1
|
Schnittman SR, Talathi R, Wilks MQ, Hedgire S, Lu MT, Fourman LT, Alagpulinsa DA, Stockman SL, White KS, Wallis ZK, Autissier P, Stanley TL, Lee H, Honigberg MC, El-Fakhri G, Williams KC, Zanni MV, Grinspoon SK, Toribio M. Association of T-cell subtypes with macrophage-specific arterial infiltration in people with HIV. AIDS 2024; 38:1940-1946. [PMID: 38905489 DOI: 10.1097/qad.0000000000003967] [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: 03/14/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
People with human immunodeficiency virus (HIV, PWH) face an increased risk of cardiovascular disease (CVD) compared to the general population. We previously demonstrated that people with (versus without) HIV have higher macrophage-specific arterial infiltration in relation to systemic monocyte activation. We now show that select T lymphocyte subpopulations (naïve CD4 + , effector memory CD4 + , and central memory CD8 + ) are differentially associated with macrophage-specific arterial infiltration among participants with versus without HIV, with evidence of interaction by HIV status. Our results suggest that among PWH, circulating T lymphocytes associate with macrophage-specific arterial infiltration, of relevance to atherogenesis and CVD risk.
Collapse
Affiliation(s)
- Samuel R Schnittman
- Division of Infectious Diseases, Department of Medicine
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ria Talathi
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Moses Q Wilks
- Yale PET Center, Yale School of Medicine, New Haven, CT
| | - Sandeep Hedgire
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - David A Alagpulinsa
- Yale Center for Molecular and Systems Metabolism & Department of Comparative Medicine, Yale School of Medicine, New Haven, CT
| | - Sara L Stockman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | | | | | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School
| | - Michael C Honigberg
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Mabel Toribio
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Panigrahi S, Mayne E, Louw S, Funderburg NT, Chakraborty A, Jacobson JM, Carpenter SM, Lederman MM, Freeman ML, Sieg SF. Deciphering the role of endothelial granulocyte macrophage-CSF in chronic inflammation associated with HIV. iScience 2024; 27:110909. [PMID: 39391731 PMCID: PMC11465086 DOI: 10.1016/j.isci.2024.110909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/29/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
People with HIV (PWH) experience endothelial dysfunction (ED) that is aggravated by chronic inflammation and microbial translocation across a damaged gut barrier. Although this paradigm is well-described, downstream pathways that terminate in endothelial dysfunction are only partially understood. This study found increased expression of granulocyte macrophage colony stimulating factor (GM-CSF), toll-like receptor-4 (TLR4), and myeloperoxidase in the aortic endothelium of PWH compared to those without HIV. Bacteria-derived lipopolysaccharide (LPS) heightened glucose uptake and induced GM-CSF expression in primary human endothelial cells. Exposure to sodium-glucose cotransporter-2 (SGLT2) inhibitors reduced glucose uptake, GM-CSF release, and ED in LPS-activated endothelial cells ex vivo, and PWH treated with SGLT2 inhibitors for diabetes had significantly lower plasma GM-CSF levels than non-diabetic PWH not on this medication. The findings suggest that microbial products trigger glucose uptake and GM-CSF expression in the endothelium, contributing to localized inflammation in PWH. Modifying this altered state could offer therapeutic benefits.
Collapse
Affiliation(s)
- Soumya Panigrahi
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elizabeth Mayne
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town and National Health Laboratory Service, Cape Town, South Africa
| | - Susan Louw
- Department of Molecular Medicine and Hematology, School of Pathology, Faculty of Health Sciences, and National Health Laboratory Service, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Archeesha Chakraborty
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jeffrey M. Jacobson
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Stephen M. Carpenter
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael M. Lederman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Scott F. Sieg
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
3
|
Takahashi N, Eltalkhawy YM, Nasu K, Abdelnaser RA, Monde K, Habash SA, Nasser H, Hiyoshi M, Ishimoto T, Suzu S. IL-10 induces activated phenotypes of monocytes observed in virally-suppressed HIV-1-infected individuals. Biochem Biophys Res Commun 2024; 729:150342. [PMID: 38981402 DOI: 10.1016/j.bbrc.2024.150342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Despite viral suppression by effective combined antiretroviral therapy, HIV-1-infected individuals have an increased risk of non-AIDS-related overall morbidity, which is due to the persistent chronic inflammation exemplified by the activation of monocytes, such as increased CD16high subset, and elevated plasma level of soluble CD163 (sCD163) and soluble CD14 (sCD14). Here, we show that IL-10, which has been recognized as anti-inflammatory, induces these activated phenotypes of monocytes in vitro. IL-10 increased CD16high monocytes, which was due to the upregulation of CD16 mRNA expression and completely canceled by an inhibitor of Stat3. Moreover, IL-10 increased the production of sCD163 and sCD14 by monocytes, which was consistent with the upregulation of cell surface expression of CD163 and CD14, and mRNA expression of CD163. However, unlike the IL-10-indeuced upregulation of CD16, that of CD14 was minimally affected by the Stat3 inhibitor. Furthermore, the IL-10-induced upregulation of CD163 protein and mRNA was partially inhibited by the Stat3 inhibitor, but completely canceled by an inhibitor of AMPK, an upstream kinase of Stat3 and PI3K/Akt/mTORC1 pathways. In this study, we also found that HIV-1 pathogenic protein Nef, which is known to persist in plasma of virally-suppressed individuals, induced IL-10 production in monocyte-derived macrophages. Our results may suggest that IL-10, which is inducible by Nef-activated macrophages, is one of drivers for activated phenotypes of monocytes in virally-suppressed individuals, and that IL-10 induces the increased CD16high monocytes and elevated level of sCD163 and sCD14 through the activation of different signaling pathways.
Collapse
MESH Headings
- Humans
- Interleukin-10/metabolism
- Monocytes/metabolism
- Monocytes/immunology
- HIV Infections/immunology
- HIV Infections/virology
- HIV Infections/metabolism
- HIV Infections/blood
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- HIV-1
- Receptors, IgG/metabolism
- Lipopolysaccharide Receptors/metabolism
- STAT3 Transcription Factor/metabolism
- Phenotype
- Up-Regulation
- Cells, Cultured
Collapse
Affiliation(s)
- Naofumi Takahashi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| | - Youssef M Eltalkhawy
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kanako Nasu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Randa A Abdelnaser
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kazuaki Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Sara A Habash
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Hesham Nasser
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Masateru Hiyoshi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinya Suzu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
4
|
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 DOI: 10.1089/aid.2024.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
5
|
Garcia JR, Olivero R, Arrieta-Aldea I, Romero JA, Riera E, Cañas-Ruano E, Garrido N, Du J, Guerri R. Association between cardiovascular inflammation and alterations in immune system induced by HIV infection detected on [ 18F]FDG PET/MRI. Rev Esp Med Nucl Imagen Mol 2024; 43:500042. [PMID: 39127392 DOI: 10.1016/j.remnie.2024.500042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To assess by [18F]FDG PET/MR the biomarkers of HIV-induced inflammation at baseline and 1 year post-antiretroviral therapy (ART). METHODS Prospective study, 14 patients, newly diagnosed HIV-positive, asymptomatic. [18F]FDG PET/MRI (PET/MR-3.0T, Signa.GE) whole body and heart was performed, baseline and 1 year post-ART. Qualitative vascular assessment (hepatic reference). Quantitative assessment (SUVmax) of the whole body. T1 and T2 value estimation in 16 myocardial segments. RESULTS Baseline CMR showed in 3 (21.4%) a decreased LVEF, normalising post-TAR. Fibrosis was ruled out (T1), with no signs of myocardial oedema (T2) at baseline or post-TAR. Four (28.6%) showed baseline vascular [18F]FDG uptake, two in ascending thoracic aorta and two in ascending and descending thoracic aorta, normalising post-TAR. All (100%) showed basal lymph-nodes activity; supra (n:14) and infradiaphragmatic (n:13), laterocervical (n:14) and inguinal (n:13), with variable number of territories (9 patients >6;64.3%). Post-ART, 7 patients (50%) showed resolution and the other 7 reduction in extension (0 patients >5): 7 supra (100%) and 2 infradiaphragmatic (28.6%), 5 in the axilla and 2 in the groin. All (100%) had persistent basal adenoid uptake post-ART, 9 (64.3%) splenic all resolved post-ART and 7 (50.5%) gastric, persistent 3 post-ART. CONCLUSIONS Cardiovascular biomarkers by [18F]FDG PET/MR have shown baseline 28.6% of patients with large vessel activity and 21.4% with low LVEF, normalising post-ART. Inflammatory/immune biomarkers showed baseline activity in 100% of lymph-nodes, 100% adenoids, 64.3% splenic and 50.5% gastric. Post-TAR the reduction was 50% lymph-nodes, 0% adenoid, 100% splenic and 57.1% gastric.
Collapse
Affiliation(s)
| | | | | | | | - E Riera
- Unidad PET/RM, CETIR ASCIRES, Spain
| | - E Cañas-Ruano
- Servicio de Infecciosas, Hospital del Mar, Barcelona, Spain
| | | | - J Du
- Instituto de Investigaciones Médicas, Hospital del Mar, Barcelona, Spain
| | - R Guerri
- Servicio de Infecciosas, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
6
|
Paternò Raddusa MS, Marino A, Celesia BM, Spampinato S, Giarratana C, Venanzi Rullo E, Cacopardo B, Nunnari G. Atherosclerosis and Cardiovascular Complications in People Living with HIV: A Focused Review. Infect Dis Rep 2024; 16:846-863. [PMID: 39311207 PMCID: PMC11417834 DOI: 10.3390/idr16050066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
The intersection of Human Immunodeficiency Virus (HIV) infection and cardiovascular disease (CVD) represents a significant area of concern; advancements in antiretroviral therapy (ART) have notably extended the life expectancy of people living with HIV (PLWH), concurrently elevating the prevalence of chronic conditions such as CVD. This paper explores the multifaceted relationship between HIV infection, ART, and cardiovascular health, focusing on the mechanisms by which HIV and ART contribute to increased cardiovascular risk, including the promotion of endothelial dysfunction, inflammation, immune activation, and metabolic disturbances. We highlight the critical roles of HIV-associated proteins-Tat, Nef, and gp120-in accelerating atherosclerosis through direct and indirect pathways that exacerbate endothelial damage and inflammation. Additionally, we address the persistent challenge of chronic inflammation and immune activation in PLWH, factors that are strongly predictive of non-AIDS-related diseases, including CVD, even in the context of effective viral suppression. The impact of ART on cardiovascular risk is examined, with particular attention to the metabolic implications of specific ART regimens, which can influence lipid profiles and body composition, thereby modifying CVD risk. The therapeutic potential of statins, aspirin, and emerging treatments such as PCSK9 inhibitors in mitigating cardiovascular morbidity and mortality among PLWH is discussed, alongside considerations for their use in conjunction with ART. Our review underscores the necessity for a comprehensive, multidisciplinary approach to cardiovascular care in PLWH, which integrates vigilant cardiovascular risk assessment and management with HIV treatment. As we navigate the evolving landscape of HIV care, the goal remains to optimize treatment outcomes while minimizing cardiovascular risk, ensuring that the gains in longevity afforded by ART translate into improved overall health and quality of life for PLWH.
Collapse
Affiliation(s)
| | - Andrea Marino
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Benedetto Maurizio Celesia
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Serena Spampinato
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Carmen Giarratana
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Emmanuele Venanzi Rullo
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy; (M.S.P.R.); (S.S.); (C.G.); (E.V.R.)
| | - Bruno Cacopardo
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, ARNAS Garibaldi Hospital, University of Catania, 95123 Catania, Italy; (B.M.C.); (B.C.); (G.N.)
| |
Collapse
|
7
|
Eigner S, Kleynhans J, Beckford Vera DR, Sathekge MM, Henke KE, Ebenhan T. Visualisation of in vivo protein synthesis during mycobacterial infection through [ 68Ga]Ga-DOTA-puromycin µPET/MRI. Sci Rep 2024; 14:19250. [PMID: 39164329 PMCID: PMC11335739 DOI: 10.1038/s41598-024-70200-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Radiolabelled puromycin analogues will allow the quantification of protein synthesis through nuclear medicine-based imaging. A particularly useful application could be the non-invasive longitudinal visualisation of mycobacterial activity through direct quantification of puromycin binding. This study assesses the value of [68Ga]Ga-DOTA-puromycin in the visualisation of mycobacteria through positron emission tomography combined with magnetic resonance imaging (µPET/MRI). The radiopharmaceutical was produced by previously published and validated methods. [68Ga]Ga-DOTA-Puromycin imaging was performed on severe immunodeficient mice infected with Bacille Calmette-Guérin-derived M. Bovis (BCG). Acute and chronic infection stages were examined by µPET/MRI. A follow-up group of animals acted as controls (animals bearing S. aureus-derived infection and sterile inflammation) to assess tracer selectivity. [68Ga]Ga-DOTA-puromycin-µPET/MRI images revealed the acute, widespread infection within the right upper shoulder and armpit. Also, [68Ga]Ga-DOTA-puromycin signal sensitivity measured after a 12-week period was lower than that of [18F]FDG-PET in the same animals. A suitable correlation between normalised uptake values (NUV) and gold standard histopathological analysis confirms accurate tracer accumulation in viable bacteria. The radiopharmaceutical showed infection selectivity over inflammation but accumulated in both M. Bovis and S. Aureus, lacking pathogen specificity. Overall, [68Ga]Ga-DOTA-puromycin exhibits potential as a tool for non-invasive protein synthesis visualization, albeit without pathogen selectivity.
Collapse
Affiliation(s)
- Sebastian Eigner
- Department of Radiopharmaceuticals, Nuclear Physics Institute of the Academy of Sciences of the Czech Republic, Husinec-Rez 130, 25068, Husinec-Rez, Czech Republic
- Department of Radiopharmacy, Charles University Prague, 11000, Prague, Czech Republic
| | - Janke Kleynhans
- Nuclear Medicine Research Infrastructure NPC, Pretoria, 0001, South Africa
| | - Dennis R Beckford Vera
- Department of Radiopharmaceuticals, Nuclear Physics Institute of the Academy of Sciences of the Czech Republic, Husinec-Rez 130, 25068, Husinec-Rez, Czech Republic
| | - Mike M Sathekge
- Nuclear Medicine Research Infrastructure NPC, Pretoria, 0001, South Africa
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
| | - Katerina Eigner Henke
- Department of Radiopharmacy, Charles University Prague, 11000, Prague, Czech Republic
- Clinical for Nuclear Medicine, University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Thomas Ebenhan
- Nuclear Medicine Research Infrastructure NPC, Pretoria, 0001, South Africa.
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa.
| |
Collapse
|
8
|
Grunblatt E, Feinstein MJ. Precision Phenotyping of Heart Failure in People with HIV: Early Insights and Challenges. Curr Heart Fail Rep 2024; 21:417-427. [PMID: 38940893 DOI: 10.1007/s11897-024-00674-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE OF REVIEW People with HIV have an elevated risk of developing heart failure even with optimally controlled disease. In this review, we outline the various mechanisms through which HIV infection may directly and indirectly contribute to heart failure pathology and highlight the emerging relationship between HIV, chronic inflammation, and cardiometabolic disease. RECENT FINDINGS HIV infection leads to chronic inflammation, immune dysregulation, and metabolic imbalances even in those with well controlled disease. These dysregulations occur through several diverse mechanisms which may lead to manifestations of different phenotypes of heart failure in people with HIV. While it has long been known that people with HIV are at risk of developing heart failure, recent studies have suggested numerous complex mechanisms involving chronic inflammation, immune dysregulation, and metabolic derangement through which this may be mediated. Further comprehensive studies are needed to elucidate the precise relationship between these mechanisms and the development of different subtypes of heart failure in people with HIV.
Collapse
Affiliation(s)
- Eli Grunblatt
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA
| | - Matthew J Feinstein
- Department of Medicine, Northwestern University Feinberg School of Medicine, 300 E Superior St, Ste 12-758, Chicago, IL, 60611, USA.
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
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
|
10
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
11
|
Afolabi JM, Kirabo A. HIV and Cardiovascular Disease. Circ Res 2024; 134:1512-1514. [PMID: 38781297 PMCID: PMC11132120 DOI: 10.1161/circresaha.124.324805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Jeremiah M Afolabi
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.M.A., A.K.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.M.A., A.K.)
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.K.)
- Vanderbilt Center for Immunobiology, Nashville, TN (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| |
Collapse
|
12
|
Hudson JA, Ferrand RA, Gitau SN, Mureithi MW, Maffia P, Alam SR, Shah ASV. HIV-Associated Cardiovascular Disease Pathogenesis: An Emerging Understanding Through Imaging and Immunology. Circ Res 2024; 134:1546-1565. [PMID: 38781300 DOI: 10.1161/circresaha.124.323890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Cardiac abnormalities were identified early in the epidemic of AIDS, predating the isolation and characterization of the etiologic agent, HIV. Several decades later, the causation and pathogenesis of cardiovascular disease (CVD) linked to HIV infection continue to be the focus of intense speculation. Before the widespread use of antiretroviral therapy, HIV-associated CVD was primarily characterized by HIV-associated cardiomyopathy linked to profound immunodeficiency. With increasing antiretroviral therapy use, viral load suppression, and establishment of immune competency, the effects of HIV on the cardiovascular system are more subtle. Yet, people living with HIV still face an increased incidence of cardiovascular pathology. Advances in cardiac imaging modalities and immunology have deepened our understanding of the pathogenesis of HIV-associated CVD. This review provides an overview of the pathogenesis of HIV-associated CVD integrating data from imaging and immunologic studies with particular relevance to the HIV population originating from high-endemic regions, such as sub-Saharan Africa. The review highlights key evidence gaps in the field and suggests future directions for research to better understand the complex HIV-CVD interactions.
Collapse
Affiliation(s)
- Jonathan A Hudson
- Kings College London BHF Centre, School of Cardiovascular and Metabolic Medicine & Sciences, United Kingdom (J.A.H.)
| | - Rashida A Ferrand
- Department of Clinical Research (R.A.F.), London School of Hygiene and Tropical Medicine, United Kingdom
- Biomedical Research and Training Institute, Harare, Zimbabwe (R.A.F.)
| | - Samuel N Gitau
- Department of Radiology, Aga Khan University Nairobi, Kenya (S.N.G.)
| | - Marianne Wanjiru Mureithi
- Department of Medical Microbiology and Immunology, Faculty of Health Sciences (M.W.M.), University of Nairobi, Kenya
| | - Pasquale Maffia
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (P.M.)
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Italy (P.M.)
- Africa-Europe Cluster of Research Excellence in Non-Communicable Diseases and Multimorbidity, African Research Universities Alliance and The Guild of European Research-Intensive Universities, Glasgow, United Kingdom (P.M.)
| | - Shirjel R Alam
- Department of Cardiology, North Bristol NHS Trust, United Kingdom (S.R.A.)
| | - Anoop S V Shah
- Department of Non-Communicable Disease Epidemiology (A.S.V.S.), London School of Hygiene and Tropical Medicine, United Kingdom
- Department of Cardiology, Imperial College NHS Trust, London, United Kingdom (A.S.V.S.)
| |
Collapse
|
13
|
van der Post J, Guerra TEJ, van den Hof M, Vaz FM, Pajkrt D, van Genderen JG. Plasma Lipidomic Profiles in cART-Treated Adolescents with Perinatally Acquired HIV Compared to Matched Controls. Viruses 2024; 16:580. [PMID: 38675922 PMCID: PMC11053976 DOI: 10.3390/v16040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Children with perinatally acquired human immunodeficiency virus (PHIV) are growing into adulthood with HIV and treatment-associated comorbidities, such as dyslipidemia and insulin resistance. HIV is identified as independent risk factor for cardiovascular disease (CVD). The hypothesis behind increased CVD risk associated with HIV includes vascular inflammation, dyslipidemia and combination antiretroviral therapy (cART) metabolomic toxicity. To investigate differences in lipid profiles and pathophysiological mechanisms of CVD risk in adolescents with PHIV, we compared the plasma lipidome of PHIV adolescents and HIV-negative controls. We additionally investigated the influence of current cART regimens and increased lipoprotein(a) (Lp(a)) levels on the plasma lipidome. We included 20 PHIV-infected adolescents and 20 HIV-negative controls matched for age, sex, ethnic origin and socio-economic status. Plasma lipidome was measured using Thermo Scientific Ultimate 3000 binary high-performance liquid chromatography (HPLC)-mass spectrometry. We evaluated the plasma lipidome in PHIV adolescents using different cART regimens (including those known to be associated with lipid alterations). The median age was 17.5 years (15.5-20.7) and 16.5 years (15.7-19.8) for PHIV adolescents and controls, respectively. Of PHIV adolescents, 45% used a non-nucleotide reverse transcriptase inhibitor (NNRTI)-based (25%) or protease inhibitor (PI)-based (20%) cART regimen. In this pilot study, we observed no significant differences between lipidomic profiles between PHIV adolescents and controls. We observed no differences in the plasma lipidome in participants with increased versus normal Lp(a) levels. Different cART regimens appear to influence chain length differences in the plasma lipidome of PHIV adolescents; however, the significance and causality of this observation remains undetermined. Further research on the influence of cART on lipid composition could further identify these alterations.
Collapse
Affiliation(s)
- Julie van der Post
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Thiara E. J. Guerra
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
| | - Malon van den Hof
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health, Ageing & Later Life, Health Behaviors and Chronic Diseases, Amsterdam, The Netherlands
| | - Frédéric M. Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children’s Hospital, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC, Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
- Amsterdam Infectious Diseases and Immunology Research Institute, Amsterdam, The Netherlands
| | - Jason G. van Genderen
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Lu MT, Ribaudo H, Foldyna B, Zanni MV, Mayrhofer T, Karady J, Taron J, Fitch KV, McCallum S, Burdo TH, Paradis K, Hedgire SS, Meyersohn NM, DeFilippi C, Malvestutto CD, Sturniolo A, Diggs M, Siminski S, Bloomfield GS, Alston-Smith B, Desvigne-Nickens P, Overton ET, Currier JS, Aberg JA, Fichtenbaum CJ, Hoffmann U, Douglas PS, Grinspoon SK. Effects of Pitavastatin on Coronary Artery Disease and Inflammatory Biomarkers in HIV: Mechanistic Substudy of the REPRIEVE Randomized Clinical Trial. JAMA Cardiol 2024; 9:323-334. [PMID: 38381407 PMCID: PMC10882511 DOI: 10.1001/jamacardio.2023.5661] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/15/2023] [Indexed: 02/22/2024]
Abstract
Importance Cardiovascular disease (CVD) is increased in people with HIV (PWH) and is characterized by premature noncalcified coronary plaque. In the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), pitavastatin reduced major adverse cardiovascular events (MACE) by 35% over a median of 5.1 years. Objective To investigate the effects of pitavastatin on noncalcified coronary artery plaque by coronary computed tomography angiography (CTA) and on inflammatory biomarkers as potential mechanisms for MACE prevention. Design, Setting, and Participants This double-blind, placebo-controlled randomized clinical trial enrolled participants from April 2015 to February 2018 at 31 US clinical research sites. PWH without known CVD who were taking antiretroviral therapy and had low to moderate 10-year CVD risk were included. Data were analyzed from April to November 2023. Intervention Oral pitavastatin calcium, 4 mg per day. Main Outcomes and Measures Coronary CTA and inflammatory biomarkers at baseline and 24 months. The primary outcomes were change in noncalcified coronary plaque volume and progression of noncalcified plaque. Results Of 804 enrolled persons, 774 had at least 1 evaluable CTA. Plaque changes were assessed in 611 who completed both CT scans. Of 611 analyzed participants, 513 (84.0%) were male, the mean (SD) age was 51 (6) years, and the median (IQR) 10-year CVD risk was 4.5% (2.6-7.0). A total of 302 were included in the pitavastatin arm and 309 in the placebo arm. The mean noncalcified plaque volume decreased with pitavastatin compared with placebo (mean [SD] change, -1.7 [25.2] mm3 vs 2.6 [27.1] mm3; baseline adjusted difference, -4.3 mm3; 95% CI, -8.6 to -0.1; P = .04; 7% [95% CI, 1-12] greater reduction relative to placebo). A larger effect size was seen among the subgroup with plaque at baseline (-8.8 mm3 [95% CI, -17.9 to 0.4]). Progression of noncalcified plaque was 33% less likely with pitavastatin compared with placebo (relative risk, 0.67; 95% CI, 0.52-0.88; P = .003). Compared with placebo, the mean low-density lipoprotein cholesterol decreased with pitavastatin (mean change: pitavastatin, -28.5 mg/dL; 95% CI, -31.9 to -25.1; placebo, -0.8; 95% CI, -3.8 to 2.2). The pitavastatin arm had a reduction in both oxidized low-density lipoprotein (-29% [95% CI, -32 to -26] vs -13% [95% CI, -17 to -9]; P < .001) and lipoprotein-associated phospholipase A2 (-7% [95% CI, -11 to -4] vs 14% [95% CI, 10-18]; P < .001) compared with placebo at 24 months. Conclusions and Relevance In PWH at low to moderate CVD risk, 24 months of pitavastatin reduced noncalcified plaque volume and progression as well as markers of lipid oxidation and arterial inflammation. These changes may contribute to the observed MACE reduction in REPRIEVE. Trial Registration ClinicalTrials.gov Identifier: NCT02344290.
Collapse
Affiliation(s)
- Michael T. Lu
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Julia Karady
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Jana Taron
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Tricia H. Burdo
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Kayla Paradis
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sandeep S. Hedgire
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Nandini M. Meyersohn
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | | | - Audra Sturniolo
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Marissa Diggs
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | - Gerald S. Bloomfield
- Department of Medicine, Duke Global Health Institute, Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Beverly Alston-Smith
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patrice Desvigne-Nickens
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Edgar T. Overton
- Division of Infectious Diseases, University of Alabama at Birmingham
- ViiV Healthcare, Research Triangle Park, North Carolina
| | - Judith S. Currier
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles
| | - Judith A. Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Pamela S. Douglas
- Duke University Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
15
|
Filip I. Aldosterone-blocking agents may reduce arterial inflammation in people with HIV. AIDS 2024; 38:N9-N10. [PMID: 38363983 DOI: 10.1097/qad.0000000000003867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Affiliation(s)
- Iulia Filip
- MedEd Medical Communications, LLC, Bluffton, SC, USA
| |
Collapse
|
16
|
Cohen MH, Benekigeri C, Anastos K. Bringing together the pieces: the need for holistic care for women with HIV. J Int AIDS Soc 2024; 27:e26228. [PMID: 38450984 PMCID: PMC10935697 DOI: 10.1002/jia2.26228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Affiliation(s)
| | | | - Kathryn Anastos
- Departments of Medicine and Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
17
|
Obeagu EI, Obeagu GU. Utilization of immunological ratios in HIV: Implications for monitoring and therapeutic strategies. Medicine (Baltimore) 2024; 103:e37354. [PMID: 38428854 PMCID: PMC10906605 DOI: 10.1097/md.0000000000037354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection remains a significant global health concern, necessitating ongoing research and innovation in the quest for improved disease management. Traditional markers for monitoring HIV progression and the effectiveness of antiretroviral therapy have limitations in capturing the intricate immune responses and inflammatory dynamics in people with HIV. In recent years, the concept of inflammation ratios has gained prominence as a valuable tool for assessing and understanding the complex interplay between inflammation, immune function, and HIV. In this abstract, we provide an overview of the emerging field of utilizing inflammation ratios in the context of HIV and its implications for disease monitoring and therapeutic strategies. These ratios, such as the CD4/CD8 ratio, neutrophil-to-lymphocyte ratio, and monocyte-to-lymphocyte ratio, offer a more comprehensive assessment of an individual's immune status and inflammatory state. By exploring the clinical implications of inflammation ratios, including their potential to predict disease complications and guide personalized treatment approaches, this publication sheds light on the potential benefits of incorporating inflammation ratios into routine HIV care. Furthermore, we emphasize the importance of ongoing research in this field to further refine our understanding of the utility and significance of inflammation ratios in improving the lives of people with HIV.
Collapse
|
18
|
Srinivasa S, Abohashem S, Walpert AR, Dunderdale CN, Iyengar S, Shen G, Jerosch-Herold M, deFilippi CR, Robbins GK, Lee H, Kwong RY, Adler GK, Tawakol A, Grinspoon SK. Mineralocorticoid Receptor Antagonism by Eplerenone and Arterial Inflammation in HIV: The MIRABELLA HIV Study. JAMA Cardiol 2024; 9:189-194. [PMID: 38090987 PMCID: PMC10719827 DOI: 10.1001/jamacardio.2023.4578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/19/2023] [Indexed: 12/17/2023]
Abstract
Importance The risk for atherosclerotic disease is increased 1.5- to 2.0-fold among persons with HIV (PWH). Increased activation of the renin-angiotensin-aldosterone system may contribute to increased arterial inflammation in this population. Objective To determine the effects of eplerenone on arterial inflammation among well-treated PWH without known cardiovascular disease (CVD). Design, Setting, and Participants Well-treated PWH who participated in the double-blinded, placebo-controlled, Mineralocorticoid Receptor Antagonism for Cardiovascular Health in HIV (MIRACLE HIV) study between February 2017 and March 2022 assessing the effects of eplerenone on myocardial perfusion were invited to participate in the Mineralocorticoid Receptor Antagonism By Eplerenone to Lower Arterial Inflammation in HIV (MIRABELLA) substudy if there was no current statin use. Participants were enrolled in the MIRABELLA study and underwent additional 18F-fludeoxyglucose-positron emission tomography/computed tomography (18F-FDG PET/CT) imaging of the aorta and carotid arteries to assess arterial inflammation over 12 months of treatment with eplerenone vs placebo. Interventions Eplerenone, 50 mg, twice a day vs identical placebo. Main Outcomes and Measures The primary outcome was change in target to background ratio (TBR), a measure of arterial wall inflammation, in the index vessel after 12 months of treatment. The index vessel was defined as the vessel (aorta, left carotid artery, or right carotid artery) with the highest TBR at baseline in each participant. Results A total of 26 participants (mean [SD] age, 54 [7] years; 18 male [69%]) were enrolled in the study. Treatment groups (eplerenone, 13 vs placebo, 13) were of similar age, sex, and body mass index. Eplerenone was associated with a reduction in TBR of the primary end point, the index vessel (eplerenone vs placebo: model treatment effect, -0.31; 95% CI, -0.50 to -0.11; P = .006; percentage change, -12.4% [IQR, -21.9% to -2.6%] vs 5.1% [IQR, -1.6% to 11.0%]; P = .003). We further observed a significant reduction of the TBR of the most diseased segment (MDS) of the index vessel (eplerenone vs placebo: -19.1% [IQR, -27.0% to -11.9%] vs 6.8% [IQR, -9.1% to 12.1%]; P = .007). A similar result was seen assessing the index vessel of the carotids (eplerenone vs placebo: -10.0% [IQR, -21.8% to 3.6%] vs 9.7% [IQR, -9.8% to 15.9%]; P = .046). Reduction in the TBR of MDS of the index vessel on 18F-FDG PET/CT correlated with improvement in the stress myocardial blood flow on cardiac magnetic resonance imaging (Spearman ρ = -0.67; P = .01). Conclusion and Relevance In this small randomized clinical trial, eplerenone was associated with reduction in arterial inflammation among well-treated PWH without known CVD. In addition, reductions in arterial inflammation as measured by 18F-FDG PET/CT were related to improvements in stress myocardial perfusion. Further larger studies should explore whether eplerenone is a potential treatment strategy for inflammatory-mediated CVD in PWH. Trial Registration ClinicalTrials.gov Identifier: NCT02740179.
Collapse
Affiliation(s)
- Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Shady Abohashem
- Cardiovascular Imaging Research Center and Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Allie R. Walpert
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | | | - Sanjna Iyengar
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Grace Shen
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michael Jerosch-Herold
- Cardiovascular Division of Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Gregory K. Robbins
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond Y. Kwong
- Cardiovascular Division of Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gail K. Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center and Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
19
|
Mu W, Patankar V, Kitchen S, Zhen A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024; 16:219. [PMID: 38399994 PMCID: PMC10893210 DOI: 10.3390/v16020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic Human Immunodeficiency Virus (HIV) infection remains a significant challenge to global public health. Despite advances in antiretroviral therapy (ART), which has transformed HIV infection from a fatal disease into a manageable chronic condition, a definitive cure remains elusive. One of the key features of HIV infection is chronic immune activation and inflammation, which are strongly associated with, and predictive of, HIV disease progression, even in patients successfully treated with suppressive ART. Chronic inflammation is characterized by persistent inflammation, immune cell metabolic dysregulation, and cellular exhaustion and dysfunction. This review aims to summarize current knowledge of the interplay between chronic inflammation, immune metabolism, and T cell dysfunction in HIV infection, and also discusses the use of humanized mice models to study HIV immune pathogenesis and develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Vaibhavi Patankar
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Scott Kitchen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anjie Zhen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Foldyna B, Mayrhofer T, Zanni MV, Lyass A, Barve R, Karady J, McCallum S, Burdo TH, Fitch KV, Paradis K, Fulda ES, Diggs MR, Bloomfield GS, Malvestutto CD, Fichtenbaum CJ, Aberg JA, Currier JS, Ribaudo HJ, Hoffmann U, Lu MT, Douglas PS, Grinspoon SK. Pericoronary Adipose Tissue Density, Inflammation, and Subclinical Coronary Artery Disease Among People With HIV in the REPRIEVE Cohort. Clin Infect Dis 2023; 77:1676-1686. [PMID: 37439633 PMCID: PMC10724469 DOI: 10.1093/cid/ciad419] [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: 04/06/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Pericoronary adipose tissue (PCAT) may influence plaque development through inflammatory mechanisms. We assessed PCAT density, as a measure of pericoronary inflammation, in relationship to coronary plaque among people with human immunodeficiency virus (HIV [PWH]) and to a matched control population. METHODS In this baseline analysis of 727 participants of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) Mechanistic Substudy, we related computed tomography-derived PCAT density to presence and extent (Leaman score) of coronary artery disease (CAD), noncalcified plaque, coronary artery calcium (CAC), and vulnerable plaque features using multivariable logistic regression analyses. We further compared the PCAT density between PWH and age, sex, body mass index, CAC score, and statin use-matched controls from the community-based Framingham Heart Study (N = 464), adjusting for relevant clinical covariates. RESULTS Among 727 REPRIEVE participants (age 50.8 ± 5.8 years; 83.6% [608/727] male), PCAT density was higher in those with (vs without) coronary plaque, noncalcified plaque, CAC >0, vulnerable plaque, and high CAD burden (Leaman score >5) (P < .001 for each comparison). PCAT density related to prevalent coronary plaque (adjusted odds ratio [per 10 HU]: 1.44; 95% confidence interval, 1.22-1.70; P < .001), adjusted for clinical cardiovascular risk factors, body mass index, and systemic immune/inflammatory biomarkers. Similarly, PCAT density related to CAC >0, noncalcified plaque, vulnerable plaque, and Leaman score >5 (all P ≤ .002). PCAT density was greater among REPRIEVE participants versus Framingham Heart Study (-88.2 ± 0.5 HU versus -90.6 ± 0.4 HU; P < .001). CONCLUSIONS Among PWH in REPRIEVE, a large primary cardiovascular disease prevention cohort, increased PCAT density independently associated with prevalence and severity of coronary plaque, linking increased coronary inflammation to CAD in PWH.
Collapse
Affiliation(s)
- Borek Foldyna
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Mayrhofer
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Health Economics, School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Asya Lyass
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Radhika Barve
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julia Karady
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tricia H Burdo
- Department of Microbiology, Immunology, and Inflammation and Center for NeuroVirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kayla Paradis
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Evelynne S Fulda
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marissa R Diggs
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald S Bloomfield
- Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Carlos D Malvestutto
- Division of Infectious Diseases, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith S Currier
- Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Innovative Imaging Consulting LLC, Waltham, Massachusetts, USA
| | - Michael T Lu
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Pamela S Douglas
- Department of Medicine (Cardiology), Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Alles M, Gunasena M, Kettelhut A, Ailstock K, Musiime V, Kityo C, Richardson B, Mulhern W, Tamilselvan B, Rubsamen M, Kasturiratna D, Demberg T, Cameron CM, Cameron MJ, Dirajlal-Fargo S, Funderburg NT, Liyanage NPM. Activated NK Cells with Pro-inflammatory Features are Associated with Atherogenesis in Perinatally HIV-Acquired Adolescents. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.06.23297580. [PMID: 37986784 PMCID: PMC10659511 DOI: 10.1101/2023.11.06.23297580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Human immunodeficiency virus (HIV) is associated with persistent immune activation and dysfunction in people with HIV despite treatment with antiretroviral therapy (ART). Modulation of the immune system may be driven by: low-level HIV replication, co-pathogens, gut dysbiosis /translocation, altered lipid profiles, and ART toxicities. In addition, perinatally acquired HIV (PHIV) and lifelong ART may alter the development and function of the immune system. Our preliminary data and published literature suggest reprogramming innate immune cells may accelerate aging and increase the risk for future end-organ complications, including cardiovascular disease (CVD). The exact mechanisms, however, are currently unknown. Natural killer (NK) cells are a highly heterogeneous cell population with divergent functions. They play a critical role in HIV transmission and disease progression in adults. Recent studies suggest the important role of NK cells in CVDs; however, little is known about NK cells and their role in HIV-associated cardiovascular risk in PHIV adolescents. Here, we investigated NK cell subsets and their potential role in atherogenesis in PHIV adolescents compared to HIV-negative adolescents in Uganda. Our data suggest, for the first time, that activated NK subsets in PHIV adolescents may contribute to atherogenesis by promoting plasma oxidized low-density lipoprotein (Ox-LDL) uptake by vascular macrophages.
Collapse
|
24
|
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
|
25
|
Blaauw MJT, Berrevoets MAH, Vos WAJW, Groenendijk AL, van Eekeren LE, Vadaq N, Weijers G, van der Ven AJAM, Rutten JHW, Riksen NP. Traditional Cardiovascular Risk Factors Are Stronger Related to Carotid Intima-Media Thickness Than to Presence of Carotid Plaques in People Living With HIV. J Am Heart Assoc 2023; 12:e030606. [PMID: 37804189 PMCID: PMC10757550 DOI: 10.1161/jaha.123.030606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/08/2023] [Indexed: 10/09/2023]
Abstract
Background Cardiovascular disease is a major cause of morbidity and mortality in people living with HIV, who are at higher risk than the general population. We assessed, in a large cohort of people living with HIV, which cardiovascular, HIV-specific, and lipoproteomic markers were associated with carotid intima-media thickness (cIMT) and carotid plaque presence. We also studied guideline adherence on lipid-lowering medication in individuals with high and very high risk for cardiovascular disease. Methods and Results In 1814 individuals with a median (interquartile range) age of 53 (44-60) years, we found a carotid plaque in 909 (50.1%) and a median (interquartile range) intima-media thickness of 0.66 (0.57-0.76) mm. Ultrasonography was used for the assessment of cIMT and plaque presence. Univariable and multivariable regression models were used for associations with cIMT and presence of plaques. Age, Black race, body mass index, type 2 diabetes, and smoking (pack years) were all positively associated with higher cIMT. Levels of high-density lipoprotein cholesterol, specifically medium and large high-density lipoprotein subclasses, were negatively associated with higher cIMT. Only age and prior myocardial infarction were positively related to the presence of a carotid plaque. Lipid-lowering treatment was prescribed in one-third of people living with HIV, who are at high and very high risk for cardiovascular disease. Conclusions Traditional cardiovascular risk factors were significantly associated with higher cIMT but not with carotid plaques, except for age. HIV-specific factors were not associated with both ultrasound measurements. Future studies are needed to elucidate which factors contribute to plaque formation. Improvement of guideline adherence on prescription of lipid-lowering treatment in high- and very high-risk patients for cardiovascular disease is recommended. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT03994835.
Collapse
Affiliation(s)
- Marc J. T. Blaauw
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
- Department of Internal MedicineElisabeth‐Tweesteden HospitalTilburgthe Netherlands
| | | | - Wilhelm A. J. W. Vos
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
- Department of Internal MedicineOLVGAmsterdamthe Netherlands
| | - Albert L. Groenendijk
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
- Department of Medical Microbiology and Infectious DiseasesErasmus Medical Center (MC)Rotterdamthe Netherlands
| | - Louise E. van Eekeren
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
| | - Nadira Vadaq
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
- Center for Tropical and Infectious Diseases, Faculty of MedicineDiponegoro University, Dr. Kariadi HospitalSemarangIndonesia
| | - Gert Weijers
- Medical UltraSound Imaging Center, Division of Medical ImagingRadboud University Medical CenterNijmegenthe Netherlands
| | - Andre J. A. M. van der Ven
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenthe Netherlands
| | - Joost H. W. Rutten
- Division of Vascular Medicine, Department of Internal MedicineRadboud University Medical CentreNijmegenthe Netherlands
| | - Niels P. Riksen
- Division of Vascular Medicine, Department of Internal MedicineRadboud University Medical CentreNijmegenthe Netherlands
| |
Collapse
|
26
|
Lau CY, Martinez-Orengo N, Lyndaker A, Flavahan K, Johnson RF, Shah S, Hammoud DA. Advances and Challenges in Molecular Imaging of Viral Infections. J Infect Dis 2023; 228:S270-S280. [PMID: 37788495 PMCID: PMC10547465 DOI: 10.1093/infdis/jiad247] [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: 10/05/2023] Open
Abstract
Molecular imaging of viral infection, using a variety of advanced imaging techniques such as optical and nuclear imaging, can and has been used for direct visualization of the virus as well as assessment of virus-host interactions. Unlike imaging of other pathogens such as bacteria and fungi, challenging aspects of imaging viral infections include the small size of viruses, the complexity of viral infection animal models (eg, species dependence), and the high-level containment needs for many high-consequence pathogens, among others. In this review, using representative viral infections, we discuss how molecular imaging can reveal real-time infection dynamics, improve our understanding of disease pathogenesis, and guide optimization of treatment and prevention strategies. Key findings from human and animal studies are highlighted.
Collapse
Affiliation(s)
- Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Neysha Martinez-Orengo
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Lyndaker
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Swati Shah
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Dima A Hammoud
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Graham SM, Nance RM, Chen J, Wurfel MM, Hunt PW, Heckbert SR, Budoff MJ, Moore RD, Jacobson JM, Martin JN, Crane HM, López JA, Liles WC. Plasma Interleukin-6 (IL-6), Angiopoietin-2, and C-Reactive Protein Levels Predict Subsequent Type 1 Myocardial Infarction in Persons With Treated HIV Infection. J Acquir Immune Defic Syndr 2023; 93:282-291. [PMID: 37018921 PMCID: PMC10330055 DOI: 10.1097/qai.0000000000003207] [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: 08/14/2022] [Accepted: 03/16/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND HIV infection leads to endothelial activation, promoting platelet adhesion, and accelerating atherosclerosis. Our goal was to determine whether biomarkers of endothelial activation and hemostasis/thrombosis were elevated in people with treated HIV (PWH) before myocardial infarction (MI). METHODS In a case-control study nested within the CFAR Network of Integrated Clinical Systems (CNICS) cohort, we compared 69 adjudicated cases with type 1 MI with 138 controls matched for antiretroviral therapy regimen. We measured angiopoietin-1, angiopoietin-2 (ANG-2), intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), von Willebrand factor, C-reactive protein (CRP), interleukin-6 (IL-6), plasminogen activation inhibitor-1, P-selectin, serum amyloid-A, soluble CD14, and apolipoprotein A1 in stored plasma. Conditional logistic regression identified associations with subsequent MI, with and without adjustment for Atherosclerotic Cardiovascular Disease (ASCVD) and Veterans Aging Cohort Study (VACS) scores. RESULTS Higher IL-6 was associated with MI after adjustment for ASCVD score (adjusted odds ratio [AOR] 1.51, 95% confidence interval [95% CI]: 1.05 to 2.17 per standard-deviation-scaled log 2 increment). In a separate model adjusting for VACS score, higher ANG-2 (AOR 1.49, 95% CI: 1.04 to 2.14), higher CRP (AOR 1.45, 95% CI: 1.06 to 2.00), and higher IL-6 (AOR 1.68, 95% CI: 1.17 to 2.41) were associated with MI. In a sensitivity analysis excluding PWH with viral load ≥400 copies/mL, higher IL-6 remained associated with MI after adjustment for ASCVD score and after adjustment for VACS score. CONCLUSIONS Among PWH, higher levels of plasma IL-6, CRP, and ANG-2 predict subsequent type 1 MI, independent of conventional risk scores. IL-6 had the most consistent associations with type 1 MI, regardless of viral load suppression.
Collapse
Affiliation(s)
- Susan M. Graham
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Robin M. Nance
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Junmei Chen
- Bloodworks Northwest Research Institute, Seattle, WA, USA
| | - Mark M. Wurfel
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Peter W. Hunt
- Department of Medicine, University of California at San Francisco, San Francisco, USA
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Matthew J. Budoff
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | | | | | - Jeffrey N. Martin
- Departments of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA USA
| | - Heidi M. Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - José A. López
- Bloodworks Northwest Research Institute, Seattle, WA, USA
| | - W. Conrad Liles
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| |
Collapse
|
28
|
Kalra DK, Vorla M, Michos ED, Agarwala A, Virani S, Duell PB, Raal FJ. Dyslipidemia in Human Immunodeficiency Virus Disease: JACC Review Topic of the Week. J Am Coll Cardiol 2023; 82:171-181. [PMID: 37407116 DOI: 10.1016/j.jacc.2023.04.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 07/07/2023]
Abstract
The advent of newer and better tolerated antiretroviral therapy has progressively shortened the life expectancy gap between people living with HIV (PWH) and the general population. However, in this aging cohort, cardiovascular disease is now a significant cause of morbidity and mortality despite advances in cardiac care. Therefore, it is critical to assess and treat all cardiovascular disease risk factors, including dyslipidemia, early and aggressively in PWH. Data are not as robust regarding the pathogenesis and management of dyslipidemia in PWH, with most evidence being extrapolated from the general uninfected population. In this review the authors describe the current understanding of the pathophysiology of HIV and antiretroviral therapy-induced dyslipidemia, and the approach to risk assessment and management, given that drug-drug interactions remain an important consideration in this population.
Collapse
Affiliation(s)
- Dinesh K Kalra
- Division of Cardiology, University of Louisville, Louisville, Kentucky, USA.
| | - Mounica Vorla
- Division of Cardiology, University of Louisville, Louisville, Kentucky, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anandita Agarwala
- Center for Cardiovascular Disease Prevention, Baylor Scott & White Health, Plano, Texas, USA
| | - Salim Virani
- Section of Cardiovascular Research, Baylor College of Medicine and Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - P Bart Duell
- Division of Cardiovascular Medicine, Oregon Health Sciences University, Portland, Oregon, USA
| | - Frederick J Raal
- Division of Endocrinology & Metabolism, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
29
|
Knudsen AD, Fuchs A, Benfield T, Gerstoft J, Køber L, Trøseid M, Kofoed KF, Nielsen SD. Coronary Artery Disease in Persons With Human Immunodeficiency Virus Without Detectable Viral Replication. Open Forum Infect Dis 2023; 10:ofad298. [PMID: 37441354 PMCID: PMC10334377 DOI: 10.1093/ofid/ofad298] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Indexed: 07/15/2023] Open
Abstract
Background We aimed to determine the prevalence of coronary artery disease (CAD) in persons with human immunodeficiency virus (HIV; PWH) and investigate whether inflammatory markers, including interleukin 6, IL-1β, and high-sensitivity C-reactive protein (hsCRP), were associated with CAD. Methods From the Copenhagen Comorbidity in HIV Infection (COCOMO) study, we included virologically suppressed PWH who underwent coronary computed tomographic (CT) angiography. Any atherosclerosis was defined as >0% stenosis, and obstructive CAD as ≥50% stenosis. Results Among 669 participants (mean age [standard deviation], 51 [11] years; 89% male), 300 (45%) had atherosclerosis, and 119 (18%) had obstructive CAD. The following risk factors were associated with any atherosclerosis and with obstructive CAD: age, male sex, hypertension, diabetes, smoking, dyslipidemia, time with HIV, and current protease inhibitor use. Interleukin 6 (IL-6) and hsCRP levels >2 mg/L were associated with any atherosclerosis and with obstructive CAD in univariable analyses but not after adjustment for traditional risk factors. IL-1β was not associated with CAD. Conclusions In a large population of PWH without viral replication, almost half had angiographically verified atherosclerosis. High concentrations of IL-6 and hsCRP were associated with CAD in univariable analyses, but adjustment for cardiovascular risk factors attenuated the association, suggesting that inflammation may mediate the association between traditional risk factors and CAD.
Collapse
Affiliation(s)
- Andreas D Knudsen
- Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Fuchs
- Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre
| | - Jan Gerstoft
- Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marius Trøseid
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Klaus F Kofoed
- Department of Cardiology, The Heart Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Radiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne D Nielsen
- Correspondence: Susanne D. Nielsen, MD, DMSc, Viro-immunology Research Unit, Department of Infectious Diseases 8632, Copenhagen University Hospital, Blegdamsvej 9B, DK-2100 Copenhagen, Denmark ()
| |
Collapse
|
30
|
Foldyna B, Mayrhofer T, Lu MT, Karády J, Kolossváry M, Ferencik M, Shah SH, Pagidipati NJ, Douglas PS, Hoffmann U. Prognostic value of CT-derived coronary artery disease characteristics varies by ASCVD risk: insights from the PROMISE trial. Eur Radiol 2023; 33:4657-4667. [PMID: 36719496 PMCID: PMC10765563 DOI: 10.1007/s00330-023-09430-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/15/2022] [Accepted: 01/07/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To compare the prognostic value of individual CT-derived coronary artery disease (CAD) characteristics across categories of clinical cardiovascular risk. METHODS The central core laboratory assessed coronary artery calcium (CAC), obstructive CAD (stenosis ≥ 50%), and high-risk plaque (HRP) in stable outpatients with suspected CAD enrolled in the PROMISE trial. Multivariable Cox regression models (endpoint: unstable angina, nonfatal myocardial infarction, or all-cause mortality; median follow-up: 2 years) were used to compare hazard ratios (HR) of the CT measures between low-borderline (< 7.5%) and moderate-high (≥ 7.5%) atherosclerotic cardiovascular disease (ASCVD) risk based on the pooled cohort equation. RESULTS Among 4356 included patients (aged 61 ± 8 years, 52% women), 67% had ASCVD risk ≥ 7.5%. Stratified by ASCVD risk, CAD ≥ 50% had nearly threefold greater HR in individuals with ASCVD < 7.5% (aHR, 6.85; 95% CI, 2.33-20.15; p < 0.001) vs. ASCVD ≥ 7.5% (aHR: 2.66, 95% CI: 1.67-4.25, p < 0.001; interaction p = 0.041). CAC predicted events solely in ASCVD ≥ 7.5% patients (aHR: 1.92, 95% CI: 1.01-3.63, p = 0.045; interaction p = 0.571), while HRP predicted events only in ASCVD < 7.5% (aHR: 3.11, 95% CI: 1.09-8.85, p = 0.034; interaction p = 0.034). CONCLUSIONS Prognostic values of CT-derived CAD characteristics differ by ASCVD risk categories. While CAD ≥ 50% has the highest prognostic value regardless of ASCVD risk, CAC is prognostic in high and HRP in low ASCVD risk. These findings suggest that CAD ≥ 50% and HRP detection rather than CAC scoring may better risk-stratify symptomatic low-risk patients and thus potentially improve downstream care. KEY POINTS • Prognostic value of individual CT-derived CAD characteristics differs by categories of cardiovascular risk. • Presence of obstructive coronary artery stenosis ≥ 50% has the highest prognostic value regardless of cardiovascular risk. • Coronary artery calcium is independently prognostic in high and high-risk plaque features in low cardiovascular risk.
Collapse
Affiliation(s)
- Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 400, Boston, MA, 02114, USA.
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 400, Boston, MA, 02114, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 400, Boston, MA, 02114, USA
| | - Júlia Karády
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 400, Boston, MA, 02114, USA
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Márton Kolossváry
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street, Suite 400, Boston, MA, 02114, USA
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Svati H Shah
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Neha J Pagidipati
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Udo Hoffmann
- Innovative Imaging Consulting LLC, 163 Longfellow Rd, Waltham, MA, 02453, USA.
| |
Collapse
|
31
|
Wang S, Singh M, Yang H, Morrell CN, Mohamad LA, Xu JJ, Nguyen T, Ture S, Tyrell A, Maggirwar SB, Schifitto G, Pang J. Monocyte-derived Dll4 is a novel contributor to persistent systemic inflammation in HIV patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537330. [PMID: 37131726 PMCID: PMC10153122 DOI: 10.1101/2023.04.18.537330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background In people living with HIV (PLWH) on combination antiretroviral therapy (cART), persistent systemic inflammation is a driving force for the progression of comorbidities, such as cardiovascular and cerebrovascular diseases. In this context, monocyte- and macrophage-related inflammation rather than T cell activation is a major cause of chronic inflammation. However, the underlying mechanism of how monocytes cause persistent systemic inflammation in PLWH is elusive. Methods and Results In vitro, we demonstrated that lipopolysaccharides (LPS) or tumor necrosis factor alpha (TNFα), induced a robust increase of Delta-like ligand 4 (Dll4) mRNA and protein expression in human monocytes and Dll4 secretion (extracellular Dll4, exDll4) from monocytes. Enhanced membrane-bound Dll4 (mDll4) expression in monocytes triggered Notch1 activation to promote pro-inflammatory factors expression. Dll4 silencing and inhibition of Nocth1 activation diminished the LPS or TNFα -induced inflammation. exDll4 releases in response to cytokines occurred in monocytes but not endothelial cells or T cells. In clinical specimens, we found that PLWH, both male and female, on cART, showed a significant increase in mDll4 expression, activation of Dll4-Notch1 signaling, and inflammatory markers in monocytes. Although there was no sex effect on mDII4 in PLWH, plasma exDll4 was significantly elevated in males but not females compared to HIV uninfected individuals. Furthermore, exDll4 plasma levels paralleled with monocytes mDll4 in male PLWH. Circulating exDll4 was also positively associated with pro-inflammatory monocytes phenotype and negatively associated with classic monocytes phenotype in male PLWH. Conclusion Pro-inflammatory stimuli increase Dll4 expression and Dll4-Notch1 signaling activation in monocytes and enhance monocyte proinflammatory phenotype, contributing to persistent systemic inflammation in male and female PLWH. Therefore, monocyte mDll4 could be a potential biomarker and therapeutic target of systemic inflammation. Plasma exDll4 may also play an additional role in systemic inflammation but primarily in men.
Collapse
|
32
|
Etemad B, Sun X, Li Y, Melberg M, Moisi D, Gottlieb R, Ahmed H, Aga E, Bosch RJ, Acosta EP, Yuki Y, Martin MP, Carrington M, Gandhi RT, Jacobson JM, Volberding P, Connick E, Mitsuyasu R, Frank I, Saag M, Eron JJ, Skiest D, Margolis DM, Havlir D, Schooley RT, Lederman MM, Yu XG, Li JZ. HIV post-treatment controllers have distinct immunological and virological features. Proc Natl Acad Sci U S A 2023; 120:e2218960120. [PMID: 36877848 PMCID: PMC10089217 DOI: 10.1073/pnas.2218960120] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023] Open
Abstract
HIV post-treatment controllers (PTCs) are rare individuals who maintain low levels of viremia after stopping antiretroviral therapy (ART). Understanding the mechanisms of HIV post-treatment control will inform development of strategies aiming at achieving HIV functional cure. In this study, we evaluated 22 PTCs from 8 AIDS Clinical Trials Group (ACTG) analytical treatment interruption (ATI) studies who maintained viral loads ≤400 copies/mL for ≥24 wk. There were no significant differences in demographics or frequency of protective and susceptible human leukocyte antigen (HLA) alleles between PTCs and post-treatment noncontrollers (NCs, n = 37). Unlike NCs, PTCs demonstrated a stable HIV reservoir measured by cell-associated RNA (CA-RNA) and intact proviral DNA assay (IPDA) during analytical treatment interruption (ATI). Immunologically, PTCs demonstrated significantly lower CD4+ and CD8+ T cell activation, lower CD4+ T cell exhaustion, and more robust Gag-specific CD4+ T cell responses and natural killer (NK) cell responses. Sparse partial least squares discriminant analysis (sPLS-DA) identified a set of features enriched in PTCs, including a higher CD4+ T cell% and CD4+/CD8+ ratio, more functional NK cells, and a lower CD4+ T cell exhaustion level. These results provide insights into the key viral reservoir features and immunological profiles for HIV PTCs and have implications for future studies evaluating interventions to achieve an HIV functional cure.
Collapse
Affiliation(s)
- Behzad Etemad
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Xiaoming Sun
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
| | - Yijia Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Meghan Melberg
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Daniela Moisi
- School of Medicine, Case Western Reserve University, Cleveland, OH44106
| | - Rachel Gottlieb
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Hayat Ahmed
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Evgenia Aga
- Harvard T. H. Chan School of Public Health, Boston, MA02115
| | | | - Edward P. Acosta
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL35233
| | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Maureen P. Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD21702
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD20814
| | - Rajesh T. Gandhi
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | | | - Paul Volberding
- School of Medicine, University of California San Francisco, San Francisco, CA94143
| | | | - Ronald Mitsuyasu
- School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| | - Ian Frank
- School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michael Saag
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL35233
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Daniel Skiest
- Department of Medicine, University of Massachusetts Chan Medical School - Baystate, Springfield, MA01199
| | - David M. Margolis
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Diane Havlir
- School of Medicine, University of California San Francisco, San Francisco, CA94143
| | - Robert T. Schooley
- Department of Medicine, University of California San Diego, San Diego, CA92103
| | | | - Xu G. Yu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
| | - Jonathan Z. Li
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| |
Collapse
|
33
|
Fichtenbaum CJ, Mallon P. Are we ready to RESPOND that the question of abacavir's role in cardiovascular disease events is settled? AIDS 2023; 37:541-543. [PMID: 36695364 DOI: 10.1097/qad.0000000000003448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Patrick Mallon
- Department of Microbial Diseases, Centre for Experimental Pathogen Host Research, University College Dublin, School of Medicine, Belfield Dublin, Ireland
| |
Collapse
|
34
|
Hileman CO, Durieux JC, Janus SE, Bowman E, Kettelhut A, Nguyen TT, Avery AK, Funderburg N, Sullivan C, McComsey GA. Heroin Use Is Associated With Vascular Inflammation in Human Immunodeficiency Virus. Clin Infect Dis 2023; 76:375-381. [PMID: 36208157 PMCID: PMC10169434 DOI: 10.1093/cid/ciac812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Heroin use may work synergistically with human immunodeficiency virus (HIV) infection to cause greater immune dysregulation than either factor alone. Unraveling how this affects end-organ disease is key as it may play a role in the excess mortality seen in people with HIV (PWH) who use heroin despite access to care and antiretroviral therapy. METHODS This is a prospectively enrolled, cross-sectional study of adults with and without HIV who use and do not use heroin using (18)F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) to compare tissue-specific inflammation including aortic (target-to-background ratio [TBR]), splenic, and bone marrow (standardized uptake value [SUV]). RESULTS A total of 120 participants were enrolled. The unadjusted mean difference in aortic TBR was 0.43 between HIV-positive [HIV+] heroin+ and HIV+ heroin-negative [heroin-] (P = .02); however, among HIV-, aortic TBR was similar regardless of heroin-use status. Further, HIV-by-heroin-use status interaction was significant (P = .02), indicating that the relationship between heroin use and higher aortic TBR depended on HIV status. On the other hand, both HIV (1.54 vs 1.68; P = .04, unadjusted estimated means for HIV+ vs HIV-) and heroin use were associated with lower bone marrow SUV, although the effect of heroin depended on sex (heroin-use-by-sex interaction, P = .03). HIV-by-heroin-use interaction was not significant for splenic or bone marrow SUV. CONCLUSIONS Aortic inflammation was greatest in PWH who use heroin, but paradoxically, bone marrow activity was the least in this group, suggesting complex and possibly divergent pathophysiology within these different end organs.
Collapse
Affiliation(s)
- Corrilynn O Hileman
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center, Cleveland, Ohio, USA
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jared C Durieux
- Department of Medicine and Pediatrics, Division of Infectious Disease, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Scott E Janus
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Emily Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, The Ohio State University, Columbus, Ohio, USA
| | - Aaren Kettelhut
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, The Ohio State University, Columbus, Ohio, USA
| | - Trong-Tuong Nguyen
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center, Cleveland, Ohio, USA
| | - Ann K Avery
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center, Cleveland, Ohio, USA
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicholas Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, The Ohio State University, Columbus, Ohio, USA
| | - Claire Sullivan
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Grace A McComsey
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Medicine and Pediatrics, Division of Infectious Disease, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Perkins MV, Joseph S, Dittmer DP, Mackman N. Cardiovascular Disease and Thrombosis in HIV Infection. Arterioscler Thromb Vasc Biol 2023; 43:175-191. [PMID: 36453273 PMCID: PMC10165851 DOI: 10.1161/atvbaha.122.318232] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
HIV infection has transitioned from an acute, fatal disease to a chronic one managed by antiretroviral therapy. Thus, the aging population of people living with HIV (PLWH) continues to expand. HIV infection results in a dysregulated immune system, wherein CD4+ T cells are depleted, particularly in the gastrointestinal tract, disrupting the gut epithelial barrier. Long-term HIV infection is associated with chronic inflammation through potentially direct mechanisms caused by viral replication or exposure to viral proteins and indirect mechanisms resulting from increased translocation of microbial products from the intestine or exposure to antiretroviral therapy. Chronic inflammation (as marked by IL [interleukin]-6 and CRP [C-reactive protein]) in PLWH promotes endothelial cell dysfunction and atherosclerosis. PLWH show significantly increased rates of cardiovascular disease, such as myocardial infarction (risk ratio, 1.79 [95% CI, 1.54-2.08]) and stroke (risk ratio, 2.56 [95% CI, 1.43-4.61]). In addition, PLWH have increased levels of the coagulation biomarker D-dimer and have a two to ten-fold increased risk of venous thromboembolism compared with the general population. Several small clinical trials analyzed the effect of different antithrombotic agents on platelet activation, coagulation, inflammation, and immune cell activation. Although some markers for coagulation were reduced, most agents failed to reduce inflammatory markers in PLWH. More studies are needed to understand the underlying mechanisms driving inflammation in PLWH to create better therapies for lowering chronic inflammation in PLWH. Such therapies can potentially reduce atherosclerosis, cardiovascular disease, and thrombosis rates in PLWH and thus overall mortality in this population.
Collapse
Affiliation(s)
- Megan V. Perkins
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah Joseph
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dirk P. Dittmer
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
36
|
Fragkou PC, Moschopoulos CD, Dimopoulou D, Triantafyllidi H, Birmpa D, Benas D, Tsiodras S, Kavatha D, Antoniadou A, Papadopoulos A. Cardiovascular disease and risk assessment in people living with HIV: Current practices and novel perspectives. Hellenic J Cardiol 2023; 71:42-54. [PMID: 36646212 DOI: 10.1016/j.hjc.2022.12.013] [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: 08/11/2022] [Revised: 11/28/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection represents a major cardiovascular risk factor, and the cumulative cardiovascular disease (CVD) burden among aging people living with HIV (PLWH) constitutes a leading cause of morbidity and mortality. To date, CVD risk assessment in PLWH remains challenging. Therefore, it is necessary to evaluate and stratify the cardiovascular risk in PLWH with appropriate screening and risk assessment tools and protocols to correctly identify which patients are at a higher risk for CVD and will benefit most from prevention measures and timely management. This review aims to accumulate the current evidence on the association between HIV infection and CVD, as well as the risk factors contributing to CVD in PLWH. Furthermore, considering the need for cardiovascular risk assessment in daily clinical practice, the purpose of this review is also to report the current practices and novel perspectives in cardiovascular risk assessment of PLWH and provide further insights into the development and implementation of appropriate CVD risk stratification and treatment strategies, particularly in countries with high HIV burden and limited resources.
Collapse
Affiliation(s)
- Paraskevi C Fragkou
- First Department of Critical Care Medicine and Pulmonary Services, Evangelismos Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Charalampos D Moschopoulos
- Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitra Dimopoulou
- Second Department of Pediatrics, Children's Hospital "Panagiotis and Aglaia Kyriakou", National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Helen Triantafyllidi
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dionysia Birmpa
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitrios Benas
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Sotirios Tsiodras
- First Department of Critical Care Medicine and Pulmonary Services, Evangelismos Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitra Kavatha
- Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anastasia Antoniadou
- Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Antonios Papadopoulos
- Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
37
|
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: 13] [Impact Index Per Article: 13.0] [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
|
38
|
Zureigat H, Abohashem S, Osborne MT, Lo J, Hsue P, Tawakol A. Evidence of an anti-inflammatory effect of statins in people living with HIV. J Nucl Cardiol 2022; 29:3069-3071. [PMID: 35277834 PMCID: PMC9464260 DOI: 10.1007/s12350-022-02922-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 01/22/2023]
Affiliation(s)
- Hadil Zureigat
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shady Abohashem
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA
| | - Janet Lo
- Endocrinology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Priscilla Hsue
- Cardiology Division, Department of Medicine, Zuckerberg San Francisco General Hospital and University of California-San Francisco, San Francisco, CA, USA
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Cardiology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Yawkey 5E, Boston, MA, 02114-2750, USA.
| |
Collapse
|
39
|
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: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
40
|
Chehab O, Kanj A, Zeitoun R, Mir T, Shafi I, Pahuja M, Briasoulis A, Doria de Vasconcellos H, Minhas A, Varadarajan V, Wu C, Arbab-Zadeh A, Post WS, Wu KC, Lima JA. Association of HIV infection with clinical features and outcomes of patients with aortic aneurysms. Vasc Med 2022; 27:557-564. [PMID: 36190774 DOI: 10.1177/1358863x221122577] [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: 01/24/2023]
Abstract
Data on the characteristics and outcomes of hospitalized patients with aortic aneurysms (AA) and HIV remain scarce. This is a cohort study of hospitalized adult patients with a diagnosis of AA from 2013 to 2019 using the US National Inpatient Readmission Database. Patients with a diagnosis of HIV were identified. Our outcomes included trends in hospitalizations and comparison of clinical characteristics, complications, and mortality in patients with AA and HIV compared to those without HIV. Among 1,905,837 hospitalized patients with AA, 4416 (0.23%) were living with HIV. There was an overall age-adjusted increase in the rate of HIV among patients hospitalized with AA over the years (14-29 per 10,000 person-years; age-adjusted p-trend < 0.001). Patients with AA and HIV were younger than those without HIV (median age: 60 vs 76 years, p < 0.001) and were less likely to have a history of smoking, hypertension, dyslipidemia, diabetes mellitus, and obesity. Thoracic aortic aneurysms were more prevalent in those with HIV (37.5% vs 26.7%, p < 0.001). On multivariable logistic regression, HIV was not associated with increased risk of aortic rupture (OR: 0.79; 95% CI: 0.61-1.01, p = 0.06), acute aortic dissection (OR: 0.73; 95% CI: 0.51-1.06, p = 0.3), readmissions (OR: 1.04; 95% CI: 0.95-1.13, p = 0.4), or aortic repair (OR: 0.89; 95% CI: 0.79-1.00, p = 0.05). Hospitalized patients with AA and HIV had a lower crude mortality rate compared to those without HIV (OR: 0.75 (0.63-0.91), p = 0.003). Hospitalized patients with AA and HIV likely constitute a distinct group of patients with AA; they are younger, have fewer traditional cardiovascular risk factors, and a higher rate of thoracic aorta involvement. Differences in clinical features may account for the lower mortality rate observed in patients with AA and HIV compared to those without HIV.
Collapse
Affiliation(s)
- Omar Chehab
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amjad Kanj
- Division of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Ralph Zeitoun
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tanveer Mir
- Department of Internal Medicine, Wayne State University / Detroit Medical Center, Detroit, MI, USA
| | - Irfan Shafi
- Department of Internal Medicine, Wayne State University / Detroit Medical Center, Detroit, MI, USA.,Department of Internal Medicine, Division of Cardiology, Wayne State University / Detroit Medical Center, Detroit, MI, USA
| | - Mohit Pahuja
- Department of Cardiology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Alexandros Briasoulis
- Division of Cardiovascular Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | | | - Anum Minhas
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vinithra Varadarajan
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colin Wu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Armin Arbab-Zadeh
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wendy S Post
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katherine C Wu
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - João Ac Lima
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Kolossváry M, deFilippi C, Lu MT, Zanni MV, Fulda ES, Foldyna B, Ribaudo H, Mayrhofer T, Collier AC, Bloomfield GS, Fichtenbaum C, Overton ET, Aberg JA, Currier J, Fitch KV, Douglas PS, Grinspoon SK. Proteomic Signature of Subclinical Coronary Artery Disease in People With HIV: Analysis of the REPRIEVE Mechanistic Substudy. J Infect Dis 2022; 226:1809-1822. [PMID: 35535576 PMCID: PMC10205625 DOI: 10.1093/infdis/jiac196] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/06/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND People with HIV (PWH) have subclinical coronary artery disease (CAD) despite low traditional atherosclerotic cardiovascular disease (ASCVD) risk scores. Coronary plaque in PWH presents as a unique phenotype, but little is known about the contributions of specific inflammatory pathways to plaque phenotypes in PWH. METHODS The REPRIEVE Mechanistic Substudy enrolled PWH on ART without known cardiovascular disease. We used a targeted discovery proteomics approach to evaluate 246 unique proteins representing cardiovascular, inflammatory, and immune pathways. Proteomic signatures were determined for presence of coronary artery calcium (CAC > 0) and presence of coronary plaque. RESULTS Data were available for 662 participants (aged 51 [SD 6] years, ASCVD risk score 4.9% [SD 3.1%]). Among 12 proteins associated with both CAC and presence of coronary plaque, independent of ASCVD risk score, the odds ratios were highest for NRP1: 5.1 (95% confidence interval [CI], 2.3-11.4) for CAC and 2.9 (95% CI, 1.4-6.1) for presence of plaque. Proteins uniquely related to presence of plaque were CST3, LTBR, MEPE, PLC, SERPINA5, and TNFSF13B; in contrast, DCN, IL-6RA, OSMR, ST2, and VCAM1 were only related to CAC. CONCLUSIONS Distinct immune and inflammatory pathways are differentially associated with subclinical CAD phenotypes among PWH. This comprehensive set of targets should be further investigated to reduce atherosclerosis and ASCVD in PWH. CLINICAL TRIALS REGISTRATION NCT02344290.
Collapse
Affiliation(s)
- Márton Kolossváry
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Chris deFilippi
- Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evelynne S Fulda
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston Massachusetts, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Ann C Collier
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Gerald S Bloomfield
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Carl Fichtenbaum
- Department of Medicine for Translational Research, University of Cincinnati, Cincinnati, Ohio, USA
| | - Edgar T Overton
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith Currier
- Division of Infectious Diseases, University of California at Los Angeles, Los Angeles, California, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Shen G, Thomas TS, Walpert AR, McClure CM, Fitch KV, deFilippi C, Torriani M, Buckless CG, Adler GK, Grinspoon SK, Srinivasa S. Role of renin-angiotensin-aldosterone system activation and other metabolic variables in relation to arterial inflammation in HIV. Clin Endocrinol (Oxf) 2022; 97:581-587. [PMID: 35614846 PMCID: PMC9532371 DOI: 10.1111/cen.14784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Arterial inflammation remains increased among persons with HIV (PWH) compared with persons without HIV (PWOH) even when controlling for traditional risk factors. We sought to understand whether increased renin-angiotensin-aldosterone system (RAAS) activation may be related to arterial inflammation in PWH and when compared with PWOH. DESIGN Twenty PWH and 9 PWOH followed a controlled, standardized low and liberal sodium diet to simulate a RAAS-activated and RAAS-suppressed state, respectively. We measured serum lipoprotein-associated phospholipase A2 (LpPLA2) concentrations following both conditions to assess the physiologic dynamics of aldosterone in relation to arterial inflammation. RESULTS LpPLA2 levels were significantly higher among PWH versus PWOH during both the RAAS-activated state[5.3(4.2, 6.1) versus 4.0(3.0, 4.8)nmol/L, median(interquartile range),p = .01]) and RAAS-suppressed state[4.4(3.9, 5.3) versus 3.8(3.4, 4.1)nmol/L,p = .01]. Among PWH, but not PWOH, LpPLA2 increased significantly with RAAS activation(p = .03). LpPLA2 levels measured during the RAAS-suppressed state among PWH remained relatively higher than LpPLA2 levels under both conditions among PWOH. Log LpPLA2 was related to log aldosterone during the RAAS-activated state(r = .39,p = .04) among all participants. Log LpPLA2 was correlated with visceral fat(r = .46,p = .04) and log systolic blood pressure(r = .57,p = .009) during a RAAS-activated state when an increase in aldosterone was stimulated in HIV. CONCLUSION LpPLA2 is increased during a RAAS-activated state among PWH, but not among PWOH. Further, LpPLA2 was increased in both RAAS-activated and suppressed states in PWH compared with PWOH. These data suggest a biological link between increased aldosterone and arterial inflammation in this population. Future studies should test RAAS blockade on arterial inflammation as a targeted treatment approach in HIV.
Collapse
Affiliation(s)
- Grace Shen
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teressa S Thomas
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Allie R Walpert
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Colin M McClure
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Martin Torriani
- Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Colleen G Buckless
- Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gail K Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Palshikar MG, Palli R, Tyrell A, Maggirwar S, Schifitto G, Singh MV, Thakar J. Executable models of immune signaling pathways in HIV-associated atherosclerosis. NPJ Syst Biol Appl 2022; 8:35. [PMID: 36131068 PMCID: PMC9492768 DOI: 10.1038/s41540-022-00246-5] [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: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Atherosclerosis (AS)-associated cardiovascular disease is an important cause of mortality in an aging population of people living with HIV (PLWH). This elevated risk has been attributed to viral infection, anti-retroviral therapy, chronic inflammation, and lifestyle factors. However, the rates at which PLWH develop AS vary even after controlling for length of infection, treatment duration, and for lifestyle factors. To investigate the molecular signaling underlying this variation, we sequenced 9368 peripheral blood mononuclear cells (PBMCs) from eight PLWH, four of whom have atherosclerosis (AS+). Additionally, a publicly available dataset of PBMCs from persons before and after HIV infection was used to investigate the effect of acute HIV infection. To characterize dysregulation of pathways rather than just measuring enrichment, we developed the single-cell Boolean Omics Network Invariant Time Analysis (scBONITA) algorithm. scBONITA infers executable dynamic pathway models and performs a perturbation analysis to identify high impact genes. These dynamic models are used for pathway analysis and to map sequenced cells to characteristic signaling states (attractor analysis). scBONITA revealed that lipid signaling regulates cell migration into the vascular endothelium in AS+ PLWH. Pathways implicated included AGE-RAGE and PI3K-AKT signaling in CD8+ T cells, and glucagon and cAMP signaling pathways in monocytes. Attractor analysis with scBONITA facilitated the pathway-based characterization of cellular states in CD8+ T cells and monocytes. In this manner, we identify critical cell-type specific molecular mechanisms underlying HIV-associated atherosclerosis using a novel computational method.
Collapse
Affiliation(s)
- Mukta G Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Alicia Tyrell
- University of Rochester Clinical & Translational Science Institute, Rochester, USA
| | - Sanjay Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Meera V Singh
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Juilee Thakar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
44
|
Alam SR, Vinayak S, Shah A, Doolub G, Kimeu R, Horn KP, Bowen SR, Jeilan M, Lee KK, Gachoka S, Riunga F, Adam RD, Vesselle H, Joshi N, Obino M, Makhdomi K, Ombati K, Nganga E, Gitau S, Chung MH, Shah ASV. Assessment of Cardiac, Vascular, and Pulmonary Pathobiology In Vivo During Acute COVID‐19. J Am Heart Assoc 2022; 11:e026399. [DOI: 10.1161/jaha.122.026399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background
Acute COVID‐19–related myocardial, pulmonary, and vascular pathology and how these relate to each other remain unclear. To our knowledge, no studies have used complementary imaging techniques, including molecular imaging, to elucidate this. We used multimodality imaging and biochemical sampling in vivo to identify the pathobiology of acute COVID‐19. Specifically, we investigated the presence of myocardial inflammation and its association with coronary artery disease, systemic vasculitis, and pneumonitis.
Methods and Results
Consecutive patients presenting with acute COVID‐19 were prospectively recruited during hospital admission in this cross‐sectional study. Imaging involved computed tomography coronary angiography (identified coronary disease), cardiac 2‐deoxy‐2‐[fluorine‐18]fluoro‐D‐glucose positron emission tomography/computed tomography (identified vascular, cardiac, and pulmonary inflammatory cell infiltration), and cardiac magnetic resonance (identified myocardial disease) alongside biomarker sampling. Of 33 patients (median age 51 years, 94% men), 24 (73%) had respiratory symptoms, with the remainder having nonspecific viral symptoms. A total of 9 patients (35%, n=9/25) had cardiac magnetic resonance–defined myocarditis. Of these patients, 53% (n=5/8) had myocardial inflammatory cell infiltration. A total of 2 patients (5%) had elevated troponin levels. Cardiac troponin concentrations were not significantly higher in patients with and without myocarditis (8.4 ng/L [interquartile range, IQR: 4.0–55.3] versus 3.5 ng/L [IQR: 2.5–5.5];
P
=0.07) or myocardial cell infiltration (4.4 ng/L [IQR: 3.4–8.3] versus 3.5 ng/L [IQR: 2.8–7.2];
P
=0.89). No patients had obstructive coronary artery disease or vasculitis. Pulmonary inflammation and consolidation (percentage of total lung volume) was 17% (IQR: 5%–31%) and 11% (IQR: 7%–18%), respectively. Neither were associated with the presence of myocarditis.
Conclusions
Myocarditis was present in a third patients with acute COVID‐19, and the majority had inflammatory cell infiltration. Pneumonitis was ubiquitous, but this inflammation was not associated with myocarditis. The mechanism of cardiac pathology is nonischemic and not attributable to a vasculitic process.
Registration
URL:
https://www.isrctn.com
; Unique identifier: ISRCTN12154994.
Collapse
Affiliation(s)
- Shirjel R. Alam
- Department of Cardiology Manchester University Manchester United Kingdom
- Department of Cardiology North Bristol Trust Bristol United Kingdom
- Non‐communicable Disease Epidemiology London School of Hygiene and Tropical Medicine London United Kingdom
| | - Sudhir Vinayak
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Adeel Shah
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Gemina Doolub
- Department of Cardiology University of Bristol Bristol United Kingdom
| | - Redemptar Kimeu
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kevin P. Horn
- Department of Radiology University of Washington Seattle WA
| | | | - Mohamed Jeilan
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kuan Ken Lee
- Department of Cardiology University of Edinburgh Edinburgh UK
| | - Sylvia Gachoka
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Felix Riunga
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Rodney D. Adam
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | | | - Nikhil Joshi
- Department of Cardiology University of Bristol Bristol United Kingdom
| | - Mariah Obino
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Khalid Makhdomi
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Kevin Ombati
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Edward Nganga
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Samuel Gitau
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| | - Michael H. Chung
- Division of Infectious Diseases of the Department of Medicine Emory University Atlanta Georgia
| | - Anoop S. V. Shah
- Department of Radiology and Department of Medicine Aga Khan University Nairobi Kenya
| |
Collapse
|
45
|
Hudson JA, Majonga ED, Ferrand RA, Perel P, Alam SR, Shah ASV. Association of HIV Infection With Cardiovascular Pathology Based on Advanced Cardiovascular Imaging: A Systematic Review. JAMA 2022; 328:951-962. [PMID: 36098725 PMCID: PMC9471974 DOI: 10.1001/jama.2022.15078] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE HIV-associated cardiovascular disease is increasing in prevalence, but its mechanisms remain poorly understood. OBJECTIVE To systematically review data from advanced cardiovascular imaging studies evaluating computed tomographic coronary angiography, positron emission tomography (PET), and cardiac magnetic resonance (MR), in people living with HIV compared with uninfected individuals. DATA SOURCES Three databases and Google Scholar were searched for studies assessing cardiovascular pathology using computed tomographic coronary angiography, cardiac MR, PET, and HIV from inception to February 11, 2022. STUDY SELECTION Two reviewers selected original studies without any restrictions on design, date, or language, investigating HIV and cardiovascular pathology. DATA EXTRACTION AND SYNTHESIS One investigator extracted data checked by a second investigator. Prevalence ratios (PRs) and differences in inflammation among people living with HIV and uninfected individuals were qualitatively synthesized in terms of cardiovascular pathology. Study quality was assessed using the National Heart, Lung, and Blood Institute quality assessment tool for observational studies. MAIN OUTCOMES AND MEASURES Primary outcomes were computed tomographic coronary angiography-defined moderate to severe (≥50%) coronary stenosis, cardiac MR-defined myocardial fibrosis identified by late gadolinium enhancement, and PET-defined vascular and myocardial target to background ratio. Prevalence of moderate to severe coronary disease, as well as myocardial fibrosis, and PRs compared with uninfected individuals were reported alongside difference in vascular target to background ratio. RESULTS Forty-five studies including 5218 people living with HIV (mean age, 48.5 years) and 2414 uninfected individuals (mean age, 49.1 years) were identified. Sixteen studies (n = 5107 participants) evaluated computed tomographic coronary angiography; 16 (n = 1698), cardiac MRs; 10 (n = 681), vascular PET scans; and 3 (n = 146), both computed tomographic coronary angiography and vascular PET scans. No studies originated from low-income countries. Regarding risk of bias, 22% were classified as low; 47% moderate; and 31% high. Prevalence of moderate to severe coronary disease among those with vs without HIV ranged from 0% to 52% and 0% to 27%, respectively, with PRs ranging from 0.33 (95% CI, 0.01-15.90) to 5.19 (95% CI, 1.26-21.42). Prevalence of myocardial fibrosis among those with vs without HIV ranged from 5% to 84% and 0% to 68%, respectively, with PRs ranging from 1.01 (95% CI, 0.85-1.21) to 17.35 (95% CI, 1.10-274.28). Differences in vascular target to background ratio among those with vs without HIV ranged from 0.06 (95% CI, 0.01-0.11) to 0.37 (95% CI, 0.02-0.72). CONCLUSIONS AND RELEVANCE In this systematic review of studies of advanced cardiovascular imaging, the estimates of the associations between HIV and cardiovascular pathologies demonstrated large amounts of heterogeneity. The findings provide a summary of the available data but may not be representative of all individuals living with HIV, including those from low-income countries with higher HIV endemicity.
Collapse
Affiliation(s)
- Jonathan A. Hudson
- Kings College London British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine & Sciences, London, United Kingdom
- Department of Cardiology, Epsom and St Helier University Hospitals Trust, London, United Kingdom
| | - Edith D. Majonga
- Biomedical Research and Training Institute, Harare, Zimbabwe
- Department of Medical Physics and Imaging Sciences, Faculty of Medicine and Health Sciences, University of Zimbabwe
| | - Rashida A. Ferrand
- Biomedical Research and Training Institute, Harare, Zimbabwe
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Pablo Perel
- Department of Non-communicable Disease, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Shirjel R. Alam
- Department of Non-communicable Disease, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Cardiology, North Bristol NHS Trust, Bristol, London, United Kingdom
| | - Anoop S. V. Shah
- Department of Non-communicable Disease, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Cardiology, Imperial College NHS Trust, London, United Kingdom
| |
Collapse
|
46
|
Riley ED, Kizer JR, Tien PC, Vittinghoff E, Lynch KL, Wu AHB, Coffin PO, Beck-Engeser G, Braun C, Hunt PW. Multiple substance use, inflammation and cardiac stretch in women living with HIV. Drug Alcohol Depend 2022; 238:109564. [PMID: 35872529 PMCID: PMC9924802 DOI: 10.1016/j.drugalcdep.2022.109564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/10/2022] [Accepted: 07/03/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) and heart failure (HF) are disproportionately high in people living with HIV and differ by sex. Few CVD-related studies focus on drug use, yet it is common in low-income women living with HIV (WLWH) and increases cardiac dysfunction. SETTING We recruited unsheltered and unstably housed WLWH from San Francisco community venues to participate in a six-month cohort study investigating linkages between drug use, inflammation, and cardiac dysfunction. METHODS Adjusting for CVD risk factors, co-infections, medications, and menopause, we examined the effects of toxicology-confirmed drug use and inflammation (C-reactive protein, sCD14, sCD163 and sTNFR2) on levels of NT-proBNP, a biomarker of cardiac stretch and HF. RESULTS Among 74 WLWH, the median age was 53 years and 45 % were Black. At baseline, 72 % of participants had hypertension. Substances used included tobacco (65 %), cannabis (53 %), cocaine (49 %), methamphetamine (31 %), alcohol (28 %), and opioids (20 %). Factors significantly associated with NT-proBNP included cannabis use (Adjusted Relative Effect [ARE]: -39.6 %) and sTNFR2 (ARE: 65.5 %). Adjusting for heart failure and restricting analyses to virally suppressed persons did not diminish effects appreciably. Cannabis use was not significantly associated with sTNFR2 and did not change the association between sTNFR2 and NT-proBNP. CONCLUSIONS Among polysubstance-using WLWH, NT-proBNP levels signaling cardiac stretch were positively associated with sTNFR2, but 40 % lower in people who used cannabis. Whether results suggest that cardiovascular pathways associated with cannabis use mitigate cardiac stress and dysfunction independent of inflammation in WLWH who use multiple substances merits further investigation.
Collapse
Affiliation(s)
- Elise D Riley
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, CA, USA.
| | - Jorge R Kizer
- San Francisco VA Health Care System, Division of Cardiology, San Francisco, CA, USA; University of California, San Francisco, School of Medicine, Department of Epidemiology and Biostatistics, San Francisco, CA, USA
| | - Phyllis C Tien
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, CA, USA
| | - Eric Vittinghoff
- University of California, San Francisco, School of Medicine, Department of Epidemiology and Biostatistics, San Francisco, CA, USA
| | - Kara L Lynch
- University of California, San Francisco, School of Medicine, Department of Laboratory Medicine, San Francisco, CA, USA
| | - Alan H B Wu
- University of California, San Francisco, School of Medicine, Department of Laboratory Medicine, San Francisco, CA, USA
| | - Phillip O Coffin
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, CA, USA; San Francisco Department of Public Health, San Francisco, CA, USA
| | - Gabriele Beck-Engeser
- University of California, San Francisco, School of Medicine, Department of Experimental Medicine, San Francisco, CA, USA
| | - Carl Braun
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, CA, USA
| | - Peter W Hunt
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, CA, USA; University of California, San Francisco, School of Medicine, Department of Experimental Medicine, San Francisco, CA, USA
| |
Collapse
|
47
|
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: 1.5] [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
|
48
|
Chan P, Spudich S. Investigating vascular diseases in people living with HIV by nuclear imaging. J Nucl Cardiol 2022; 29:1576-1582. [PMID: 33884573 DOI: 10.1007/s12350-021-02613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Phillip Chan
- SEARCH, Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Serena Spudich
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
49
|
Taglieri N, Bonfiglioli R, Bon I, Malosso P, Corovic A, Bruno M, Le E, Granozzi B, Palmerini T, Ghetti G, Tamburello M, Bruno AG, Saia F, Tarkin JM, Rudd JHF, Calza L, Fanti S, Re MC, Galié N. Pattern of arterial inflammation and inflammatory markers in people living with HIV compared with uninfected people. J Nucl Cardiol 2022; 29:1566-1575. [PMID: 33569752 PMCID: PMC9345795 DOI: 10.1007/s12350-020-02522-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/25/2020] [Indexed: 11/03/2022]
Abstract
STUDY DESIGN To compare arterial inflammation (AI) between people living with HIV (PLWH) and uninfected people as assessed by 18F-Fluorodeoxyglucose (18F-FDG)-positron emission tomography (PET). METHODS We prospectively enrolled 20 PLWH and 20 uninfected people with no known cardiovascular disease and at least 3 traditional cardiovascular risk factors. All patients underwent 18F-FDG-PET/computed tomography (CT) of the thorax and neck. Biomarkers linked to inflammation and atherosclerosis were also determined. The primary outcome was AI in ascending aorta (AA) measured as mean maximum target-to-background ratio (TBRmax). The independent relationships between HIV status and both TBRmax and biomarkers were evaluated by multivariable linear regression adjusted for body mass index, creatinine, statin therapy, and atherosclerotic cardiovascular 10-year estimated risk (ASCVD). RESULTS Unadjusted mean TBRmax in AA was slightly higher but not statistically different (P = .18) in PLWH (2.07; IQR 1.97, 2.32]) than uninfected people (2.01; IQR 1.85, 2.16]). On multivariable analysis, PLWH had an independent risk of increased mean log-TBRmax in AA (coef = 0.12; 95%CI 0.01,0.22; P = .032). HIV infection was independently associated with higher values of interleukin-10 (coef = 0.83; 95%CI 0.34, 1.32; P = .001), interferon-γ (coef. = 0.90; 95%CI 0.32, 1.47; P = .003), and vascular cell adhesion molecule-1 (VCAM-1) (coef. = 0.75; 95%CI: 0.42, 1.08, P < .001). CONCLUSIONS In patients with high cardiovascular risk, HIV status was an independent predictor of increased TBRmax in AA. PLWH also had an increased independent risk of IFN-γ, IL-10, and VCAM-1 levels.
Collapse
Affiliation(s)
- Nevio Taglieri
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Rachele Bonfiglioli
- Division of Nuclear Medicine, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Isabella Bon
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Pietro Malosso
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Andrej Corovic
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Matteo Bruno
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Elizabeth Le
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Bianca Granozzi
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Tullio Palmerini
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Gabriele Ghetti
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Martina Tamburello
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Antonio Giulio Bruno
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Francesco Saia
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Leonardo Calza
- Department of Medical and Surgical Sciences, Clinics of Infectious Diseases, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Division of Nuclear Medicine, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Carla Re
- Division of Microbiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St. Orsola, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Nazzareno Galié
- Division of Cardiology, Department of Experimental Diagnostic and Specialty Medicine, IRCCS Policlinico di St.Orsola, Alma Mater Studiorum-University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| |
Collapse
|
50
|
Toribio M, Wilks MQ, Hedgire S, Lu MT, Cetlin M, Wang M, Alhallak I, Durbin CG, White KS, Wallis Z, Schnittman SR, Stanley TL, El-Fakhri G, Lee H, Autissier P, Zanni MV, Williams KC, Grinspoon SK. Increased Macrophage-Specific Arterial Infiltration Relates to Non-calcified Plaque and Systemic Immune Activation in People with HIV. J Infect Dis 2022; 226:1823-1833. [PMID: 35856671 DOI: 10.1093/infdis/jiac301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Persistent immune activation is thought to contribute to heightened atherosclerotic cardiovascular disease (ASCVD) risk among people with HIV (PWH). METHODS Participants (≥18 years) with versus without HIV and without history of clinical ASCVD were enrolled. We hypothesized that increased macrophage-specific arterial infiltration would relate to plaque composition and systemic immune activation among PWH. We applied a novel targeted molecular imaging approach [technetium-99 m (99mTc)-tilmanocept single photon emission computed tomography (SPECT)/CT] and comprehensive immune phenotyping. RESULTS Aortic 99mTc-tilmanocept uptake was significantly higher among PWH (N = 20) versus participants without HIV (N = 10) with similar 10-year ASCVD risk (P = 0.02). Among PWH, but not among participants without HIV, non-calcified aortic plaque volume related directly to aortic 99mTc-tilmanocept uptake at different uptake thresholds. An interaction (P = 0.001) was seen between HIV status and non-calcified plaque volume, but not calcified plaque (P = 0.83). Systemic levels of caspase-1 (P = 0.004), CD14-CD16+ (non-classical/patrolling/homing) monocytes (P = 0.0004) and CD8+ T-cells (P = 0.005) related positively and CD4+/CD8 + T-cell ratio (P = 0.02) inversely to aortic 99mTc-tilmanocept uptake volume. CONCLUSIONS Macrophage-specific arterial infiltration was higher among PWH and related to non-calcified aortic plaque volume only among PWH. Key systemic markers of immune activation relating to macrophage-specific arterial infiltration may contribute to heightened ASCVD risk among PWH.
Collapse
Affiliation(s)
- Mabel Toribio
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Moses Q Wilks
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sandeep Hedgire
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Madeline Cetlin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Melissa Wang
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Iad Alhallak
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claudia G Durbin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin S White
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Zoey Wallis
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Samuel R Schnittman
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Takara L Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Georges El-Fakhri
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|