, Longenecker CT. Sex, HIV Status, and Measures of Cardiac Stress and Fibrosis in Uganda. J Am Heart Assoc 2021; 10:e018767. [PMID: 33998251 PMCID: PMC8483535 DOI: 10.1161/jaha.120.018767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Biomarkers of myocardial stress and fibrosis are elevated in people living with HIV and are associated with cardiac dysfunction. It is unknown whether sex influences these markers of heart failure risk in sub‐Saharan Africa, where HIV burden is high and where the vast majority of women with HIV live. Methods and Results Echocardiograms and 6 plasma biomarkers (suppression of tumorigenicity‐2, growth differentiation factor 15, galectin 3, soluble fms‐like tyrosine kinase‐1, NT‐proBNP [N‐terminal pro‐B‐type natriuretic peptide], and cystatin C) were obtained from 100 people living with HIV on antiretroviral therapy and 100 HIV‐negative controls in Uganda. All participants were ≥45 years old with ≥1 major cardiovascular risk factor. Multivariable linear and logistic regression models were used to assess associations between biomarkers, echocardiographic variables, HIV status, and sex, and to assess whether sex modified these associations. Overall, mean age was 56 years and 62% were women. Suppression of tumorigenicity‐2 was higher in men versus women (P<0.001), and growth differentiation factor 15 was higher in people living with HIV versus controls (P<0.001). Sex modified the HIV effect on cystatin C and NT‐proBNP (both P for interaction <0.025). Women had more diastolic dysfunction than men (P=0.02), but there was no evidence of sex‐modifying HIV effects on cardiac structure and function. Cardiac biomarkers were more strongly associated with left ventricular mass index in men compared with women. Conclusions There are prominent differences in biomarkers of cardiac fibrosis and stress by sex and HIV status in Uganda. The predictive value of cardiac biomarkers for heart failure in people living with HIV in sub‐Saharan Africa should be examined, and novel risk markers for women should be further explored.
Collapse
Affiliation(s)
- Jasmine Kipke
- Case Western Reserve University School of MedicineClevelandOH
| | | | | | | | | | | | | | - Grace A. McComsey
- Case Western Reserve University School of MedicineClevelandOH
- University Hospitals Cleveland Medical CenterClevelandOH
| | - Chris T. Longenecker
- Case Western Reserve University School of MedicineClevelandOH
- University Hospitals Cleveland Medical CenterClevelandOH
| |
Collapse
|
43
|
Harding BN, Avoundjian T, Heckbert SR, Whitney BM, Nance RM, Ruderman SA, Kalani R, Tirschwell DL, Ho EL, Becker KJ, Zunt J, Chow F, Huffer A, Mathews WC, Eron J, Moore RD, Marra CM, Burkholder G, Saag MS, Kitahata MM, Crane HM, Delaney JC. HIV Viremia and Risk of Stroke Among People Living with HIV Who Are Using Antiretroviral Therapy. Epidemiology 2021; 32:457-464. [PMID: 33591056 PMCID: PMC8012252 DOI: 10.1097/ede.0000000000001331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rates of stroke are higher in people living with HIV compared with age-matched uninfected individuals. Causes of elevated stroke risk, including the role of viremia, are poorly defined. METHODS Between 1 January 2006 and 31 December 2014, we identified incident strokes among people living with HIV on antiretroviral therapy at five sites across the United States. We considered three parameterizations of viral load (VL) including (1) baseline (most recent VL before study entry), (2) time-updated, and (3) cumulative VL (copy-days/mL of virus). We used Cox proportional hazards models to estimate hazard ratios (HRs) for stroke risk comparing the 75th percentile ("high VL") to the 25th percentile ("low VL") of baseline and time-updated VL. We used marginal structural Cox models, with most models adjusted for traditional stroke risk factors, to estimate HRs for stroke associated with cumulative VL. RESULTS Among 15,974 people living with HIV, 139 experienced a stroke (113 ischemic; 18 hemorrhagic; eight were unknown type) over a median follow-up of 4.2 years. Median baseline VL was 38 copies/mL (interquartile interval: 24, 3,420). High baseline VL was associated with increased risk of both ischemic (HR: 1.3; 95% CI = 0.96-1.7) and hemorrhagic stroke (HR: 3.1; 95% CI = 1.6-5.9). In time-updated models, high VL was also associated with an increased risk of any stroke (HR: 1.8; 95% CI = 1.4-2.3). We observed no association between cumulative VL and stroke risk. CONCLUSIONS Our findings are consistent with the hypothesis that elevated HIV VL may increase stroke risk, regardless of previous VL levels.
Collapse
Affiliation(s)
| | | | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA
| | | | - Robin M Nance
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Rizwan Kalani
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Emily L Ho
- Department of Medicine, University of Washington, Seattle, WA
| | - Kyra J Becker
- Department of Medicine, University of Washington, Seattle, WA
| | - Joseph Zunt
- Department of Medicine, University of Washington, Seattle, WA
| | - Felicia Chow
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Andrew Huffer
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Joseph Eron
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | | | | | - Greer Burkholder
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Michael S Saag
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Mari M Kitahata
- Department of Medicine, University of Washington, Seattle, WA
| | - Heidi M Crane
- Department of Medicine, University of Washington, Seattle, WA
| | - Joseph C Delaney
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
44
|
Zhang E, Chai JC, Deik AA, Hua S, Sharma A, Schneider MF, Gustafson D, Hanna DB, Lake JE, Rubin LH, Post WS, Anastos K, Brown T, Clish CB, Kaplan RC, Qi Q. Plasma Lipidomic Profiles and Risk of Diabetes: 2 Prospective Cohorts of HIV-Infected and HIV-Uninfected Individuals. J Clin Endocrinol Metab 2021; 106:999-1010. [PMID: 33420793 PMCID: PMC7993589 DOI: 10.1210/clinem/dgab011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Antiretroviral therapy (ART) use is associated with disrupted lipid and glucose metabolism in people with HIV infection. We aimed to identify plasma lipid species associated with risk of diabetes in the context of HIV infection. RESEARCH DESIGN AND METHODS We profiled 211 plasma lipid species in 491 HIV-infected and 203 HIV-uninfected participants aged 35 to 55 years from the Women's Interagency HIV Study and the Multicenter AIDS Cohort Study. Cox proportional hazards model was used to examine associations between baseline lipid species and incident diabetes (166 diabetes cases were identified during a median follow-up of 12.6 years). RESULTS We identified 11 lipid species, representing independent signals for 8 lipid classes/subclasses, associated with risk of diabetes (P < 0.05 after FDR correction). After adjustment for multiple covariates, cholesteryl ester (CE) (22:4), lysophosphatidylcholine (LPC) (18:2), phosphatidylcholine (PC) (36:4), phosphatidylcholine plasmalogen (34:3), and phosphatidylethanolamine (PE) (38:2) were associated with decreased risk of diabetes (HRs = 0.70 to 0.82 per SD increment), while diacylglycerol (32:0), LPC (14:0), PC (38:3), PE (36:1), and triacylglycerol (50:1) were associated with increased risk of diabetes (HRs = 1.26 to 1.56 per SD increment). HIV serostatus did not modify any lipid-diabetes associations; however, most of these lipid species were positively associated with HIV and/or ART use, including 3 diabetes-decreased ( CE [22:4], LPC [18:2], PE [38:2]) and all 5 diabetes-increased lipid species. CONCLUSIONS This study identified multiple plasma lipid species associated with incident diabetes. Regardless of the directions of their associations with diabetes, most diabetes-associated lipid species were elevated in ART-treated people with HIV infection. This suggests a complex role of lipids in the link between ART and diabetes in HIV infection.
Collapse
Affiliation(s)
- Eric Zhang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jin Choul Chai
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amy A Deik
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Simin Hua
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anjali Sharma
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael F Schneider
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Deborah Gustafson
- Department of Neurology, State University of New York-Downstate Medical Center, Brooklyn, NY, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jordan E Lake
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Leah H Rubin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Neurology and Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wendy S Post
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Todd Brown
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle WA, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
45
|
Feinstein MJ, Doyle MF, Stein JH, Sitlani CM, Fohner AE, Huber SA, Landay AL, Heckbert SR, Rice K, Kronmal RA, Hedrick C, Manichaikul A, McNamara C, Rich S, Tracy RP, Olson NC, Psaty BM, Delaney JAC. Nonclassical Monocytes (CD14dimCD16+) Are Associated With Carotid Intima-Media Thickness Progression for Men but Not Women: The Multi-Ethnic Study of Atherosclerosis-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:1810-1817. [PMID: 33761764 PMCID: PMC8057525 DOI: 10.1161/atvbaha.120.315886] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Supplemental Digital Content is available in the text. Objective: Few studies of population-based cohorts have investigated prospective associations of lymphoid and myeloid cell subsets in cardiovascular disease onset and progression. The purpose of this analysis was to determine associations of prespecified myeloid and lymphoid lineage cell subsets with common carotid artery intima-media thickness (IMT) progression. Approach and Results: We performed a prospective case-cohort study of 1195 participants from the Multi-Ethnic Study of Atherosclerosis who had peripheral blood mononuclear cells stored from the baseline examination. Key exposure variables were prespecified subsets of lymphoid and myeloid lineage immune cells, phenotyped by multicolor flow cytometry. The primary outcome was progression from baseline (Exam 1) to year 10 (Exam 5) in common carotid IMT. Higher proportions of nonclassical monocytes (CD14dimCD16++) were significantly associated with IMT progression over 10 years, but classical monocytes (CD14++CD16−), CD4+CD28− T cells, and T helper cells producing IL-17 (interleukin 17; T helper 17 cells) were not associated with significant changes in IMT over 10 years. There were significant interactions between monocyte subsets and sex with respect to IMT progression: in sex-stratified analyses, nonclassical monocytes were associated with significant IMT progression and classical monocytes were associated with significant IMT regression for men, whereas there were no significant associations of monocyte subsets with IMT change for women. Conclusions: Nonclassical monocytes were associated with progression of carotid IMT. There were significant sex differences in associations of monocyte subsets with IMT progression: for men, nonclassical monocytes were associated with IMT progression and classical monocytes were associated with regression, whereas these associations were null for women.
Collapse
Affiliation(s)
- Matthew J Feinstein
- Departments of Medicine and Preventive Medicine, Northwestern University, Chicago, IL (M.J.F.)
| | - Margaret F Doyle
- University of Vermont, Burlington (M.F.D., S.A.H., R.P.T., N.C.O.)
| | | | - Colleen M Sitlani
- University of Washington, Seattle (C.M.S., A.E.F., S.R.H., K.R., R.A.K.)
| | - Alison E Fohner
- University of Washington, Seattle (C.M.S., A.E.F., S.R.H., K.R., R.A.K.)
| | - Sally A Huber
- University of Vermont, Burlington (M.F.D., S.A.H., R.P.T., N.C.O.)
| | - Alan L Landay
- Rush University Medical Center, Chicago, IL (A.L.L.)
| | - Susan R Heckbert
- University of Washington, Seattle (C.M.S., A.E.F., S.R.H., K.R., R.A.K.)
| | - Kenneth Rice
- University of Washington, Seattle (C.M.S., A.E.F., S.R.H., K.R., R.A.K.)
| | - Richard A Kronmal
- University of Washington, Seattle (C.M.S., A.E.F., S.R.H., K.R., R.A.K.)
| | | | | | | | - Stephen Rich
- University of Virginia, Charlottesville (A.M., C.M., S.R.)
| | - Russell P Tracy
- University of Vermont, Burlington (M.F.D., S.A.H., R.P.T., N.C.O.)
| | - Nels C Olson
- University of Vermont, Burlington (M.F.D., S.A.H., R.P.T., N.C.O.)
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle (B.M.P.).,Kaiser Permanente Washington Health Research Institute, Seattle (B.M.P.)
| | | |
Collapse
|
46
|
Xhakollari L, Jujic A, Molvin J, Nilsson P, Holm H, Bachus E, Leosdottir M, Grubb A, Christensson A, Magnusson M. Proteins linked to atherosclerosis and cell proliferation are associated with the shrunken pore syndrome in heart failure patients: Shrunken pore syndrome and proteomic associations. Proteomics Clin Appl 2021; 15:e2000089. [PMID: 33682349 DOI: 10.1002/prca.202000089] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/12/2021] [Accepted: 03/02/2021] [Indexed: 01/23/2023]
Abstract
PURPOSE The "Shrunken pore syndrome" (SPS) is characterized by a difference in renal filtration between cystatin C and creatinine, resulting in a low eGFRcystatinC /eGFRcreatinine -ratio. Studies have demonstrated a high risk for cardiovascular morbidity and mortality for patients with SPS. In this discovery study, we explored associations between SPS and proteins implicated in cardiovascular disease and inflammation in patients with heart failure. EXPERIMENTAL DESIGN Plasma samples from 300 individuals in HARVEST-Malmö trial hospitalized for the diagnosis of heart failure (mean age 74.9 ± 11.5 years; 30.0% female), were analyzed with a proximity extension assay consisting of 92 proteins. A Bonferroni-corrected p-value of 0.05/92 = 5.4 × 10-4 was considered significant in the initial age and sex-adjusted analyses. Presence of SPS was defined as eGFRcystatinC ≤ 60% of eGFRcreatinine . RESULTS SPS presented with significant associations (p < 5.4 × 10-4 ) in age and sex-adjusted logistic regressions with elevated levels of six proteins; scavenger receptor cysteine rich type 1 protein M130, tumor necrosis factor receptor 1, tumor necrosis factor receptor 2, osteoprotegerin, interleukin-2 receptor subunit alpha, and tyrosine-protein kinase receptor UFO. All proteins remained associated (p < 0.05) with SPS after multivariate adjustments. CONCLUSIONS AND CLINICAL RELEVANCE In heart failure patients, SPS was associated with proteins linked to atherosclerosis and cell proliferation.
Collapse
Affiliation(s)
- Liana Xhakollari
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Nephrology, Skåne University Hospital, Malmö, Sweden
| | - Amra Jujic
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Skane University Hospital, Department of Cardiology, Lund University, Malmö, Sweden
| | - John Molvin
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Skane University Hospital, Department of Cardiology, Lund University, Malmö, Sweden
| | - Peter Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Hannes Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Skane University Hospital, Department of Cardiology, Lund University, Malmö, Sweden
| | - Erasmus Bachus
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Margret Leosdottir
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Skane University Hospital, Department of Cardiology, Lund University, Malmö, Sweden
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund University, Lund, Sweden
| | - Anders Christensson
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Nephrology, Skåne University Hospital, Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Skane University Hospital, Department of Cardiology, Lund University, Malmö, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.,Hypertension in Africa Research Team, North-West University Potchefstroom, Potchefstroom, South Africa
| |
Collapse
|
47
|
Ghosn J, Abdoul H, Fellahi S, Merlet A, Salmon D, Morini JP, Deleuze J, Blacher J, Capeau J, Bastard JP, Viard JP. Prevalence of Silent Atherosclerosis and Other Comorbidities in an Outpatient Cohort of Adults Living with HIV: Associations with HIV Parameters and Biomarkers. AIDS Res Hum Retroviruses 2021; 37:101-108. [PMID: 33076677 DOI: 10.1089/aid.2020.0182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
People living with HIV (PLWH) are at risk of noninfectious comorbidities. It is important to individualize those at higher risk. In a single-center cohort of PLWH, we performed a cross-sectional analysis of comorbidities, diagnosed according to standard procedures. The primary endpoint was the prevalence of subclinical carotid/coronary atherosclerosis. Secondary endpoints were its association with selected inflammatory/immune activation biomarkers and with other comorbidities. Associations were examined by using Chi-square or Fisher's exact test for categorical variables and Student or Wilcoxon tests for quantitative variables, and a stepwise multivariate logistical model was performed for further exploration. Among 790 participants [median age: 49.8 years (interquartile range, IQR: 44.5-55.6), 77.1% males, median CD4: 536/mm3 (IQR: 390-754), 83.6% with undetectable viral load], asymptomatic atherosclerosis was found in 26% and was associated in multivariate analysis with older age, longer known duration of infection, higher sCD14, and lower adiponectin levels. Hypertension was found in 33.5% of participants, diabetes in 19.4%, renal impairment in 14.6%, elevated low-density lipoprotein-cholesterol in 13.3%, elevated triglyceride/high-density lipoprotein (HDL)-cholesterol ratio in 6.6%, and osteoporosis in 7.9%. The presence of two or more comorbidities was found in 42.1% of participants and was associated in multivariate analysis with older age and longer exposure to antiretrovirals. Comorbidities were diversely associated with biomarkers: osteoporosis with higher IL-6, renal impairment with higher sCD14, hypertension with higher D-dimer, diabetes and elevated triglyceride/HDL-cholesterol ratio both with lower adiponectin and lower 25-hydroxyvitamin D. Asymptomatic atherosclerosis and multimorbidity were frequent in a cohort of middle-aged, well-controlled, PLWH and were associated with traditional and HIV-specific factors. Associations between morbidities and inflammatory/immune activation biomarkers were diverse.
Collapse
Affiliation(s)
- Jade Ghosn
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Hendy Abdoul
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Soraya Fellahi
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Audrey Merlet
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Dominique Salmon
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jean-Pierre Morini
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jean Deleuze
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jacques Blacher
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jacqueline Capeau
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jean-Philippe Bastard
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| | - Jean-Paul Viard
- Immunology-Infectious Diseases Unit, Hôtel-Dieu Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Centre-Université de Paris, Paris, France
| |
Collapse
|
48
|
Utay NS, Overton ET. Immune Activation and Inflammation in People With Human Immunodeficiency Virus: Challenging Targets. J Infect Dis 2021; 221:1567-1570. [PMID: 31282534 DOI: 10.1093/infdis/jiz351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Netanya S Utay
- Division of General Internal Medicine, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston
| | - Edgar T Overton
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham School of Medicine
| |
Collapse
|
49
|
Kuk M, Ward NC, Dwivedi G. Extrinsic and Intrinsic Responses in the Development and Progression of Atherosclerosis. Heart Lung Circ 2021; 30:807-816. [PMID: 33468387 DOI: 10.1016/j.hlc.2020.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 11/25/2022]
Abstract
Atherosclerosis is a multifactorial disease that is thought to be primarily inflammatory in origin. Given the contribution of inflammation to the development and progression of atherosclerosis, other conditions that are characterised by a dysregulated inflammatory response have also been proposed to play a role. The purpose of this review is to organise and present the various inflammatory processes that can affect atherosclerosis into two broad categories: extrinsic or host-independent and intrinsic or host-dependent. Within these two categories, we will discuss various processes that may contribute to the development and progression of atherosclerosis and the clinical studies describing these associations. Although the clinical trials investigating anti-inflammatory therapies have to date provided mixed results, further studies, particularly in conjunction with lipid-lowering and blood pressure lowering therapies should be considered.
Collapse
Affiliation(s)
- Mariya Kuk
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada; McGill University Health Centre, McGill University, Montreal, Canada
| | - Natalie C Ward
- School of Public Health, Curtin University, Perth, WA, Australia; Medical School, University of Western Australia, Perth, WA, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, WA, Australia; Harry Perkins Institute for Medical Research, Fiona Stanley Hospital, Perth, WA, Australia.
| |
Collapse
|
50
|
Temu TM, Zifodya JS, Polyak SJ, Wagoner J, Wanjalla CN, Masyuko S, Nyabiage J, Kinuthia J, Bloomfield GS, Page ST, Farquhar C. Antiretroviral therapy reduces but does not normalize immune and vascular inflammatory markers in adults with chronic HIV infection in Kenya. AIDS 2021; 35:45-51. [PMID: 33055570 PMCID: PMC7718419 DOI: 10.1097/qad.0000000000002729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Markers of monocyte/macrophage activation and vascular inflammation are associated with HIV-related cardiovascular diseases (CVD) and mortality. We compared these markers among African people living with HIV (PLWH) and HIV-negative adults, and examined risk factors associated with elevated biomarkers (>75th percentile) in PLWH on antiretroviral therapy (ART). DESIGN Cross-sectional study. METHODS We measured serum concentrations of a gut integrity biomarker (intestinal-fatty acid binding protein), monocyte/macrophage activation biomarkers (soluble CD14 and CD163), and vascular inflammation biomarkers [soluble intercellular adhesion molecule 1 (sICAM-1) and soluble vascular adhesion molecule 1 (sVCAM-1)]. We assessed the relationship of these inflammatory parameters with HIV, using logistic regression adjusting for traditional CVD risk factors. RESULTS Among the 541 participants, median age was 43 years and half were female. Among 275 PLWH, median CD4 T-cell count and duration of ART use was 509 cells/μl and 8 years, respectively. PLWH had significantly higher prevalence of elevated inflammatory biomarkers compared with HIV-negative individuals even after adjustment for traditional CVD risk factors. Compared with individuals without HIV, the prevalence of elevated biomarkers was highest among persons with detectable viral load and CD4 T-cell counts 200 cells/μl or less. In a subanalysis among PLWH, nadir CD4 T-cell count 200 cells/μl or less was associated with elevated soluble CD14 (sCD14); dyslipidemia with elevated sCD14, sICAM-1, and sVCAM-1; and overweight/obesity with reduced sCD14. Longer ART exposure (>4 years) was associated with reduced sVCAM-1 and sICAM-1. CONCLUSION HIV and not traditional CVD risk factors is a primary contributor of monocyte/macrophage activation and inflammation despite ART. Anti-inflammatory therapies in addition to ART may be necessary to reduce these immune dysregulations and improve health outcomes of African PLWH.
Collapse
Affiliation(s)
- Tecla M Temu
- Department of Global Health, University of Washington, Seattle, Washington
| | - Jerry S Zifodya
- Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Jessica Wagoner
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | | | - Sarah Masyuko
- Department of Global Health, University of Washington, Seattle, Washington
- Ministry of Health
- Kenyatta National Hospital, Nairobi, Kenya
| | - Jerusha Nyabiage
- Department of Global Health, University of Washington, Seattle, Washington
| | | | - Gerald S Bloomfield
- Department of Medicine and Duke Global Health Institute, Duke University, Durham, North Carolina
| | | | - Carey Farquhar
- Department of Global Health, University of Washington, Seattle, Washington
- Department of Medicine
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|