101
|
Choi H, Dey AK, Sharma G, Bhoite R, Burkholder G, Fedson S, Jneid H. Etiology and pathophysiology of heart failure in people with HIV. Heart Fail Rev 2021; 26:497-505. [PMID: 33619685 DOI: 10.1007/s10741-020-10048-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 11/25/2022]
Abstract
HIV-associated cardiomyopathy is a well-established sequela in people infected with HIV (PHIV). Despite significant advances in HIV management through the use of highly active anti-retroviral therapy (HAART), PHIV on HAART continue to have elevated risk of cardiomyopathy and heart failure, even when accounting for known cardiovascular risk factors. This review article will explore the proposed mechanisms by which chronic HIV infection induces cardiomyopathy and heart failure in the setting of HAART. Evaluation, work-up, and management of cardiomyopathy in PHIV will also be briefly discussed. The advent of HAART has altered the pathophysiology HIV-associated cardiomyopathy from a rapidly progressive cardiomyopathy, often with pericardial involvement, into a chronic process involving inflammation and persistent immune dysregulation. With the significant decrease in AIDS-related deaths, the prevalence of cardiomyopathy and the mortality associated with heart failure in PHIV have increased. Multiple immune-related and inflammatory mechanisms have been proposed, which may provide insight into evaluation and management of cardiomyopathy in PHIV.
Collapse
Affiliation(s)
- Harry Choi
- Section of Inflammation and Cardiometabolic Diseases, National Institute of Health, Bethesda, MD, USA
| | - Amit K Dey
- Section of Inflammation and Cardiometabolic Diseases, National Institute of Health, Bethesda, MD, USA
| | - Gaurav Sharma
- Division of Medicine, University of South Alabama, Mobile, AL, USA
| | - Rahul Bhoite
- Division of Medicine, MedStar Union Memorial Hospital and Good Samaritan Hospital, Baltimore, MD, USA
| | - Greer Burkholder
- Division of Infectious Diseases, University of Alabama, Birmingham, AL, USA
| | - Savitri Fedson
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VA Hospital, Houston, TX, 77030, USA
| | - Hani Jneid
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VA Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
102
|
Bonou M, Kapelios CJ, Athanasiadi E, Mavrogeni SI, Psichogiou M, Barbetseas J. Imaging modalities for cardiovascular phenotyping in asymptomatic people living with HIV. Vasc Med 2021; 26:326-337. [PMID: 33475050 DOI: 10.1177/1358863x20978702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) has emerged as a leading cause of non-HIV-related mortality among people living with HIV (PLWH). Despite the growing CVD burden in PLWH, there is concern that general population risk score models may underestimate CVD risk in these patients. Imaging modalities have received mounting attention lately to better understand the pathophysiology of subclinical CVD and provide improved risk assessment in this population. To date, traditional and well-established techniques such as echocardiography, pulse wave velocity, and carotid intima thickness continue to be the basis for the diagnosis and subsequent monitoring of vascular atherosclerosis and heart failure. Furthermore, novel imaging tools such as cardiac computed tomography (CT) and cardiac CT angiography (CCTA), positron emission tomography/CT (PET/CT), and cardiac magnetic resonance (CMR) have provided new insights into accelerated cardiovascular abnormalities in PLWH and are currently evaluated with regards to their potential to improve risk stratification.
Collapse
Affiliation(s)
- Maria Bonou
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | - Chris J Kapelios
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | - Eleni Athanasiadi
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | | | - Mina Psichogiou
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - John Barbetseas
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| |
Collapse
|
103
|
Kuk M, Ward NC, Dwivedi G. Extrinsic and Intrinsic Responses in the Development and Progression of Atherosclerosis. Heart Lung Circ 2021; 30:807-816. [PMID: 33468387 DOI: 10.1016/j.hlc.2020.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 11/25/2022]
Abstract
Atherosclerosis is a multifactorial disease that is thought to be primarily inflammatory in origin. Given the contribution of inflammation to the development and progression of atherosclerosis, other conditions that are characterised by a dysregulated inflammatory response have also been proposed to play a role. The purpose of this review is to organise and present the various inflammatory processes that can affect atherosclerosis into two broad categories: extrinsic or host-independent and intrinsic or host-dependent. Within these two categories, we will discuss various processes that may contribute to the development and progression of atherosclerosis and the clinical studies describing these associations. Although the clinical trials investigating anti-inflammatory therapies have to date provided mixed results, further studies, particularly in conjunction with lipid-lowering and blood pressure lowering therapies should be considered.
Collapse
Affiliation(s)
- Mariya Kuk
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada; McGill University Health Centre, McGill University, Montreal, Canada
| | - Natalie C Ward
- School of Public Health, Curtin University, Perth, WA, Australia; Medical School, University of Western Australia, Perth, WA, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, WA, Australia; Harry Perkins Institute for Medical Research, Fiona Stanley Hospital, Perth, WA, Australia.
| |
Collapse
|
104
|
Stein JH, Kime N, Korcarz CE, Ribaudo H, Currier JS, Delaney JC. Effects of HIV Infection on Arterial Endothelial Function: Results From a Large Pooled Cohort Analysis. Arterioscler Thromb Vasc Biol 2021; 41:512-522. [PMID: 33327750 PMCID: PMC7770018 DOI: 10.1161/atvbaha.120.315435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/26/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To determine the effects of HIV serostatus and disease severity on endothelial function in a large pooled cohort study of people living with HIV infection and HIV- controls. Approach and Results: We used participant-level data from 9 studies: 7 included people living with HIV (2 treatment-naïve) and 4 had HIV- controls. Brachial artery flow-mediated dilation (FMD) was measured using a standardized ultrasound imaging protocol with central reading. After data harmonization, multiple linear regression was used to examine the effects of HIV- serostatus, HIV disease severity measures, and cardiovascular disease risk factors on FMD. Of 2533 participants, 986 were people living with HIV (mean 44.4 [SD 11.8] years old) and 1547 were HIV- controls (42.9 [12.2] years old). The strongest and most consistent associates of FMD were brachial artery diameter, age, sex, and body mass index. The effect of HIV+ serostatus on FMD was strongly influenced by kidney function. In the highest tertile of creatinine (1.0 mg/dL), the effect of HIV+ serostatus was strong (β=-1.59% [95% CI, -2.58% to -0.60%], P=0.002), even after covariate adjustment (β=-1.36% [95% CI, -2.46% to -0.47%], P=0.003). In the lowest tertile (0.8 mg/dL), the effect of HIV+ serostatus was strong (β=-1.90% [95% CI, -2.58% to -1.21%], P<0.001), but disappeared after covariate adjustment. HIV RNA viremia, CD4+ T-cell count, and use of antiretroviral therapy were not meaningfully associated with FMD. CONCLUSIONS The significant effect of HIV+ serostatus on FMD suggests that people living with HIV are at increased cardiovascular disease risk, especially if they have kidney disease.
Collapse
Affiliation(s)
- James H. Stein
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Noah Kime
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
| | - Claudia E. Korcarz
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | | | - Judith S. Currier
- David Geffen School of Medicine at University of California -Los Angeles; Los Angeles, CA
| | - Joseph C. Delaney
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba; Winnipeg, MB
| |
Collapse
|
105
|
Bogorodskaya M, Lyass A, Mahoney TF, Borowsky LH, Sen P, Swirski FK, Srinivasa S, Longenecker CT, Massaro JM, D'Agostino RB, Triant VA. Utilization of absolute monocyte counts to predict cardiovascular events in people living with HIV. HIV Med 2020; 22:314-320. [PMID: 33295150 DOI: 10.1111/hiv.13018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Cardiovascular risk is increased in people living with HIV (PLWH). In HIV-uninfected populations, total absolute monocyte count (AMC) has been shown to be predictive of future cardiovascular events (CVEs). We sought to determine whether AMC predicts CVEs in PLWH independent of established and HIV-related cardiovascular risk factors. METHODS We identified all PLWH within the Partners HIV Cohort without factors that could confound the monocyte count. CVE was defined as fatal or non-fatal acute myocardial infarction or ischaemic stroke. Baseline-measured AMC was defined as the average of all outpatient AMC counts a year before and after the baseline date. Multivariable Cox proportional hazards models were used to assess the association of baseline AMC with CVEs. RESULTS Our cohort consisted of 1980 patients, with median follow-up of 10.9 years and 182 CVEs. Mean (± SD) age was 41.9 ± 9.3 years; 73.0% were male. Mean CD4 count was 506.3 ± 307.1 cells/µL, 48% had HIV viral load (VL) < 400 copies/mL, and 87% were on antiretroviral therapy. Mean AMC was 0.38 × 103 ± 0.13 cells/µL. In multivariable modelling adjusted for traditional CV risk factors, CD4 cell count, and HIV VL, AMC quartile 2 (Q2) (HR = 1.01, P = 0.98), Q3 (HR = 1.07, P = 0.76), and Q4 (HR = 0.97, P = 0.89) were not significantly predictive of CVE compared with Q1. DISCUSSION Baseline AMC was not associated with long-term CVEs in PLWH. AMC obtained in routine clinical encounters does not appear to enhance CV risk stratification in PLWH.
Collapse
Affiliation(s)
- M Bogorodskaya
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - A Lyass
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - T F Mahoney
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - L H Borowsky
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - P Sen
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - F K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - S Srinivasa
- Program in Nutritional Metabolism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - C T Longenecker
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - J M Massaro
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - R B D'Agostino
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - V A Triant
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA.,Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
106
|
Lawal IO, Popoola GO, Mahapane J, Kaufmann J, Davis C, Ndlovu H, Maserumule LC, Mokoala KMG, Bouterfa H, Wester HJ, Zeevaart JR, Sathekge MM. [ 68Ga]Ga-Pentixafor for PET Imaging of Vascular Expression of CXCR-4 as a Marker of Arterial Inflammation in HIV-Infected Patients: A Comparison with 18F[FDG] PET Imaging. Biomolecules 2020; 10:E1629. [PMID: 33287237 PMCID: PMC7761707 DOI: 10.3390/biom10121629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022] Open
Abstract
People living with human immunodeficiency virus (PLHIV) have excess risk of atherosclerotic cardiovascular disease (ASCVD). Arterial inflammation is the hallmark of atherogenesis and its complications. In this study we aimed to perform a head-to-head comparison of fluorine-18 fluorodeoxyglucose positron emission tomography/computed tomography ([18F]FDG PET/CT) and Gallium-68 pentixafor positron emission tomography/computed tomography [68Ga]Ga-pentixafor PET/CT for quantification of arterial inflammation in PLHIV. We prospectively recruited human immunodeficiency virus (HIV)-infected patients to undergo [18F]FDG PET/CT and [68Ga]Ga-pentixafor PET/CT within two weeks of each other. We quantified the levels of arterial tracer uptake on both scans using maximum standardized uptake value (SUVmax) and target-background ratio. We used Bland and Altman plots to measure the level of agreement between tracer quantification parameters obtained on both scans. A total of 12 patients were included with a mean age of 44.67 ± 7.62 years. The mean duration of HIV infection and mean CD+ T-cell count of the study population were 71.08 ± 37 months and 522.17 ± 260.33 cells/µL, respectively. We found a high level of agreement in the quantification variables obtained using [18F]FDG PET and [68Ga]Ga-pentixafor PET. There is a good level of agreement in the arterial tracer quantification variables obtained using [18F]FDG PET/CT and [68Ga]Ga-pentixafor PET/CT in PLHIV. This suggests that [68Ga]Ga-pentixafor may be applied in the place of [18F]FDG PET/CT for the quantification of arterial inflammation.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa; (I.O.L.); (H.N.); (L.C.M.); (K.M.G.M.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Gbenga O. Popoola
- Department of Epidemiology and Community Health, University of Ilorin, Ilorin 240102, Nigeria;
| | - Johncy Mahapane
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| | - Jens Kaufmann
- PentixaPharm GmbH, 97082 Wuerzburg, Germany; (J.K.); (H.B.)
| | - Cindy Davis
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa; (I.O.L.); (H.N.); (L.C.M.); (K.M.G.M.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| | - Letjie C. Maserumule
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa; (I.O.L.); (H.N.); (L.C.M.); (K.M.G.M.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| | - Kgomotso M. G. Mokoala
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa; (I.O.L.); (H.N.); (L.C.M.); (K.M.G.M.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| | - Hakim Bouterfa
- PentixaPharm GmbH, 97082 Wuerzburg, Germany; (J.K.); (H.B.)
| | - Hans-Jürgen Wester
- Pharmazeutische Radiochemie, Technische Universität München, 85748 Garching, Germany;
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Radiochemistry, South African Nuclear Energy Corporation SOC (Necsa), Pelindaba 0204, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa; (I.O.L.); (H.N.); (L.C.M.); (K.M.G.M.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria 0001, South Africa; (J.M.); (C.D.)
| |
Collapse
|
107
|
Wong K, Nguyen J, Blair L, Banjanin M, Grewal B, Bowman S, Boyd H, Gerstner G, Cho HJ, Panfilov D, Tam CK, Aguilar D, Venketaraman V. Pathogenesis of Human Immunodeficiency Virus- Mycobacterium tuberculosis Co-Infection. J Clin Med 2020; 9:E3575. [PMID: 33172001 PMCID: PMC7694603 DOI: 10.3390/jcm9113575] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023] Open
Abstract
Given that infection with Mycobacterium tuberculosis (Mtb) is the leading cause of death amongst individuals living with HIV, understanding the complex mechanisms by which Mtb exacerbates HIV infection may lead to improved treatment options or adjuvant therapies. While it is well-understood how HIV compromises the immune system and leaves the host vulnerable to opportunistic infections such as Mtb, less is known about the interplay of disease once active Mtb is established. This review explores how glutathione (GSH) depletion, T cell exhaustion, granuloma formation, and TNF-α upregulation, as a result of Mtb infection, leads to an increase in HIV disease severity. This review also examines the difficulties of treating coinfected patients and suggests further research on the clinical use of GSH supplementation.
Collapse
Affiliation(s)
- Kevin Wong
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - James Nguyen
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Lillie Blair
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Marina Banjanin
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Bunraj Grewal
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Shane Bowman
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Hailey Boyd
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Grant Gerstner
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Hyun Jun Cho
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - David Panfilov
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Cho Ki Tam
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Delaney Aguilar
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific-NorthWest, Western University of Health Sciences, Lebanon, OR 97355, USA; (K.W.); (J.N.); (L.B.); (M.B.); (B.G.); (S.B.); (H.B.); (G.G.); (H.J.C.); (D.P.); (C.K.T.); (D.A.)
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
108
|
Severino P, D’Amato A, Pucci M, Infusino F, Adamo F, Birtolo LI, Netti L, Montefusco G, Chimenti C, Lavalle C, Maestrini V, Mancone M, Chilian WM, Fedele F. Ischemic Heart Disease Pathophysiology Paradigms Overview: From Plaque Activation to Microvascular Dysfunction. Int J Mol Sci 2020; 21:E8118. [PMID: 33143256 PMCID: PMC7663258 DOI: 10.3390/ijms21218118] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Ischemic heart disease still represents a large burden on individuals and health care resources worldwide. By conventions, it is equated with atherosclerotic plaque due to flow-limiting obstruction in large-medium sized coronary arteries. However, clinical, angiographic and autoptic findings suggest a multifaceted pathophysiology for ischemic heart disease and just some cases are caused by severe or complicated atherosclerotic plaques. Currently there is no well-defined assessment of ischemic heart disease pathophysiology that satisfies all the observations and sometimes the underlying mechanism to everyday ischemic heart disease ward cases is misleading. In order to better examine this complicated disease and to provide future perspectives, it is important to know and analyze the pathophysiological mechanisms that underline it, because ischemic heart disease is not always determined by atherosclerotic plaque complication. Therefore, in order to have a more complete comprehension of ischemic heart disease we propose an overview of the available pathophysiological paradigms, from plaque activation to microvascular dysfunction.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Mariateresa Pucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Fabio Infusino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Francesco Adamo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Lucrezia Netti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Giulio Montefusco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Carlo Lavalle
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| | - William M. Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA;
| | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (P.S.); (A.D.); (M.P.); (F.I.); (F.A.); (L.I.B.); (L.N.); (G.M.); (C.C.); (C.L.); (V.M.); (M.M.)
| |
Collapse
|
109
|
Bloch M, John M, Smith D, Rasmussen TA, Wright E. Managing HIV-associated inflammation and ageing in the era of modern ART. HIV Med 2020; 21 Suppl 3:2-16. [PMID: 33022087 DOI: 10.1111/hiv.12952] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This paper aims to address the concerns around ongoing immune activation, inflammation, and resistance in those ageing with HIV that represent current challenges for clinicians. METHODS Presentations at a symposium addressing issues of ageing with HIV infection were reviewed and synthesised. RESULTS The changing natural history and demographics of human immunodeficiency virus (HIV)-infected individuals means new challenges in contemporary management. In the early years of the epidemic,management was focussed on acute, potentially life-threatening AIDS-related complications. From initial monotherapy with first-generation antiretroviral therapy (ART), the development of combination highly active ART (HAART) allowed HIV control but ART toxicities, treatment adherence and drug resistance emerged as major issues. Today, the availability of potent and tolerable ART has made viral suppression achievable in most people living with HIV (PLHIV), and clinicians are confronted with managing a chronic condition among an ageing population. The combination of diseases of ageing and the co-morbidities associated with HIV-infection, even when well controlled, results in a complex set of challenges for many older PLHIV. There is a growing appreciation that many non-AIDS-related co-morbidities are caused, at least in part, by persistent, low-grade immune activation, inflammation, and hypercoagulability, despite suppressive ART. CONCLUSIONS In order to further improve HIV management, it is important to understand the enduring effects of chronically suppressed HIV infection, the potential contribution of these factors to the ageing process, the possibility of drug resistance, and the impact of different treatment strategies, including early ART initiation.
Collapse
Affiliation(s)
- M Bloch
- Holdsworth House Medical Practice, Sydney, NSW, Australia.,Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - M John
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.,Royal Perth Hospital, Perth, WA, Australia.,Institute of Immunology and Infectious Disease, Perth, WA, Australia
| | - D Smith
- School of Public Health and Community Medicine, University of New South Wales, Sydney, NSW, Australia.,The Albion Centre, Sydney, NSW, Australia
| | - T A Rasmussen
- Doherty Institute for Infection and Immunity, Melbourne, Vic., Australia.,University of Melbourne, Melbourne, Vic., Australia
| | - E Wright
- The Alfred Hospital, Melbourne, Vic., Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Vic., Australia.,The Burnett Institute, Melbourne, Vic., Australia
| |
Collapse
|
110
|
McLaughlin MM, Ma Y, Scherzer R, Rahalkar S, Martin JN, Mills C, Milush J, Deeks SG, Hsue PY. Association of Viral Persistence and Atherosclerosis in Adults With Treated HIV Infection. JAMA Netw Open 2020; 3:e2018099. [PMID: 33119103 PMCID: PMC7596582 DOI: 10.1001/jamanetworkopen.2020.18099] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE Persons living with HIV (PLWH) have increased risk for cardiovascular disease, and inflammation is thought to contribute to this excess risk. Production of HIV during otherwise effective antiretroviral therapy (ART) has been associated with inflammation. OBJECTIVE To determine whether higher levels of viral persistence are associated with atherosclerosis as assessed by changes in carotid artery intima-media thickness (IMT) over time. DESIGN, SETTING, AND PARTICIPANTS In this cohort study, intima-media thickness, a validated marker of atherosclerosis, was assessed over time in a cohort of treated PLWH with viral suppression. Cell-associated HIV DNA and RNA and change in IMT, adjusted for demographics, cardiovascular risk factors, and HIV-related factors, were examined, as well as which factors were associated with viral persistence. One hundred fifty-two PLWH with undetectable viral loads for at least 6 months before study enrollment were recruited from HIV clinics affiliated with 2 hospitals in San Francisco, California, from January 1, 2003, to December 31, 2012. Data were analyzed from February 7, 2018, to May 12, 2020. EXPOSURES Cell-associated HIV RNA and DNA were measured using enriched CD4+ T cells from cryopreserved peripheral blood mononuclear cells. MAIN OUTCOMES AND MEASURES Carotid IMT was measured at baseline and the last visit, with a mean (SD) follow-up of 4.2 (2.7) years, using high-resolution B mode ultrasonography. The main study outcomes were baseline IMT, annual IMT progression, and incident plaque, defined as a focal region of carotid IMT of greater than 1.5 mm. RESULTS The analysis included 152 PLWH (140 [92.1%] male; median age, 48.5 [interquartile range {IQR}, 43.3-53.7] years). Older age, smoking, medications for hypertension, higher low-density lipoprotein levels, and higher interleukin 6 levels were associated with higher baseline mean IMT, whereas cell-associated HIV DNA (estimate, -0.07% [95% CI, -6.1% to 6.4%]; P = .98), and HIV RNA levels (estimate, -0.8% [95% CI, -5.9% to 4.4%]; P = .75) were not. Levels of HIV RNA (0.017 [95% CI, 0.000-0.034] mm/y; P = .047) and HIV DNA (0.022 [95% CI, 0.001-0.044] mm/y; P = .042) were significantly associated with annual carotid artery IMT progression in unadjusted models only. Both HIV RNA (incidence risk ratio [IRR], 3.05 [95% CI, 1.49-6.27] per IQR; P = .002) and HIV DNA (IRR, 3.15 [95% CI, 1.51-6.57] per IQR; P = .002) were significantly associated with incident plaque, which remained significant after adjusting for demographics, cardiovascular risk factors, and HIV-related factors (IRR for HIV RNA, 4.05 [95% CI, 1.44-11.36] per IQR [P = .008]; IRR for HIV DNA, 3.35 [95% CI, 1.22-9.19] per IQR [P = .02]). Higher C-reactive protein levels were associated with higher cell-associated HIV RNA (estimate, 20.7% [95% CI, 0.9%-44.4%] per doubling; P = .04), whereas higher soluble CD14 levels were associated with HIV DNA (estimate, 18.6% [95% CI, 3.5%-35.8%] per 10% increase; P = .01). Higher soluble CD163 levels were associated with a higher HIV RNA:DNA ratio (difference, 63.8% [95% CI, 3.5%-159.4%]; P = .04). CONCLUSIONS AND RELEVANCE These findings suggest that measurements of viral persistence in treated HIV disease are independently associated with incident carotid plaque development. The size and transcriptional activity of the HIV reservoir may be important contributors to HIV-associated atherosclerosis.
Collapse
Affiliation(s)
| | - Yifei Ma
- Department of Medicine, San Francisco Veterans Affairs Medical Center, UCSF
| | - Rebecca Scherzer
- Department of Medicine, San Francisco Veterans Affairs Medical Center, UCSF
| | - Smruti Rahalkar
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, UCSF
| | | | - Claire Mills
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, UCSF
| | - Jeffrey Milush
- Department of Medicine, Division of Experimental Medicine, UCSF
| | - Steven G. Deeks
- Positive Health Program, San Francisco General Hospital, San Francisco, California
| | - Priscilla Y. Hsue
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, UCSF
| |
Collapse
|
111
|
Positron Emission Tomography to Characterize Dorsocervical Fat Pad Hypertrophy in People With Treated HIV Infection. J Acquir Immune Defic Syndr 2020; 83:e23-e25. [PMID: 31904705 DOI: 10.1097/qai.0000000000002273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
112
|
Abdelrahman KM, Chen MY, Dey AK, Virmani R, Finn AV, Khamis RY, Choi AD, Min JK, Williams MC, Buckler AJ, Taylor CA, Rogers C, Samady H, Antoniades C, Shaw LJ, Budoff MJ, Hoffmann U, Blankstein R, Narula J, Mehta NN. Coronary Computed Tomography Angiography From Clinical Uses to Emerging Technologies: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76:1226-1243. [PMID: 32883417 PMCID: PMC7480405 DOI: 10.1016/j.jacc.2020.06.076] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Evaluation of coronary artery disease (CAD) using coronary computed tomography angiography (CCTA) has seen a paradigm shift in the last decade. Evidence increasingly supports the clinical utility of CCTA across various stages of CAD, from the detection of early subclinical disease to the assessment of acute chest pain. Additionally, CCTA can be used to noninvasively quantify plaque burden and identify high-risk plaque, aiding in diagnosis, prognosis, and treatment. This is especially important in the evaluation of CAD in immune-driven conditions with increased cardiovascular disease prevalence. Emerging applications of CCTA based on hemodynamic indices and plaque characterization may provide personalized risk assessment, affect disease detection, and further guide therapy. This review provides an update on the evidence, clinical applications, and emerging technologies surrounding CCTA as highlighted at the 2019 National Heart, Lung and Blood Institute CCTA Summit.
Collapse
Affiliation(s)
- Khaled M Abdelrahman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Ramzi Y Khamis
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew D Choi
- Division of Cardiology and Department of Radiology, The George Washington University School of Medicine, Washington, DC
| | - James K Min
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Habib Samady
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leslee J Shaw
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Matthew J Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ron Blankstein
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jagat Narula
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josée and Henry R. Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, New York, New York
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
113
|
Monocyte activation and gut barrier dysfunction in South African youth on antiretroviral therapy and their associations with endothelial dysfunction. AIDS 2020; 34:1615-1623. [PMID: 32769763 DOI: 10.1097/qad.0000000000002615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is evidence for endothelial dysfunction in youth living with perinatally acquired HIV (YLPHIV). However, little data exist on its mechanisms. METHODS YLPHIV and age-matched HIV-uninfected (HIV-) youth enrolled in the Cape Town Adolescent Antiretroviral Cohort in South Africa between 9 and 14 years of age were included. YLPHIV were on antiretroviral therapy more than 6 months with viral load less than 400 copies/ml at baseline and 24 months. Serum biomarkers of systemic inflammation, monocyte activation, intestinal integrity, and oxidized LDL-cholesterol were measured at baseline and after 24 months. Endothelial function was measured at 24 months using reactive hyperemic index (RHI); endothelial dysfunction was defined as RHI less than 1.35. Spearman correlation coefficient and quantile regression were used to examine associations between RHI and different biomarkers. RESULTS We included 266 YLPHIV and 69 HIV- participants. At baseline, median (Q1, Q3) age was 12 (11, 13) years and 53% were females. YLPHIV had poorer endothelial function compared with HIV- youth (RHI = 1.36 vs. 1.52, P < 0.01). At baseline and 24 months, YLPHIV had higher markers of monocyte activation (soluble CD14), gut barrier dysfunction (intestinal fatty acid binding protein) and oxidized LDL-cholesterol (P ≤ 0.04) compared with HIV- youth. Among YLPHIV, soluble CD14 remained associated with endothelial dysfunction after adjusting for age, sex, Tanner stage, and antiretroviral therapy duration (β: -0.05, P = 0.01). CONCLUSION Despite viral suppression, South African YLPHIV have poor endothelial function and persistent evidence of monocyte activation and gut barrier dysfunction compared with HIV- youth. The long-term clinical significance of gut integrity and monocyte activation needs to be further assessed in YLPHIV.
Collapse
|
114
|
Hsue PY, Ribaudo HJ, Deeks SG, Bell T, Ridker PM, Fichtenbaum C, Daar ES, Havlir D, Yeh E, Tawakol A, Lederman M, Currier JS, Stein JH. Safety and Impact of Low-dose Methotrexate on Endothelial Function and Inflammation in Individuals With Treated Human Immunodeficiency Virus: AIDS Clinical Trials Group Study A5314. Clin Infect Dis 2020; 68:1877-1886. [PMID: 30219823 DOI: 10.1093/cid/ciy781] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/07/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Chronic inflammation in treated HIV infection is associated with mortality and atherosclerotic cardiovascular disease (ASCVD). We evaluated the safety and potential efficacy of low-dose methotrexate (LDMTX) in treated HIV. METHODS This was a phase 2 randomized, double-blind, multicenter trial in adults ≥40 years old with treated HIV, with CD4+ T-cell count ≥400 cells/μL and with/at increased risk for ASCVD. Participants received LDMTX (5-15 mg/week) or placebo (plus folic acid) for 24 weeks and were followed for an additional 12 weeks. Primary endpoints were safety and brachial artery flow-mediated dilation (FMD). RESULTS The 176 participants (90% male) had a median (Q1, Q3) age of 54 (49, 59) years. LDMTX was associated with decreases in CD4+ T cells at week 24 and CD8+ T cells at weeks 8, 12, and 24. Eleven participants (12.8%) experienced safety events in the LDMTX group vs 5 (5.6%) in placebo (Δ = 7.2%, upper 1-sided 90% CI, 13.4%; Pnoninferiority = .037). Week 24 change in FMD was 0.47% with LDMTX and 0.09% with placebo (P = .55). No inflammatory markers changed differentially with LDMTX compared to placebo. CONCLUSIONS Adults with HIV and increased ASCVD risk treated with LDMTX had more safety events than with placebo, but the prespecified noninferiority margin of 15% was not exceeded. LDMTX had no significant effect on endothelial function or inflammatory biomarkers but was associated with a significant decrease in CD8+ T cells. The balance of risks and potential benefits of LDMTX in this population will require additional investigation. CLINICAL TRIALS REGISTRATION NCT01949116.
Collapse
Affiliation(s)
- Priscilla Y Hsue
- Department of Medicine, University of California, San Francisco School of Medicine, Boston, Massachusetts
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco School of Medicine, Boston, Massachusetts
| | - Tanvir Bell
- McGovern Medical School, University of Texas Health Science Center at Houston, Boston, Massachusetts
| | - Paul M Ridker
- Cardiology Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Eric S Daar
- David Geffen School of Medicine, University of California, Los Angeles, Boston
| | - Diane Havlir
- Department of Medicine, University of California, San Francisco School of Medicine, Boston, Massachusetts
| | - Eunice Yeh
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Ahmed Tawakol
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michael Lederman
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Judith S Currier
- David Geffen School of Medicine, University of California, Los Angeles, Boston
| | - James H Stein
- University of Wisconsin School of Medicine and Public Health, Madison
| |
Collapse
|
115
|
Dar T, Osborne MT, Abohashem S, Abbasi T, Choi KW, Ghoneem A, Naddaf N, Smoller JW, Pitman RK, Denninger JW, Shin LM, Fricchione G, Tawakol A. Greater Neurobiological Resilience to Chronic Socioeconomic or Environmental Stressors Associates With Lower Risk for Cardiovascular Disease Events. Circ Cardiovasc Imaging 2020; 13:e010337. [PMID: 32787499 PMCID: PMC7820711 DOI: 10.1161/circimaging.119.010337] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/13/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Chronic exposure to socioeconomic or environmental stressors associates with greater stress-related neurobiological activity (ie, higher amygdalar activity [AmygA]) and higher risk of major adverse cardiovascular events (MACE). However, among individuals exposed to such stressors, it is unknown whether neurobiological resilience (NBResilience, defined as lower AmygA despite stress exposure) lowers MACE risk. We tested the hypotheses that NBResilience protects against MACE, and that it does so through decreased bone marrow activity and arterial inflammation. METHODS Individuals underwent 18F-fluorodeoxyglucose positron emission tomography/computed tomography; AmygA, bone marrow activity, and arterial inflammation were quantified. Chronic socioeconomic and environmental stressors known to associate with AmygA and MACE (ie, transportation noise exposure, neighborhood median household income, and crime rate) were quantified. Heightened stress exposure was defined as exposure to at least one chronic stressor (ie, the highest tertile of noise exposure or crime or lowest tertile of income). MACE within 5 years of imaging was adjudicated. Relationships were evaluated using linear and Cox regression, Kaplan-Meier survival, and mediation analyses. RESULTS Of 254 individuals studied (median age [interquartile range]: 57 years [46-67], 36.7% male), 166 were exposed to at least one chronic stressor. Among stress-exposed individuals, 12 experienced MACE over a median follow-up of 3.75 years. Among this group, higher AmygA (ie, lower resilience) associated with higher bone marrow activity (standardized β [95% CI]: 0.192 [0.030-0.353], P=0.020), arterial inflammation (0.203 [0.055-0.351], P=0.007), and MACE risk (standardized hazard ratio [95% CI]: 1.927 [1.370-2.711], P=0.001). The effect of NBResilience on MACE risk was significantly mediated by lower arterial inflammation (P<0.05). CONCLUSIONS Among individuals who are chronically exposed to socioeconomic or environmental stressors, NBResilience (AmygA <1 SD above the mean) associates with a >50% reduction in MACE risk, potentially via reduced arterial inflammation. These data raise the possibility that enhancing NBResilience may decrease the burden of cardiovascular disease.
Collapse
Affiliation(s)
- Tawseef Dar
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Cardiology Division (T.D., M.T.O., S.A., T.A., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michael T Osborne
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Cardiology Division (T.D., M.T.O., S.A., T.A., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Medicine (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Shady Abohashem
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Cardiology Division (T.D., M.T.O., S.A., T.A., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Taimur Abbasi
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Cardiology Division (T.D., M.T.O., S.A., T.A., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Karmel W Choi
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (K.W.C., J.W.S.)
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston (K.W.C., J.W.S.)
- Stanley Center for Psychiatric Research, Broad Institute, Boston, MA (K.W.C., J.W.S.)
| | - Ahmed Ghoneem
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Nicki Naddaf
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Jordan W Smoller
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Roger K Pitman
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - John W Denninger
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (K.W.C., J.W.S.)
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston (K.W.C., J.W.S.)
- Stanley Center for Psychiatric Research, Broad Institute, Boston, MA (K.W.C., J.W.S.)
| | - Lisa M Shin
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Psychology, Tufts University, Medford, MA (L.M.S.)
| | - Gregory Fricchione
- Department of Psychiatry (K.W.C., J.W.S., R.K.P., J.W.D., L.M.S., G.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center (T.D., M.T.O., S.D., T.A., A.G., N.N., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Cardiology Division (T.D., M.T.O., S.A., T.A., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Medicine (M.T.O., A.T.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
116
|
Hanna DB, Ramaswamy C, Kaplan RC, Kizer JR, Daskalakis D, Anastos K, Braunstein SL. Sex- and Poverty-Specific Patterns in Cardiovascular Disease Mortality Associated With Human Immunodeficiency Virus, New York City, 2007-2017. Clin Infect Dis 2020; 71:491-498. [PMID: 31504325 PMCID: PMC7384322 DOI: 10.1093/cid/ciz852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) may affect the risk of death due to cardiovascular disease (CVD) differently in men versus women. METHODS We examined CVD mortality rates between 2007 and 2017 among all New York City residents living with HIV and aged 13+ by sex, using data from city HIV surveillance and vital statistics and the National Death Index. Residents without HIV were enumerated using modified US intercensal estimates. We determined associations of HIV status with CVD mortality by sex and neighborhood poverty, defined as the percent of residents living below the federal poverty level, after accounting for age, race/ethnicity, and year. RESULTS There were 3234 CVD deaths reported among 147 915 New Yorkers living with HIV, with the proportion of deaths due to CVD increasing from 11% in 2007 to 22% in 2017. The age-standardized CVD mortality rate was 2.7/1000 person-years among both men and women with HIV. The relative rate of CVD mortality associated with HIV status was significantly higher among women (adjusted rate ratio [aRR] 1.7, 95% confidence interval [CI] 1.6-1.8) than men (aRR 1.2, 95% CI 1.1-1.3) overall, and within strata defined by neighborhood poverty. Sex differences in CVD mortality rates were the greatest when comparing individuals living with HIV and having detectable HIV RNA and CD4+ T-cell counts <500 cells/uL with individuals living without HIV. CONCLUSIONS Among people with HIV, 1 in 5 deaths is now associated with CVD. HIV providers should recognize the CVD risk among women with HIV, and reinforce preventive measures (eg, smoking cessation, blood pressure control, lipid management) and viremic control among people living with HIV regardless of neighborhood poverty to reduce CVD mortality.Human immunodeficiency virus (HIV) increases cardiovascular disease mortality risks to a greater degree among women than men, even after accounting for neighborhood poverty. HIV providers should emphasize cardiovascular disease prevention (eg, smoking cessation, hypertension control, lipid management) and viremic control.
Collapse
Affiliation(s)
- David B Hanna
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, USA
| | - Chitra Ramaswamy
- Bureau of Human Immunodeficiency Virus Prevention and Control, New York City Department of Health and Mental Hygiene, New York, USA
| | - Robert C Kaplan
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, University of California San Francisco, San Francisco, California, USA; and Departments of
- Medicine and, University of California San Francisco, San Francisco, California, USA
- Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Demetre Daskalakis
- Division of Disease Control, New York City Department of Health and Mental Hygiene, New York
| | - Kathryn Anastos
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, USA
| | - Sarah L Braunstein
- Bureau of Human Immunodeficiency Virus Prevention and Control, New York City Department of Health and Mental Hygiene, New York, USA
| |
Collapse
|
117
|
Bami K, Tewari S, MacPherson PA, Corrales-Medina VF, Verma S, Yanagawa B, Ruel M, Dwivedi G. Knowledge and Attitudes of Canadian Cardiac Surgeons Regarding Patients With Human Immunodeficiency Virus. Ann Thorac Surg 2020; 111:945-950. [PMID: 32710847 DOI: 10.1016/j.athoracsur.2020.05.133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/06/2020] [Accepted: 05/13/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Current data on cardiac surgical practices for people living with human immunodeficiency virus (HIV) are lacking. We hypothesized that cardiac surgeons would consider people living with HIV as candidates for the full scope of cardiac surgery, including heart transplant for these patients. METHODS We conducted a prospective survey of 155 cardiac surgeons across Canada to evaluate their current clinical perceptions regarding cardiac surgery in people living with HIV. Specifically, we evaluated their assessment of eligibility toward a wide scope of cardiac surgeries by using representative clinical scenarios. RESULTS A total of 63 surgeon responses (40.6%) were completed. The majority of surgeons agreed that a 50-year-old man with HIV and no other comorbidities, who had been receiving combination antiretroviral therapy for 5 years with an undetectable viral load since starting therapy and a CD4 count greater than 350 cells/μL, would be a candidate for valve replacement (73%), valve repair surgery (74.6%), or coronary artery bypass graft surgery (79.4%). Few surgeons believed that this patient would be eligible for cardiac transplantation (7.9%) or could be a cardiac transplant donor (1.6%). There was clinical equipoise over the eligibility for ventricular assist device surgery. CONCLUSIONS A majority of cardiac surgeons would perform coronary artery bypass graft surgery or valve surgery on patients with controlled HIV, but most consider HIV status as a prohibitive risk factor for cardiac transplantation. Although this may represent an opportunity for continuing medical education for cardiac surgeons, it also highlights the need for contemporary, high-quality evidence in this patient population.
Collapse
Affiliation(s)
- Karan Bami
- Division of Cardiology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shrankhala Tewari
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Paul A MacPherson
- Division of Infectious Disease, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Vicente F Corrales-Medina
- Division of Infectious Disease, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael's Hospital, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Bobby Yanagawa
- Division of Cardiac Surgery, St Michael's Hospital, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Marc Ruel
- Division of Cardiac Surgery, Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Girish Dwivedi
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada; Harry Perkins Institute of Medical Research and Fiona Stanley Hospital (Murdoch), University of Western Australia, Nedlands, Western Australia, Australia.
| |
Collapse
|
118
|
Lawal IO, Stoltz AC, Sathekge MM. Molecular imaging of cardiovascular inflammation and infection in people living with HIV infection. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00370-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
119
|
Heart Failure among People with HIV: Evolving Risks, Mechanisms, and Preventive Considerations. Curr HIV/AIDS Rep 2020; 16:371-380. [PMID: 31482297 DOI: 10.1007/s11904-019-00458-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE People with HIV (PHIV) with access to modern antiretroviral therapy (ART) face a two-fold increased risk of heart failure as compared with non-HIV-infected individuals. The purpose of this review is to consider evolving risks, mechanisms, and preventive considerations pertaining to heart failure among PHIV. RECENT FINDINGS While unchecked HIV/AIDS has been documented to precipitate heart failure characterized by overtly reduced cardiac contractile function, ART-treated HIV may be associated with either heart failure with reduced ejection fraction (HFrEF) or with heart failure with preserved ejection fraction (HFpEF). In HFpEF, a "stiff" left ventricle cannot adequately relax in diastole-a condition known as diastolic dysfunction. Diastolic dysfunction, in turn, may result from processes including myocardial fibrosis (triggered by hypertension and/or immune activation/inflammation) and/or myocardial steatosis (triggered by metabolic dysregulation). Notably, hypertension, systemic immune activation, and metabolic dysregulation are all common conditions among even those PHIV who are well-treated with ART. Of clinical consequence, HFpEF is uniquely intransigent to conventional medical therapies and portends high morbidity and mortality. However, diastolic dysfunction is reversible-as are contributing processes of myocardial fibrosis and myocardial steatosis. Our challenges in preserving myocardial health among PHIV are two-fold. First, we must continue working to realize UNAIDS 90-90-90 goals. This achievement will reduce AIDS-related mortality, including cardiovascular deaths from AIDS-associated heart failure. Second, we must work to elucidate the detailed mechanisms continuing to predispose ART-treated PHIV to heart failure and particularly HFpEF. Such efforts will enable the development and implementation of targeted preventive strategies.
Collapse
|
120
|
Abstract
PURPOSE OF REVIEW People infected with HIV through injection drug use are more likely to experience progression to AIDS, death due to AIDS, and all-cause mortality even when controlling for access to care and antiretroviral therapy. While high-risk behavior and concurrent infections most certainly are contributors, chronic immune activation, downstream metabolic comorbidities may play an important role. RECENT FINDINGS Altered intestinal integrity plays a major role in HIV-related immune activation and microbial translocation markers are heightened in active heroin users. Additionally, greater injection frequency drives systemic inflammation and is associated with HIV viral rebound. Finally, important systemic inflammation markers have been linked with frailty and mortality in people who inject drugs with and without concurrent HIV infection. Heroin use may work synergistically with HIV infection to cause greater immune activation than either factor alone. Further research is needed to understand the impact on downstream metabolic comorbidities including cardiovascular disease. Medication-assisted treatment for opioid use disorder with methadone or buprenorphine may ameliorate some of this risk; however, there is presently limited research in humans, including in non-HIV populations, describing changes in immune activation on these treatments which is of paramount importance for those with HIV infection.
Collapse
|
121
|
Brusca RM, Hanna DB, Wada NI, Blankson JN, Witt MD, Jacobson LP, Kingsley L, Palella FJ, Budoff M, Brown TT, Anastos K, Lazar JM, Mack WJ, Bacchetti P, Tien PC, Golzar Y, Plankey M, Golub E, Kaplan RC, Post WS. Subclinical cardiovascular disease in HIV controller and long-term nonprogressor populations. HIV Med 2020; 21:217-227. [PMID: 31729142 PMCID: PMC7069771 DOI: 10.1111/hiv.12820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Elite controllers (ECs), viraemic controllers (VCs), and long-term nonprogressors (LTNPs) control HIV viral replication or maintain CD4 T-cell counts without antiretroviral therapy, but may have increased cardiovascular disease (CVD) risk compared to HIV-uninfected persons. We evaluated subclinical carotid and coronary atherosclerosis and inflammatory biomarker levels among HIV controllers, LTNPs and noncontrollers and HIV-uninfected individuals in the Multicenter AIDS Cohort Study (MACS) and the Women's Interagency HIV Study (WIHS). METHODS We measured carotid plaque presence and common carotid artery intima-media thickness (IMT) in 1729 women and 1308 men, and the presence of coronary artery calcium and plaque in a subgroup of men. Associations between HIV control category and carotid and coronary plaque prevalences were assessed by multivariable regression analyses adjusting for demographics and CVD risk factors. Serum inflammatory biomarker concentrations [soluble CD163 (sCD163), soluble CD14 (sCD14), galectin-3 (Gal-3), galectin-3 binding protein (Gal-3BP) and interleukin (IL)-6] were measured and associations with HIV control category assessed. RESULTS We included 135 HIV controllers (30 ECs) and 135 LTNPs in the study. Carotid plaque prevalence and carotid IMT were similar in HIV controllers, LTNPs and HIV-uninfected individuals. HIV controllers and LTNPs had lower prevalences of carotid plaque compared to viraemic HIV-infected individuals. The prevalence of coronary atherosclerosis was similar in HIV controllers/LTNPs compared to HIV-uninfected and viraemic HIV-infected men. Controllers and LTNPs had higher concentrations of sCD163 and sCD14 compared to HIV-uninfected persons. CONCLUSIONS Subclinical CVD was similar in HIV controllers, LTNPs and HIV-uninfected individuals despite elevated levels of some inflammatory biomarkers. Future studies of HIV controllers and LTNPs are needed to characterize the risk of CVD among HIV-infected persons.
Collapse
Affiliation(s)
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolas I. Wada
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | - Mallory D. Witt
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | | | | | | | - Matthew Budoff
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Todd T. Brown
- Johns Hopkins University School of Medicine, Baltimore MD
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Montefiore Medical Center, Bronx, NY
| | - Jason M. Lazar
- Department of Medicine, SUNY-Downstate Medical Center, Brooklyn, NY
| | - Wendy J. Mack
- Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Phyllis C. Tien
- Department of Medicine and Clinical Pharmacy, University of California, San Francisco, CA and Department of Veterans Affairs Medical Center, San Francisco, CA
| | | | - Michael Plankey
- Department of Medicine, Division of Infectious Diseases, Georgetown University Medical Center, Washington, DC
| | - Elizabeth Golub
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Wendy S. Post
- Johns Hopkins University School of Medicine, Baltimore MD
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
122
|
Monocytes from men living with HIV exhibit heightened atherogenic potential despite long-term viral suppression with antiretroviral therapy. AIDS 2020; 34:513-518. [PMID: 32108672 DOI: 10.1097/qad.0000000000002460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE People living with HIV have an increased risk of cardiovascular disease (CVD) despite effective antiretroviral therapy (ART). Monocytes play a key role in the early stages of atherosclerosis-driven CVD by forming lipid-laden foam cells within artery walls. HIV infection potentiates foam cell formation ex vivo, but the mechanisms contributing to this are not known. METHODS We investigated the atherosclerosis-promoting potential of monocytes from 39 virologically suppressed men living with HIV (MLHIV) on ART and no evidence of CVD, and 25 HIV-uninfected controls of comparable age, sex, smoking status and CVD risk. RESULTS Despite absence of clinical atherosclerosis in both MLHIV and uninfected cohorts (evidenced by a carotid intima-media thickness of 0.6 mm for both groups; P = 0.254), monocytes from MLHIV showed increased potential to form atherosclerosis-promoting foam cells compared with controls in an ex-vivo assay (36.6% vs. 27.6%, respectively, P = 0.003). Consistent with observations of persistent inflammation and immune/endothelial activation in ART-treated HIV infection, levels of soluble tumour necrosis factor receptor II, CXCL10 and soluble VCAM-1 were elevated in MLHIV (P ≤ 0.005 for all), but were not significantly associated with foam cell formation. Foam cell formation was associated with an impaired ability of monocytes to undergo reverse transmigration, and a reduced ability to efflux cholesterol ex vivo (P < 0.05 for both). Importantly, foam cell formation declined significantly with duration of viral suppression (P = 0.004). CONCLUSION These findings highlight the persistence of HIV-related changes to the atherogenic potential of monocytes despite long-term viral suppression, and provide insights into mechanisms potentially driving increased CVD in ART-treated HIV infection.
Collapse
|
123
|
Mabuchi S, Komura N, Sasano T, Shimura K, Yokoi E, Kozasa K, Kuroda H, Takahashi R, Kawano M, Matsumoto Y, Kato H, Hatazawa J, Kimura T. Pretreatment tumor-related leukocytosis misleads positron emission tomography-computed tomography during lymph node staging in gynecological malignancies. Nat Commun 2020; 11:1364. [PMID: 32170086 PMCID: PMC7069975 DOI: 10.1038/s41467-020-15186-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 02/11/2020] [Indexed: 01/08/2023] Open
Abstract
The accuracy of fluorine-18-fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG-PET/CT) can be influenced by the increased glycolytic activity of inflammatory lesions. Here, using clinical data obtained from gynecological cancer patients, tumor samples and animal models, we investigate the impact of pretreatment tumor-related leukocytosis (TRL) on the diagnostic performance of 18F-FDG-PET/CT in detecting pelvic and paraaortic lymph node metastasis. We demonstrate that pretreatment TRL misleads 18F-FDG-PET/CT during lymph node staging in gynecological malignancies. In the mechanistic investigations, we show that the false-positive 18F-FDG-PET/CT result for detecting nodal metastasis can be reproduced in animal models of TRL-positive cancer bearing G-CSF expressing cervical cancer cells. We also show that increased 18F-FDG uptake in non-metastatic nodes can be explained by the MDSC-mediated premetastatic niche formation in which proinflammatory factors, such as S100A8 or S100A9, are abundantly expressed. Together, our results suggest that the MDSC-mediated premetastatic niche created in the lymph node of TRL-positive patients misleads 18F-FDG-PET/CT for detecting nodal metastasis.
Collapse
Affiliation(s)
- Seiji Mabuchi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan. .,Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara, Japan.
| | - Naoko Komura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomoyuki Sasano
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kotaro Shimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Eriko Yokoi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Katsumi Kozasa
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromasa Kuroda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryoko Takahashi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mahiru Kawano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuri Matsumoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroki Kato
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
124
|
Sinha A, Feinstein M. Epidemiology, pathophysiology, and prevention of heart failure in people with HIV. Prog Cardiovasc Dis 2020; 63:134-141. [PMID: 31987806 PMCID: PMC7237287 DOI: 10.1016/j.pcad.2020.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 12/21/2022]
Abstract
Heart failure (HF) has been a known complication of HIV/AIDS for three decades. As the treatment of HIV has changed, so has the epidemiology and pathophysiology of HF in people with HIV (PWH). Initial manifestations of HF in uncontrolled HIV primarily included a rapidly evolving cardiomyopathy with pericardial involvement. With the widespread uptake of effective antiretroviral therapy (ART), HF in PWH has become a chronic disease reflective of the aging population and associated comorbidities, albeit with a contribution from HIV-associated chronic immune dysregulation and inflammation. Despite viral suppression, PWH remain at elevated risk for both HF with reduced ejection fraction and HF with preserved ejection fraction. In this review, we discuss the changing epidemiology and mechanisms of HF in PWH and how that may inform HF prevention in this vulnerable population.
Collapse
Affiliation(s)
- Arjun Sinha
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, 60611; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 60611
| | - Matthew Feinstein
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, 60611; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 60611.
| |
Collapse
|
125
|
Abstract
PURPOSE OF REVIEW This article describes the use of biomarkers in expanding our understanding of chronic non-AIDS comorbidities among persons living with HIV (PLWH) receiving antiretroviral therapy (ART). RECENT FINDINGS We review current evidence that biomarkers of chronic immune activation and inflammation associate with a broad spectrum of end-organ diseases in PLWH. We discuss how ART may impact inflammation associated with HIV infection and the degree to which inflammation persists despite effective suppression of viral replication in plasma. We then discuss the limitations of the current literature, which lacks evidence of causality and disproportionately involves a few protein biomarkers that are unable to disentangle complex and overlapping biological pathways. SUMMARY Premature end-organ disease among PLWH has been repeatedly associated with higher levels of blood biomarkers reflecting inflammation and immune activation, which, despite viral suppression and CD4 T-cell increases after ART treatment, remain elevated relative to uninfected persons. There remain important unanswered questions with implications for the development of anti-inflammatory treatment strategies aimed at mitigating excess risk for end-organ comorbidities among PLWH.
Collapse
|
126
|
Titanji B, Gavegnano C, Hsue P, Schinazi R, Marconi VC. Targeting Inflammation to Reduce Atherosclerotic Cardiovascular Risk in People With HIV Infection. J Am Heart Assoc 2020; 9:e014873. [PMID: 31973607 PMCID: PMC7033865 DOI: 10.1161/jaha.119.014873] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Boghuma Titanji
- Division of Infectious Diseases Emory University School of Medicine Atlanta GA
| | - Christina Gavegnano
- Center for AIDS Research Laboratory of Biochemical Pharmacology Department of Pediatrics Emory University Atlanta GA
| | - Priscilla Hsue
- Department of Cardiology Zuckerberg San Francisco General Hospital University of California-San Francisco CA
| | - Raymond Schinazi
- Center for AIDS Research Laboratory of Biochemical Pharmacology Department of Pediatrics Emory University Atlanta GA
| | - Vincent C Marconi
- Division of Infectious Diseases Emory University School of Medicine Atlanta GA.,Emory Vaccine Center Atlanta GA.,Rollins School of Public Health Emory University Atlanta GA.,Atlanta VA Medical Center Decatur GA
| |
Collapse
|
127
|
Gupta SK, Liu Z, Sims EC, Repass MJ, Haneline LS, Yoder MC. Endothelial Colony-Forming Cell Function Is Reduced During HIV Infection. J Infect Dis 2020; 219:1076-1083. [PMID: 30239747 DOI: 10.1093/infdis/jiy550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/12/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) may be related to cardiovascular disease through monocyte activation-associated endothelial dysfunction. METHODS Blood samples from 15 HIV-negative participants (the uninfected group), 8 HIV-positive participants who were not receiving antiretroviral therapy (ART) (the infected, untreated group), and 15 HIV-positive participants who were receiving ART (the infected, treated group) underwent flow cytometry of endothelial colony-forming cells (ECFCs) and monocyte proportions. IncuCyte live cell imaging of 8 capillary proliferative capacity parameters were obtained from cord blood ECFCs treated with participant plasma. RESULTS The ECFC percentage determined by flow cytometry was not different between the study groups; however, values of the majority of capillary proliferative capacity parameters (ie, cell area, network length, network branch points, number of networks, and average tube width uniformity) were significantly lower in infected, untreated participants as compared to values for uninfected participants or infected, treated participants (P < .00625 for all comparisons). CD14+CD16+ intermediate monocytes and soluble CD163 were significantly and negatively correlated with several plasma-treated, cord blood ECFC proliferative capacity parameters in the combined HIV-positive groups but not in the uninfected group. CONCLUSIONS Cord blood ECFC proliferative capacity was significantly impaired by plasma from infected, untreated patients, compared with plasma from uninfected participants and from infected, treated participants. Several ECFC functional parameters were adversely associated with monocyte activation in the HIV-positive groups, thereby suggesting a mechanism by which HIV-related inflammation may impair vascular reparative potential and consequently increase the risk of cardiovascular disease during HIV infection.
Collapse
Affiliation(s)
- Samir K Gupta
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Ziyue Liu
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis
| | - Emily C Sims
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Matthew J Repass
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Laura S Haneline
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis
| |
Collapse
|
128
|
Kung BT, Seraj SM, Zadeh MZ, Rojulpote C, Kothekar E, Ayubcha C, Ng KS, Ng KK, Au-Yong TK, Werner TJ, Zhuang H, Hunt SJ, Hess S, Alavi A. An update on the role of 18F-FDG-PET/CT in major infectious and inflammatory diseases. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2019; 9:255-273. [PMID: 31976156 PMCID: PMC6971480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/27/2019] [Indexed: 06/10/2023]
Abstract
For decades, conventional nuclear medicine techniques have been utilized for the assessment of many infectious and inflammatory diseases. Most of these techniques have limitations such as the relatively low spatial resolution, being time consuming and low sensitivity or specificity. In recent years, FDG-PET/CT has shown promising role in the management of such diseases. An expanding set of studies illustrate the multifarious roles of FDG-PET/CT in the assessment of these conditions, both systemic diseases and more regional. Specifically, PET can provide vital information at a molecular level and consequently detect the disease activity at their earliest manifestation. With the continuing research on the diagnosis and treatment monitoring of patients with infectious and inflammatory diseases, the role of PET/CT can be further extended.
Collapse
Affiliation(s)
- Boom Ting Kung
- Clinical PET Centre and Nuclear Medicine Unit, Queen Elizabeth HospitalHong Kong, China
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
- Department of Radiology, Children’s Hospital of PhiladelphiaPA, USA
| | | | - Mahdi Zirakchian Zadeh
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
- Department of Radiology, Children’s Hospital of PhiladelphiaPA, USA
| | | | - Esha Kothekar
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
| | - Cyrus Ayubcha
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
| | - Kwok Sing Ng
- Clinical PET Centre and Nuclear Medicine Unit, Queen Elizabeth HospitalHong Kong, China
| | - Koon Kiu Ng
- Clinical PET Centre and Nuclear Medicine Unit, Queen Elizabeth HospitalHong Kong, China
| | - Ting Kun Au-Yong
- Clinical PET Centre and Nuclear Medicine Unit, Queen Elizabeth HospitalHong Kong, China
| | - Thomas J Werner
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
| | - Hongming Zhuang
- Department of Radiology, Children’s Hospital of PhiladelphiaPA, USA
| | - Stephen J Hunt
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
| | - Søren Hess
- Department of Radiology and Nuclear Medicine, Hospital of South West JutlandEsbjerg, Denmark
- Department of Regional Health Research, Faculty of Health Sciences, University of Southern DenmarkOdense, Denmark
| | - Abass Alavi
- Department of Radiology, Hospital of University of PennsylvaniaPA, USA
| |
Collapse
|
129
|
Kruize Z, Kootstra NA. The Role of Macrophages in HIV-1 Persistence and Pathogenesis. Front Microbiol 2019; 10:2828. [PMID: 31866988 PMCID: PMC6906147 DOI: 10.3389/fmicb.2019.02828] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
Current antiretroviral therapy (ART) effectively suppresses Human Immunodeficiency Virus type 1 (HIV-1) in infected individuals. However, even long term ART does not eradicate HIV-1 infected cells and the virus persists in cellular reservoirs. Beside memory CD4+ T cells, cells of the myeloid lineage, especially macrophages, are believed to be an important sanctuary for HIV-1. Monocytes and macrophages are key players in the innate immune response to pathogens and are recruited to sites of infection and inflammation. Due to their long life span and ability to reside in virtually every tissue, macrophages have been proposed to play a critical role in the establishment and persistence of the HIV-1 reservoir. Current HIV-1 cure strategies mainly focus on the concept of “shock and kill” to purge the viral reservoir. This approach aims to reactivate viral protein production in latently infected cells, which subsequently are eliminated as a consequence of viral replication, or recognized and killed by the immune system. Macrophage susceptibility to HIV-1 infection is dependent on the local microenvironment, suggesting that molecular pathways directing differentiation and polarization are involved. Current latency reversing agents (LRA) are mainly designed to reactivate the HIV-1 provirus in CD4+ T cells, while their ability to abolish viral latency in macrophages is largely unknown. Moreover, the resistance of macrophages to HIV-1 mediated kill and the presence of infected macrophages in immune privileged regions including the central nervous system (CNS), may pose a barrier to elimination of infected cells by current “shock and kill” strategies. This review focusses on the role of monocytes/macrophages in HIV-1 persistence. We will discuss mechanisms of viral latency and persistence in monocytes/macrophages. Furthermore, the role of these cells in HIV-1 tissue distribution and pathogenesis will be discussed.
Collapse
Affiliation(s)
- Zita Kruize
- Laboratory for Viral Immune Pathogenesis, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Neeltje A Kootstra
- Laboratory for Viral Immune Pathogenesis, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
130
|
Abstract
OBJECTIVE Individuals with HIV suffer a higher burden of cardiovascular diseases. Traditional cardiovascular risk scores consistently underestimate cardiovascular risk in this population. Subsets of microRNAs (miRNAs) are differentially expressed among individuals with cardiovascular disease and individuals infected with HIV. However, no study has clarified whether specific miRNAs may be biomarkers for cardiovascular disease in individuals with HIV. DESIGN/METHODS We compared the miRNA expression profiles of 34 HIV-positive individuals who had experienced clinically adjudicated type I myocardial infarctions (MI) with the profiles of 76 HIV-positive controls matched by traditional cardiovascular risk factors and HIV-specific measures. Using the elastic net algorithm, we selected miRNAs most strongly associated with incident MI and then used conditional Cox proportional hazards regression and cross-validation to evaluate miRNAs and their association with incident MI. We evaluated whether miRNA markers would improve risk classification relative to the Framingham Risk Score. RESULTS Higher miR-125a-5p and miR-139-5p expression levels were each associated with increased risk of developing MI after adjustment for Framingham Risk Score and HIV-related factors (hazard ratio 2.43, P = 0.018; hazard ratio 2.13, P = 0.048, respectively). Compared with the Framingham Risk Score alone, adding expression levels of miR-125a-5p or miR-139-5p resulted in an integrated discrimination improvement of 10.1 or 5.8%, respectively. CONCLUSION MiR-125a-5p and miR-139-5p, transcripts known to be differentially expressed in HIV-positive individuals, may serve as unique biomarkers predictive of cardiovascular disease in these patients and may help clarify processes because of HIV infection that contribute to cardiovascular disorders in this population.
Collapse
|
131
|
Abstract
Antiretroviral therapy has largely transformed HIV infection into a chronic disease condition. As such, physicians and other providers caring for individuals living with HIV infection need to be aware of the potential cardiovascular complications of HIV infection and the nuances of how HIV infection increases the risk of cardiovascular diseases, including acute myocardial infarction, stroke, peripheral artery disease, heart failure and sudden cardiac death, as well as how to select available therapies to reduce this risk. In this Review, we discuss the epidemiology and clinical features of cardiovascular disease, with a focus on coronary heart disease, in the setting of HIV infection, which includes a substantially increased risk of myocardial infarction even when the HIV infection is well controlled. We also discuss the mechanisms underlying HIV-associated atherosclerotic cardiovascular disease, such as the high rates of traditional cardiovascular risk factors in patients with HIV infection and HIV-related factors, including the use of antiretroviral therapy and chronic inflammation in the setting of effectively treated HIV infection. Finally, we highlight available therapeutic strategies, as well as approaches under investigation, to reduce the risk of cardiovascular disease and lower inflammation in patients with HIV infection.
Collapse
Affiliation(s)
- Priscilla Y Hsue
- University of California-San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA.
| | - David D Waters
- University of California-San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
132
|
Cedarbaum E, Ma Y, Scherzer R, Price JC, Adimora AA, Bamman M, Cohen M, Fischl MA, Matsushita K, Ofotokun I, Plankey M, Seaberg EC, Yin MT, Grunfeld C, Vartanian S, Sharma A, Tien PC. Contributions of HIV, hepatitis C virus, and traditional vascular risk factors to peripheral artery disease in women. AIDS 2019; 33:2025-2033. [PMID: 31335806 PMCID: PMC6774831 DOI: 10.1097/qad.0000000000002319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES HIV and hepatitis C virus (HCV) have been associated with cardiovascular disease (CVD), but it is unclear whether HIV and HCV are also associated with peripheral artery disease (PAD). We examined the association of HIV, HCV, and traditional CVD risk factors with PAD in the Women's Interagency HIV Study, a multicenter US cohort. METHODS In this cross-sectional study, ankle-brachial index was estimated using Doppler ultrasound and manual sphygmomanometer in 1899 participants aged more than 40 years with HIV/HCV coinfection, HCV or HIV monoinfection, or neither infection. Multivariable logistic regression was used to estimate the odds of PAD (ankle-brachial index ≤0.9) after controlling for demographic, behavioral, and CVD risk factors. RESULTS Over two-thirds were African-American, median age was 50 years, and PAD prevalence was 7.7% with little difference by infection status. After multivariable adjustment, neither HIV nor HCV infection was associated with greater odds of PAD. Factors associated with PAD included older age [adjusted odds ratio (aOR): 2.01 for age 61-70 vs. 40-50 years; 95% confidence interval (CI): 1.04, 3.87], Black race (aOR: 2.30; 95% CI: 1.15, 4.63), smoking (aOR: 1.27 per 10-pack-year increment; 95% CI: 1.09, 1.48), and higher SBP (aOR: 1.14 per 10 mmHg; 95% CI: 1.01, 1.28). CONCLUSION The high PAD prevalence in this nationally representative cohort of women with or at risk for HIV is on par with general population studies in individuals a decade older than our study's median age. HIV and HCV infection are not associated with greater PAD risk relative to uninfected women with similar risk factors. Modifiable traditional CVD risk factors may be important early intervention targets in women with and at risk for HIV.
Collapse
Affiliation(s)
- Emily Cedarbaum
- aDepartment of Medicine, University of California, San Francisco bMedical Service, Department of Veterans Affairs Medical Center, San Francisco, California cDepartment of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina dDepartment of Cell, Developmental, and Integrative Biology eDepartment of Medicine fDepartment of Neurology, University of Alabama, Birmingham, Alabama gDepartment of Medicine, Cook County Health and Hospitals System, Chicago, Illinois hDepartment of Medicine, University of Miami, Miami, Florida iDepartment of Epidemiology, Johns Hopkins University, Baltimore, Maryland jDepartment of Medicine, Emory University, Atlanta, Georgia kDepartment of Medicine, Georgetown University Medical Center, Washington, District of Columbia lDepartment of Medicine, Columbia University, New York, New York mDepartment of Surgery, University of California, San Francisco, San Francisco, California nDepartment of Medicine, Albert Einstein College of Medicine, The Bronx, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Jabs DA, Van Natta ML, Trang G, Jones N, Milush JM, Cheu R, Klatt NR, Pak JW, Danis RP, Hunt PW. Association of Systemic Inflammation With Retinal Vascular Caliber in Patients With AIDS. Invest Ophthalmol Vis Sci 2019; 60:2218-2225. [PMID: 31108552 PMCID: PMC6528842 DOI: 10.1167/iovs.18-26070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose To evaluate relationships among retinal vascular caliber and biomarkers of systemic inflammation in patients with AIDS. Methods A total of 454 participants with AIDS had retinal vascular caliber (central retinal artery equivalent and central retinal vein equivalent) determined from enrollment retinal photographs by reading center graders masked to clinical and biomarker information. Cryopreserved plasma specimens were assayed for inflammatory biomarkers, including C-reactive protein (CRP), IL-6, interferon-γ inducible protein (IP)-10, kynurenine/tryptophan (KT) ratio, and intestinal fatty acid binding protein (I-FABP). Results In the simple linear regression of retinal vascular caliber on plasma biomarkers, elevated CRP, IL-6, and IP-10 were associated with retinal venular dilation, and elevated KT ratio with retinal arteriolar narrowing. In the multiple linear regression, including baseline characteristics and plasma biomarkers, AMD was associated with dilation of retinal arterioles (mean difference: 9.1 μm; 95% confidence interval [CI] 5.2, 12.9; P < 0.001) and venules (mean difference, 10.9 μm; 95% CI, 5.3, 16.6; P < 0.001), as was black race (P < 0.001). Hyperlipidemia was associated with retinal venular narrowing (mean difference, -7.5 μm; 95% CI, -13.7, -1.2; P = 0.02); cardiovascular disease with arteriolar narrowing (mean difference, -5.2 μm; 95% CI, -10.3, -0.1; P = 0.05); age with arteriolar narrowing (slope, -0.26 μm/year; 95% CI, -0.46, -0.06; P = 0.009); and IL-6 with venular dilation (slope, 5.3 μm/standard deviation log10[plasma IL-6 concentration]; 95% CI, 2.7, 8.0; P < 0.001). Conclusions These data suggest that retinal vascular caliber is associated with age, race, AMD, hyperlipidemia, cardiovascular disease, and selected biomarkers of systemic inflammation.
Collapse
Affiliation(s)
- Douglas A Jabs
- Department of Ophthalmology, the Icahn School of Medicine at Mount Sinai, New York, New York, United States.,Department of Medicine, the Icahn School of Medicine at Mount Sinai, New York, New York, United States.,Department of Epidemiology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States
| | - Mark L Van Natta
- Department of Epidemiology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States
| | - Garrett Trang
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California, United States
| | - Norman Jones
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California, United States
| | - Jeffrey M Milush
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California, United States
| | - Ryan Cheu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States
| | - Nichole R Klatt
- Department of Pediatrics, the University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jeong Won Pak
- Department of Ophthalmology and Visual Sciences, The University of Wisconsin, Madison, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Ronald P Danis
- Department of Ophthalmology and Visual Sciences, The University of Wisconsin, Madison, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Peter W Hunt
- Department of Medicine, the University of California, San Francisco, School of Medicine, San Francisco, California, United States
| |
Collapse
|
134
|
Wan Yusuf WN, Wan Mohammad WMZ, Gan SH, Mustafa M, Abd Aziz CB, Sulaiman SA. Tualang honey ameliorates viral load, CD4 counts and improves quality of life in asymptomatic human immunodeficiency virus infected patients. J Tradit Complement Med 2019; 9:249-256. [PMID: 31453119 PMCID: PMC6702152 DOI: 10.1016/j.jtcme.2018.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 12/28/2022] Open
Abstract
This is the first study to report on the effects of honey in asymptomatic HIV positive subjects in ameliorating CD4 count, viral load (VL) and quality of life (QOL). It is a randomized, controlled, open labelled study, comparing the effects of Tualang honey (TH) administration for six months at three different doses: 20 g (THL), 40 g (THI) or 60 g (THH) daily compared with control (no administered treatment, THC). Only asymptomatic HIV positive subjects (n=95) having CD4 count 250-600 cell/ml, not on antiretrovirals were enrolled. Blood, (together with QOL questionnaires administration) were investigated at baseline, three and six months (CD4 cell count) while VL was determined only at baseline and six months. Significant reductions in CD4 counts in THL and THC groups (p= 0.003 for both) were seen with no significant reductions in the CD4 counts in THI and THH groups (p=0.447 and 0.053 respectively). There was improvement in VL in THC and THI (130% and 32% respectively) and reductions in THL and THH (26% and 8% respectively). Within and between group analyses for VL indicated significant differences between THL and THH compared to THC. In addition, significant improvement in QOL of groups which received TH was noted. TH has the potential to improve the QOL (physical and psychological) and CD4 counts. There was a trend of lower VL in asymptomatic HIV subjects following TH administration thus supporting the possible role of TH in boosting the immune system by improving CD4 counts, causing VL reductions in HIV positive subjects.
Collapse
Affiliation(s)
- Wan Nazirah Wan Yusuf
- Pharmacology Department, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | | | - Siew Hua Gan
- School of Pharmacy, Building 2, Level 5, Room 40 (2-5-40), Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Mahiran Mustafa
- Infectious Disease Unit, Department of Medicine, Raja Perempuan Zainab II Hospital, 15586, Kota Bharu, Kelantan, Malaysia
| | - Che Badariah Abd Aziz
- Physiology Department, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Siti Amrah Sulaiman
- Pharmacology Department, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
135
|
A novel method to measure vascular inflammation by [18F]fluorodeoxyglucose PET/computed tomography scanning of the aorta. Nucl Med Commun 2019; 40:1087-1089. [PMID: 31517809 DOI: 10.1097/mnm.0000000000001055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
136
|
Henrich TJ, Hsue PY, VanBrocklin H. Seeing Is Believing: Nuclear Imaging of HIV Persistence. Front Immunol 2019; 10:2077. [PMID: 31572355 PMCID: PMC6751256 DOI: 10.3389/fimmu.2019.02077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/16/2019] [Indexed: 12/19/2022] Open
Abstract
A major obstacle to HIV eradication is the presence of infected cells that persist despite suppressive antiretroviral therapy (ART). HIV largely resides outside of the peripheral circulation, and thus, numerous anatomical and lymphoid compartments that have the capacity to harbor HIV are inaccessible to routine sampling. As a result, there is a limited understanding of the tissue burden of HIV infection or anatomical distribution of HIV transcriptional and translational activity. Novel, non-invasive, in vivo methods are urgently needed to address this fundamental gap in knowledge. In this review, we discuss past and current nuclear imaging approaches that have been applied to HIV infection with an emphasis on current strategies to implement positron emission tomography (PET)-based imaging to directly visualize and characterize whole-body HIV burden. These imaging approaches have various limitations, such as the potential for limited PET sensitivity and specificity in the setting of ART suppression or low viral burden. However, recent advances in high-sensitivity, total-body PET imaging platforms and development of new radiotracer technologies that may enhance anatomical penetration of target-specific tracer molecules are discussed. Potential strategies to image non-viral markers of HIV tissue burden or focal immune perturbation are also addressed. Overall, emerging nuclear imaging techniques and platforms may play an important role in the development of novel therapeutic and HIV reservoir eradication strategies.
Collapse
Affiliation(s)
- Timothy J Henrich
- Division of Experimental Medicine, Department of Medicine, University of San Francisco, San Francisco, CA, United States
| | - Priscilla Y Hsue
- Division of Cardiology, Department of Medicine, University of San Francisco, San Francisco, CA, United States
| | - Henry VanBrocklin
- Radiopharmaceutical Research Program, Center for Molecular and Functional Imaging, University of San Francisco, San Francisco, CA, United States
| |
Collapse
|
137
|
Kearns AC, Liu F, Dai S, Robinson JA, Kiernan E, Tesfaye Cheru L, Peng X, Gordon J, Morgello S, Abuova A, Lo J, Zanni MV, Grinspoon S, Burdo TH, Qin X. Caspase-1 Activation Is Related With HIV-Associated Atherosclerosis in an HIV Transgenic Mouse Model and HIV Patient Cohort. Arterioscler Thromb Vasc Biol 2019; 39:1762-1775. [PMID: 31315440 PMCID: PMC6703939 DOI: 10.1161/atvbaha.119.312603] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Atherosclerotic cardiovascular disease (ASCVD) is an increasing cause of morbidity and mortality in people with HIV since the introduction of combination antiretroviral therapy. Despite recent advances in our understanding of HIV ASCVD, controversy still exists on whether this increased risk of ASCVD is due to chronic HIV infection or other risk factors. Mounting biomarker studies indicate a role of monocyte/macrophage activation in HIV ASCVD; however, little is known about the mechanisms through which HIV infection mediates monocyte/macrophage activation in such a way as to engender accelerated atherogenesis. Here, we experimentally investigated whether HIV expression is sufficient to accelerate atherosclerosis and evaluated the role of caspase-1 activation in monocytes/macrophages in HIV ASCVD. Approach and Results: We crossed a well-characterized HIV mouse model, Tg26 mice, which transgenically expresses HIV-1, with ApoE-/- mice to promote atherogenic conditions (Tg26+/-/ApoE-/-). Tg26+/-/ApoE-/- have accelerated atherosclerosis with increased caspase-1 pathway activation in inflammatory monocytes and atherosclerotic vasculature compared with ApoE-/-. Using a well-characterized cohort of people with HIV and tissue-banked aortic plaques, we documented that serum IL (interleukin)-18 was higher in people with HIV compared with non-HIV-infected controls, and in patients with plaques, IL-18 levels correlated with monocyte/macrophage activation markers and noncalcified inflammatory plaques. In autopsy-derived aortic plaques, caspase-1+ cells and CD (clusters of differentiation) 163+ macrophages correlated. CONCLUSIONS These data demonstrate that expression of HIV is sufficient to accelerate atherogenesis. Further, it highlights the importance of caspase-1 and monocyte/macrophage activation in HIV atherogenesis and the potential of Tg26+/-/ApoE-/- as a tool for mechanistic studies of HIV ASCVD.
Collapse
Affiliation(s)
- Alison C. Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
- Co-first author, these authors contributed equally to this work
| | - Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA 70433
- Co-first author, these authors contributed equally to this work
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Jake A. Robinson
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Elizabeth Kiernan
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Lediya Tesfaye Cheru
- Program in Nutritional Metabolism, Mass General Hospital and Harvard Medical School, Boston, MA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Jennifer Gordon
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Susan Morgello
- Departments of Neurology, Neuroscience, and Pathology, Mount Sinai Medical Center, New York, NY
| | - Aishazhan Abuova
- Departments of Neurology, Neuroscience, and Pathology, Mount Sinai Medical Center, New York, NY
| | - Janet Lo
- Program in Nutritional Metabolism, Mass General Hospital and Harvard Medical School, Boston, MA
| | - Markella V. Zanni
- Program in Nutritional Metabolism, Mass General Hospital and Harvard Medical School, Boston, MA
| | - Steven Grinspoon
- Program in Nutritional Metabolism, Mass General Hospital and Harvard Medical School, Boston, MA
| | - Tricia H. Burdo
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA 19140
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA 70433
| |
Collapse
|
138
|
Babu H, Ambikan AT, Gabriel EE, Svensson Akusjärvi S, Palaniappan AN, Sundaraj V, Mupanni NR, Sperk M, Cheedarla N, Sridhar R, Tripathy SP, Nowak P, Hanna LE, Neogi U. Systemic Inflammation and the Increased Risk of Inflamm-Aging and Age-Associated Diseases in People Living With HIV on Long Term Suppressive Antiretroviral Therapy. Front Immunol 2019; 10:1965. [PMID: 31507593 PMCID: PMC6718454 DOI: 10.3389/fimmu.2019.01965] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
The ART program in low- and middle-income countries (LMIC) like India, follows a public health approach with a standardized regimen for all people living with HIV (PLHIV). Based on the evidence from high-income countries (HIC), the risk of an enhanced, and accentuated onset of premature-aging or age-related diseases has been observed in PLHIV. However, very limited data is available on residual inflammation and immune activation in the populations who are on first-generation anti-HIV drugs like zidovudine and lamivudine that have more toxic side effects. Therefore, the aim of the present study was to evaluate the levels of systemic inflammation and understand the risk of age-associated diseases in PLHIV on long-term suppressive ART using a large number of biomarkers of inflammation and immune activation. Blood samples were obtained from therapy naïve PLHIV (Pre-ART, n = 43), PLHIV on ART for >5 years (ART, n = 53), and HIV-negative healthy controls (HIVNC, n = 41). Samples were analyzed for 92 markers of inflammation, sCD14, sCD163, and telomere length. Several statistical tests were performed to compare the groups under study. Multivariate linear regression was used to investigate the associations. Despite a median duration of 8 years of successful ART, sCD14 (p < 0.001) and sCD163 (p = 0.04) levels continued to be significantly elevated in ART group as compared to HIVNC. Eleven inflammatory markers, including 4E-BP1, ADA, CCL23, CD5, CD8A, CST5, MMP1, NT3, SLAMF1, TRAIL, and TRANCE, were found to be significantly different (p < 0.05) between the groups. Many of these markers are associated with age-related co-morbidities including cardiovascular disease, neurocognitive decline and some of these markers are being reported for the first time in the context of HIV-induced inflammation. Linear regression analysis showed a significant negative association between HIV-1-positivity and telomere length (p < 0.0001). In ART-group CXCL1 (p = 0.048) and TGF-α (p = 0.026) showed a significant association with the increased telomere length and IL-10RA was significantly associated with decreased telomere length (p = 0.042). This observation warrants further mechanistic studies to generate evidence to highlight the need for enhanced treatment monitoring and special interventions in HIV-infected individuals.
Collapse
Affiliation(s)
- Hemalatha Babu
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anoop T. Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Erin E. Gabriel
- Department of Medical Epidemiology and Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Naveen Reddy Mupanni
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Narayanaiah Cheedarla
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | | | - Srikanth P. Tripathy
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Piotr Nowak
- Unit of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Luke Elizabeth Hanna
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
139
|
Lawal IO, Ankrah AO, Popoola GO, Lengana T, Sathekge MM. Arterial inflammation in young patients with human immunodeficiency virus infection: A cross-sectional study using F-18 FDG PET/CT. J Nucl Cardiol 2019; 26:1258-1265. [PMID: 29417417 DOI: 10.1007/s12350-018-1207-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND HIV infection is associated with the risk of development of atherosclerosis at a younger age. We compared arterial inflammation in HIV-infected and HIV-uninfected patients with otherwise low-risk factors for cardiovascular disease (CVD) using FDG PET/CT. METHODS 242 patients aged 18-40 years with low-risk factors for CVD consisting of 121 HIV-infected patients and 121 HIV-uninfected age- and gender-matched controls were studied, mean age = 34.95 ± 5.46 years. We calculated and compared the target-to-background ratio of FDG uptake in ascending aorta of HIV-infected and non-infected patients. RESULTS Median CD4 count and viral load were 375.5 cells/mm3 (range 2-1094) and 6391.00 copies/mL (range 24-1,348,622), respectively. There was slightly higher but significant overlap in the TBR between HIV-infected group compared with control (1.22, 0.87-2.02 vs. 1.12, 0.38-1.40, P < 0.001). TBR was neither affected by CD4 count levels nor the presence or absence of detectable viremia. We also found no significant difference in TBR between male and female patients with HIV infection. We found a weak positive correlation between TBR and CD4 count, TBR and duration of HIV infection, and a very weak negative correlation between TBR and viral load. There was no significant difference in TBR between patients on HAART and those not yet commenced on therapy. CONCLUSION Marginally higher TBR with a significant overlap exist in HIV-infected patients compared with control. Arterial F-18 FDG uptake is not affected by the CD 4 count, viral load, gender, or duration of HIV infection.
Collapse
Affiliation(s)
- Ismaheel O Lawal
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001, South Africa
| | - Alfred O Ankrah
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001, South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gbenga O Popoola
- Department of Epidemiology and Community Health, University of Ilorin, Ilorin, Nigeria
| | - Thabo Lengana
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001, South Africa
| | - Mike M Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001, South Africa.
| |
Collapse
|
140
|
Schoepf IC, Buechel RR, Kovari H, Hammoud DA, Tarr PE. Subclinical Atherosclerosis Imaging in People Living with HIV. J Clin Med 2019; 8:E1125. [PMID: 31362391 PMCID: PMC6723163 DOI: 10.3390/jcm8081125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
In many, but not all studies, people living with HIV (PLWH) have an increased risk of coronary artery disease (CAD) events compared to the general population. This has generated considerable interest in the early, non-invasive detection of asymptomatic (subclinical) atherosclerosis in PLWH. Ultrasound studies assessing carotid artery intima-media thickness (CIMT) have tended to show a somewhat greater thickness in HIV+ compared to HIV-, likely due to an increased prevalence of cardiovascular (CV) risk factors in PLWH. Coronary artery calcification (CAC) determination by non-contrast computed tomography (CT) seems promising to predict CV events but is limited to the detection of calcified plaque. Coronary CT angiography (CCTA) detects calcified and non-calcified plaque and predicts CAD better than either CAC or CIMT. A normal CCTA predicts survival free of CV events over a very long time-span. Research imaging techniques, including black-blood magnetic resonance imaging of the vessel wall and 18F-fluorodeoxyglucose positron emission tomography for the assessment of arterial inflammation have provided insights into the prevalence of HIV-vasculopathy and associated risk factors, but their clinical applicability remains limited. Therefore, CCTA currently appears as the most promising cardiac imaging modality in PLWH for the evaluation of suspected CAD, particularly in patients <50 years, in whom most atherosclerotic coronary lesions are non-calcified.
Collapse
Affiliation(s)
- Isabella C Schoepf
- University Department of Medicine and Infectious Diseases Service, Kantonsspital Baselland, University of Basel, 4101 Bruderholz, Switzerland
| | - Ronny R Buechel
- Department of Nuclear Medicine, Cardiac Imaging, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Helen Kovari
- Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, 8091 Zurich, Switzerland
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip E Tarr
- University Department of Medicine and Infectious Diseases Service, Kantonsspital Baselland, University of Basel, 4101 Bruderholz, Switzerland.
| |
Collapse
|
141
|
Stein JH, Yeh E, Weber JM, Korcarz C, Ridker PM, Tawakol A, Hsue PY, Currier JS, Ribaudo H, Mitchell CKC. Brachial Artery Echogenicity and Grayscale Texture Changes in HIV-Infected Individuals Receiving Low-Dose Methotrexate. Arterioscler Thromb Vasc Biol 2019; 38:2870-2878. [PMID: 30571173 DOI: 10.1161/atvbaha.118.311807] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective- We evaluated the biological effects of low-dose methotrexate on 3 novel brachial artery grayscale ultrasound measures that may indicate subclinical arterial injury. Approach and Results- Exploratory analysis from a clinical trial of people with HIV infection at increased cardiovascular disease risk who were randomly assigned to low-dose methotrexate (target dose 15 mg/wk) or placebo. Brachial artery ultrasound grayscale median, gray level difference statistic texture-contrast (GLDS-CON), and gray level texture entropy were measured at baseline and after 24 weeks of intervention. Findings from the intention-to-treat (N=148) and adequately-dosed (N=118) populations were consistent, so the adequately-dosed population results are presented. Participants were a median (Q1, Q3) age of 54 (50, 60) years. After 24 weeks, the low-dose methotrexate intervention was associated with a 25.4% (-18.1, 58.6; P=0.007) increase in GLDS-CON compared with 1.3% (-29.1, 44.7; P=0.97) with placebo ( P=0.05) and a 0.10 u (-0.06, 0.23; P=0.026) increase in entropy compared with 0.02 u (-0.11, 0.14; P=0.54) with placebo ( P=0.14). At week 24, changes in CD4+ T cells correlated inversely with changes in GLDS-CON (ρ=-0.20; P=0.031), and entropy (ρ=-0.21; P=0.023). Changes in D-dimer levels, but no other inflammatory biomarkers, also correlated inversely with changes in GLDS-CON (ρ=-0.23; P=0.014) and entropy (ρ=-0.26; P=0.005). Conclusions- Brachial artery GLDS-CON and entropy increased after 24 weeks of low-dose methotrexate, though the latter was not significantly different from placebo. Grayscale changes were associated with decreases in CD4+ T-cell and D-dimer concentrations and may indicate favorable arterial structure changes.
Collapse
Affiliation(s)
- James H Stein
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Eunice Yeh
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA (E.Y., H.R.)
| | - Joanne M Weber
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Claudia Korcarz
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Paul M Ridker
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (P.M.R., A.T.)
| | - Ahmed Tawakol
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (P.M.R., A.T.)
| | - Priscilla Y Hsue
- Department of Medicine, University of California-San Francisco School of Medicine (P.Y.H.)
| | - Judith S Currier
- Division of Infectious Diseases, David Geffen School of Medicine at University of California-Los Angeles (J.S.C.)
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA (E.Y., H.R.)
| | - Carol K C Mitchell
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| |
Collapse
|
142
|
Conceptualizing the Risks of Coronary Heart Disease and Heart Failure Among People Aging with HIV: Sex-Specific Considerations. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2019; 21:41. [DOI: 10.1007/s11936-019-0744-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
143
|
Feinstein MJ, Hsue PY, Benjamin L, Bloomfield GS, Currier JS, Freiberg MS, Grinspoon SK, Levin J, Longenecker CT, Post. WS. Characteristics, Prevention, and Management of Cardiovascular Disease in People Living With HIV: A Scientific Statement From the American Heart Association. Circulation 2019; 140:e98-e124. [PMID: 31154814 PMCID: PMC7993364 DOI: 10.1161/cir.0000000000000695] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As early and effective antiretroviral therapy has become more widespread, HIV has transitioned from a progressive, fatal disease to a chronic, manageable disease marked by elevated risk of chronic comorbid diseases, including cardiovascular diseases (CVDs). Rates of myocardial infarction, heart failure, stroke, and other CVD manifestations, including pulmonary hypertension and sudden cardiac death, are significantly higher for people living with HIV than for uninfected control subjects, even in the setting of HIV viral suppression with effective antiretroviral therapy. These elevated risks generally persist after demographic and clinical risk factors are accounted for and may be partly attributed to chronic inflammation and immune dysregulation. Data on long-term CVD outcomes in HIV are limited by the relatively recent epidemiological transition of HIV to a chronic disease. Therefore, our understanding of CVD pathogenesis, prevention, and treatment in HIV relies on large observational studies, randomized controlled trials of HIV therapies that are underpowered to detect CVD end points, and small interventional studies examining surrogate CVD end points. The purpose of this document is to provide a thorough review of the existing evidence on HIV-associated CVD, in particular atherosclerotic CVD (including myocardial infarction and stroke) and heart failure, as well as pragmatic recommendations on how to approach CVD prevention and treatment in HIV in the absence of large-scale randomized controlled trial data. This statement is intended for clinicians caring for people with HIV, individuals living with HIV, and clinical and translational researchers interested in HIV-associated CVD.
Collapse
Affiliation(s)
| | - Priscilla Y. Hsue
- University of California-San Francisco School of Medicine, San Francisco, CA
| | | | | | - Judith S. Currier
- University of California-Los Angeles School of Medicine, Los Angeles, CA
| | | | | | - Jules Levin
- National AIDS Treatment Advocacy Program, New York, NY
| | | | - Wendy S. Post.
- Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
144
|
Jaworowski A, Hearps AC, Angelovich TA, Hoy JF. How Monocytes Contribute to Increased Risk of Atherosclerosis in Virologically-Suppressed HIV-Positive Individuals Receiving Combination Antiretroviral Therapy. Front Immunol 2019; 10:1378. [PMID: 31275317 PMCID: PMC6593090 DOI: 10.3389/fimmu.2019.01378] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/31/2019] [Indexed: 12/27/2022] Open
Abstract
Combination antiretroviral therapy (ART) is effective at suppressing HIV viremia to achieve persistently undetectable levels in peripheral blood in the majority of individuals with access and ability to maintain adherence to treatment. However, evidence suggests that ART is less effective at eliminating HIV-associated inflammation and innate immune activation. To the extent that residual inflammation and immune activation persist, virologically suppressed people living with HIV (PLWH) may have increased risk of inflammatory co-morbidities, and adjunctive therapies may need to be considered to reduce HIV-related inflammation and fully restore the health of virologically suppressed HIV+ individuals. Cardiovascular disease (CVD) is the single leading cause of death in the developed world and is becoming more important in PLWH with access to ART. Arterial disease due to atherosclerosis, leading to acute myocardial infarction (AMI) and stroke, is a major component of CVD. Atherosclerosis is an inflammatory disease, and epidemiological comparisons of atherosclerosis and AMI show a higher prevalence and suggest a greater risk in PLWH compared to the general population. The reasons for greater prevalence of CVD in PLWH can be broadly grouped into four categories: (a) the higher prevalence of traditional risk factors e.g., smoking and hypertension (b) dyslipidemia (also a traditional risk factor) caused by off-target effects of ART drugs (c) HIV-related inflammation and immune activation and (d) other undefined HIV-related factors. Management strategies aimed at reducing the impact of traditional risk factors in PLWH are similar to those for the general population and their effectiveness is currently being evaluated. Together with improvements in ART regimens and guidelines for treatment, and a greater awareness of its impact on CVD, the HIV-related risk of AMI and stroke is decreasing but remains elevated compared to the general community. Monocytes are key effector cells which initiate the formation of atherosclerotic plaques by migrating into the intima of coronary arteries and accumulating as foam cells full of lipid droplets. This review considers the specific role of monocytes as effector cells in atherosclerosis which progresses to AMI and stroke, and explores mechanisms by which HIV may promote an atherogenic phenotype and function independent of traditional risk factors. Altered monocyte function may represent a distinct HIV-related factor which increases risk of CVD in PLWH.
Collapse
Affiliation(s)
- Anthony Jaworowski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia.,Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
| | - Anna C Hearps
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia.,Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
| | - Thomas A Angelovich
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,Life Sciences Discipline, Burnet Institute, Melbourne, VIC, Australia
| | - Jennifer F Hoy
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia
| |
Collapse
|
145
|
Abstract
PURPOSE OF REVIEW We aim to provide an in-depth review of recent literature highlighting the role of inflammation involving the adipose tissue, liver, skeletal muscles, and gastrointestinal tract in the development of metabolic complications among persons living with HIV (PLWH). RECENT FINDINGS Recent studies in PLWH have demonstrated a significant association between circulating inflammatory markers and development of insulin resistance and metabolic complications. In adipose tissue, pro-inflammatory cytokine expression inhibits adipocyte insulin signaling, which alters lipid and glucose homeostasis. Increased lipolysis and lipogenesis elevate levels of circulating free fatty acids and promote ectopic fat deposition in liver and skeletal muscles. This leads to lipotoxicity characterized by a pro-inflammatory response with worsening insulin resistance. Finally, HIV is associated with gastrointestinal tract inflammation and changes in the gut microbiome resulting in reduced diversity, which is an additional risk factor for diabetes. Metabolic complications in PLWH are in part due to chronic, multisite tissue inflammation resulting in dysregulation of glucose and lipid trafficking, utilization, and storage.
Collapse
|
146
|
Grinspoon SK, Fitch KV, Overton ET, Fichtenbaum CJ, Zanni MV, Aberg JA, Malvestutto C, Lu MT, Currier JS, Sponseller CA, Waclawiw M, Alston-Smith B, Cooper-Arnold K, Klingman KL, Desvigne-Nickens P, Hoffmann U, Ribaudo HJ, Douglas PS. Rationale and design of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE). Am Heart J 2019; 212:23-35. [PMID: 30928825 PMCID: PMC6535121 DOI: 10.1016/j.ahj.2018.12.016] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/15/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is more frequent among people with HIV (PWH) and may relate to traditional and nontraditional factors, including inflammation and immune activation. A critical need exists to develop effective strategies to prevent CVD in this population. METHODS The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) (A5332) is a prospective, randomized, placebo-controlled trial of a statin strategy for the primary prevention of major adverse cardiovascular events (MACE) in PWH with low to moderate traditional risk. At least 7,500 PWH, 40-75 years of age, on stable antiretroviral therapy, will be randomized to pitavastatin calcium (4 mg/d) or identical placebo and followed for up to 8 years. Participants are enrolled based on the 2013 American College of Cardiology (ACC)/American Heart Association (AHA) atherosclerotic cardiovascular disease (ASCVD) risk score and low-density lipoprotein cholesterol (LDL-C) level with a goal to identify a low- to moderate-risk population who might benefit from a pharmacologic CVD prevention strategy. Potential participants with a risk score ≤ 15% were eligible based on decreasing LDL-C thresholds for increasing risk score >7.5% (LDL-C <190 mg/dL for risk score <7.5%, LDL-C <160 mg/dL for risk score 7.6%-10%, and LDL-C<130 mg/dL for risk score 10.1%-15%). The primary objective is to determine effects on a composite end point of MACE. Formal and independent adjudication of clinical events will occur using standardized criteria. Key secondary end points include effects on MACE components, all-cause mortality, specified non-CVD events, AIDS and non-AIDS events, and safety. RESULTS To date, REPRIEVE has enrolled >7,500 participants at approximately 120 sites across 11 countries, generating a diverse and representative population of PWH to investigate the primary objective of the trial. CONCLUSIONS REPRIEVE is the first trial investigating a primary CVD prevention strategy in PWH. REPRIEVE will inform the field of the efficacy and safety of a statin strategy among HIV-infected participants on antiretroviral therapy and provide critical information on CVD mechanisms and non-CVD events in PWH.
Collapse
Affiliation(s)
- Steven K Grinspoon
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA.
| | - Kathleen V Fitch
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Edgar Turner Overton
- Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Markella V Zanni
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Carlos Malvestutto
- Division of Infectious Diseases, Ohio State University Wexner Medical Center, Columbus, OH
| | - Michael T Lu
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Judith S Currier
- David Geffen School of Medicine at University of California Los Angeles, Division of Infectious Diseases, Los Angeles, CA
| | | | - Myron Waclawiw
- National Institutes of Health/National Heart, Lung, and Blood Institute, Bethesda, MD
| | | | | | | | | | - Udo Hoffmann
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
147
|
Hoffmann U, Lu MT, Olalere D, Adami EC, Osborne MT, Ivanov A, Aluru JS, Lee S, Arifovic N, Overton ET, Fichtenbaum CJ, Aberg JA, Alston-Smith B, Klingman KL, Waclawiw M, Burdo TH, Williams KC, Zanni MV, Desvigne-Nickens P, Cooper-Arnold K, Fitch KV, Ribaudo H, Douglas PS, Grinspoon SK. Rationale and design of the Mechanistic Substudy of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE): Effects of pitavastatin on coronary artery disease and inflammatory biomarkers. Am Heart J 2019; 212:1-12. [PMID: 30928823 PMCID: PMC6596304 DOI: 10.1016/j.ahj.2019.02.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/23/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND People with HIV (PWH) have increased cardiovascular events, inflammation, and high-risk coronary atherosclerosis. Statin therapy has been shown to lower the risk of cardiovascular disease (CVD) in the general population, but whether this results from reductions in coronary atherosclerosis and is mediated by decreased inflammation remains unknown. METHODS REPRIEVE is a randomized, placebo-controlled trial of pitavastatin calcium (4 mg/day) vs. placebo enrolling at least 7500 PWH between 40-75 years, on antiretroviral therapy (ART), with low to moderate traditional CVD risk. The Mechanistic Substudy of REPRIEVE (A5333s) is co-enrolling 800 participants from 31 US sites. These participants undergo serial contrast enhanced coronary computed tomography angiography (CCTA) and measurements of biomarkers of inflammation and immune activation at baseline and after 2 years of follow-up. The primary objectives are to determine the effects of pitavastatin on noncalcified coronary atherosclerotic plaque (NCP) volume, low attenuation plaque, and positive remodeling and on changes in immune activation and inflammation and to assess relationships between the two. Changes in CAD will be assessed in a standardized fashion by a core lab with expert readers blinded to time points and participant information; immune activation and inflammation assessment is also performed centrally. RESULTS To date the Mechanistic Substudy has completed planned enrollment, with 805 participants. CONCLUSION This study represents the first large, randomized, CCTA-based assessment of the effects of a primary prevention strategy for CVD on high-risk CAD, immune activation and inflammation among PWH. The study will assess pitavastatin's effects on coronary plaque, and the interrelationship of these changes with biomarkers of immune activation and inflammation in PWH to determine mechanisms of CVD prevention and improved outcomes in this population.
Collapse
Affiliation(s)
- Udo Hoffmann
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA.
| | - Michael T Lu
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Devvora Olalere
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Elizabeth C Adami
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Michael T Osborne
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Alex Ivanov
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - John Sukumar Aluru
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Saeyun Lee
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Nadja Arifovic
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Edgar Turner Overton
- Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | - Myron Waclawiw
- National Institutes of Health/National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Tricia H Burdo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA
| | | | - Markella V Zanni
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | | | | | - Kathleen V Fitch
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Steven K Grinspoon
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| |
Collapse
|
148
|
Hadigan C, Paules CI, Fauci AS. Association Between Human Immunodeficiency Virus Infection and Cardiovascular Diseases: Finding a Solution to Double Jeopardy. JAMA Cardiol 2019; 2:123-124. [PMID: 28002559 DOI: 10.1001/jamacardio.2016.5177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Colleen Hadigan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Catharine I Paules
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Anthony S Fauci
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
149
|
D'Souza RR, Gopalan BP, Rajnala N, Phetsouphanh C, Shet A. Increased monocyte activation with age among HIV-infected long term non-progressor children: implications for early treatment initiation. HIV Med 2019; 20:513-522. [PMID: 31131542 DOI: 10.1111/hiv.12751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The key to newer therapeutic and eradication approaches often lies in understanding slow disease progression in HIV infection. The paediatric population has been poorly studied in this regard. We aimed to describe a cohort of perinatally infected long-term nonprogressor (LTNP) children living with HIV in India and to evaluate the immune biomarkers of disease progression. METHODS LTNPs (ART-naïve, with a CD4 count ≥ 500 cells/μL at age ≥ 7 years) among the cohort of HIV-infected children were identified and monitored longitudinally, and their CD4 T-cell counts and plasma viral loads were measured every 6 months. The plasma monocyte/macrophage activation markers, namely soluble CD14 (sCD14), soluble CD163 (sCD163) and interferon-inducible protein-10 (IP-10) were measured by enzyme-linked immunosorbent assay (ELISA) in LTNPs and progressors. The Mann-Whitney U-test was used to compare the two groups and P values < 0.05 were considered statistically significant. Spearman's rank or Pearson's correlation coefficient (r) was calculated to determine the associations between variables. RESULTS Among 378 children living with HIV-1 surveyed in our cohort, 40 (10.6%) were LTNPs. Longitudinal analysis of the LTNP data showed that both CD4 count and viral load declined significantly with age (P < 0.0001 for both). Plasma sCD14 levels were significantly (P < 0.005) higher in progressors and sCD163 levels were significantly (P < 0.0001) higher in LTNPs. CONCLUSIONS The prevalence of LTNPs in our cohort of perinatally infected children living with HIV was 10.6%. We observed a trend for associations between the increasing sCD163 monocyte/macrophage activation marker levels, declining CD4 counts and the gradual loss of nonprogressor status with age in the LTNPs. These findings underscore the need for early antiretroviral therapy in those children with proven slow disease progression.
Collapse
Affiliation(s)
- R R D'Souza
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK.,Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | - B P Gopalan
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India.,The University of Trans-disciplinary Health Sciences and Technology, Bangalore, India
| | - N Rajnala
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | - C Phetsouphanh
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - A Shet
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
150
|
Sarfo FS, Nichols M, Singh A, Hardy Y, Norman B, Mensah G, Tagge R, Jenkins C, Ovbiagele B. Characteristics of hypertension among people living with HIV in Ghana: Impact of new hypertension guideline. J Clin Hypertens (Greenwich) 2019; 21:838-850. [PMID: 31125188 DOI: 10.1111/jch.13561] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/02/2019] [Accepted: 03/23/2019] [Indexed: 12/15/2022]
Abstract
Data on the burden of hypertension among people living with HIV (PLWH) in Africa are limited, especially after new expert consensus hypertension guidelines were published in 2017. The authors sought to assess the prevalence and factors associated with hypertension among PLWH. This is a cross-sectional study involving PLWH on combination antiretroviral therapy (cART) (n = 250) compared with sex-matched cART-naïve PLWH (n = 201) in Ghana. Hypertension was defined as blood pressure ≥ 140/90 mm Hg or use of antihypertensive drugs. The authors also assessed the prevalence and predictors associated with hypertension using the recent guideline recommended cutoff BP ≥ 130/80 mm Hg. Multivariate logistic regression models were fitted to identify factors associated with hypertension among PLWH. The mean age of PLWH on cART was 45.7 ± 8.6 years, and 42.9 ± 8.8 years among PLWH cART-naive with 81% of study participants being women. The prevalence of hypertension among PLWH on cART and PLWH cART-naïve was 36.9% and 23.4%, P = 0.002 at BP ≥ 140/90 mm Hg and 57.2% and 42.3%, respectively, P = 0.0009, at BP ≥ 130/80 mm Hg. Factors associated with hypertension at BP ≥ 140/90 mm Hg in the PLWH group with adjusted odds ratio (95% CI) were increasing age, 2.08 (1.60-2.71) per 10 years, and body mass index, 1.53 (1.24-1.88) per 5 kg/m2 rise. At BP ≥ 130/80 mm Hg, cART exposure, aOR of 1.77 (95% CI: 1.20-2.63), family history of hypertension, aOR of 1.43 (1.12-1.83), and hypertriglyceridemia, aOR of 0.54 (0.31-0.93), were associated with hypertension. Among PLWH, cART exposure was associated with higher prevalence of hypertension per the new guideline definition, a finding which warrants further investigation and possible mitigation.
Collapse
Affiliation(s)
- Fred Stephen Sarfo
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Michelle Nichols
- Medical University of South Carolina, Charleston, South Carolina
| | - Arti Singh
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Betty Norman
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | - Ralle Tagge
- Medical University of South Carolina, Charleston, South Carolina
| | - Carolyn Jenkins
- Medical University of South Carolina, Charleston, South Carolina
| | | |
Collapse
|