1
|
Watanabe M, Davidson L, Smith P, Castellucio PF, Jergovic M, Uhrlaub JL, Smithey MJ, Fantry LE, Dechambre B, Wilson RC, Knox KC, Ren J, Stowe RP, Weinstock G, Twigg H, Nikolich JŽ. Anti-cytomegalovirus antibody levels stratify human immune profiles across the lifespan. GeroScience 2024; 46:4225-4242. [PMID: 38512581 PMCID: PMC11336022 DOI: 10.1007/s11357-024-01124-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Human cytomegalovirus (hCMV) is a ubiquitous latent persistent herpesvirus infecting 60-90% of the population worldwide. hCMV carriage in immunocompetent people is asymptomatic; thus, hCMV can be considered a component of normative aging. However, hCMV powerfully modulates many features of the immune, and likely other, systems and organs. Questions remain as to how hCMV carriage affects the human host. We used anti-CMV antibody titers as a stratifying criterion to examine the impact of "intensity" of hCMV infection as a potential biomarker of aging, inflammation, and immune homeostasis in a cohort of 247 participants stratified into younger (21-40 years) and older (> 65 years of age) groups. We showed that anti-CMV antibody titers increased with age and directly correlated to increased levels of soluble tumor necrosis factor (sTNFR) I in younger but not older participants. CD8 + cell numbers were reduced in the older group due to the loss in CD8 + T naïve (Tn) cells. In CMV carriers and, in particular, in anti-CMV Ab-high participants, this loss was mitigated or reversed by an increase in the numbers of CD8 + T effector memory (Tem) and T effector memory reexpressing CD45RA (Temra) cells. Analysis of CD38, HLA-DR, and CD57 expression revealed subset (CD4 or CD8)-specific changes that correlated with anti-CMV Ab levels. In addition, anti-CMV Ab levels predicted anti-CMV CD8 T cell responsiveness to different CMV open reading frames (ORFs) selectively in older participants, which correlated to the transcriptional order of expression of specific CMV ORFs. Implications of these results for the potential predictive value of anti-CMV Ab titers during aging are discussed.
Collapse
Affiliation(s)
- Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Lisa Davidson
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Patricia Smith
- Division of Pulmonary Medicine, Department of Medicine, Indiana University College of Medicine, Bloomington, IN, USA
| | - Peter F Castellucio
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mladen Jergovic
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Megan J Smithey
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA
| | - Lori E Fantry
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Brett Dechambre
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Department of Health Services, Phoenix, AZ, USA
| | - Rachel C Wilson
- Division of Pulmonary Medicine, Department of Medicine, Indiana University College of Medicine, Bloomington, IN, USA
| | - Kenneth C Knox
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jie Ren
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Homer Twigg
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Arizona Center On Aging, University of Arizona College of Medicine-Tucson, 1501 N. Campbell Ave, P.O. Box 245221, Tucson, AZ, 85724, USA.
| |
Collapse
|
2
|
Abidi MZ, Umbleja T, Overton ET, Burdo T, Flynn JM, Lu MT, Taron J, Schnittman SR, Fitch KV, Zanni MV, Fichtenbaum CJ, Malvestutto C, Aberg JA, Fulda ES, Eckard AR, Manne-Goehler J, Tuan JJ, Ribaudo HJ, Douglas PS, Grinspoon SK, Brown TT, Erlandson KM. Cytomegalovirus IgG is Associated With Physical Function But Not Muscle Density in People With HIV. J Acquir Immune Defic Syndr 2024; 95:470-478. [PMID: 38180893 PMCID: PMC10947880 DOI: 10.1097/qai.0000000000003377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Cytomegalovirus (CMV) seropositivity is associated with poor outcomes, including physical function impairment, in people without HIV. We examined associations between CMV IgG titer and physical function in virologically suppressed people with HIV (PWH). METHODS REPRIEVE is a double-blind randomized trial evaluating pitavastatin for primary prevention of atherosclerotic cardiovascular disease in PWH. This analysis focused on participants enrolled in a substudy with additional biomarker testing, imaging [coronary CT angiography], and physical function measures at entry. CMV IgG was measured using quantitative enzyme immunoassay, physical function by Short Physical Performance Battery, and muscle density and area by CT. Associations between CMV IgG (risk factor) and outcomes were evaluated using the partial Spearman correlation and linear and log-binomial regression. RESULTS Among 717 participants, 82% male, the median CMV IgG was 2716 (Q1, Q3: 807, 6672) IU/mL, all above the limit of quantification. Among 631 participants with imaging, there was no association between CMV IgG and CT-based muscle density or area, controlling for age (r = -0.03 and r = -0.01, respectively; P ≥ 0.38). Among 161 participants with physical function data, higher CMV IgG was associated with poorer overall modified Short Physical Performance Battery score ( P = 0.02), adjusted for age, nadir CD4, and high-sensitivity C-reactive protein. CONCLUSIONS Higher CMV IgG titer was associated with poorer physical function, not explained by previous immune compromise, inflammation, or muscle density or area. Further mechanistic studies are needed to understand this association and whether CMV-specific therapy can affect physical function in PWH.
Collapse
Affiliation(s)
- Maheen Z. Abidi
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Triin Umbleja
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edgar T. Overton
- Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Tricia Burdo
- Department of Microbiology, Immunology, and Inflammation and Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jacqueline M. Flynn
- Department of Microbiology, Immunology, and Inflammation and Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jana Taron
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Medical Center – University of Freiburg, Germany
| | - Samuel R. Schnittman
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Carlos Malvestutto
- Division of Infectious Diseases, Ohio State University Medical Center, Columbus, OH, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evelynne S. Fulda
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Allison Ross Eckard
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, Charleston, SC, USA
| | - Jennifer Manne-Goehler
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica J. Tuan
- Division of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Heather J. Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, U.S.A
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Todd T. Brown
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine M. Erlandson
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Denver, CO, USA
| |
Collapse
|
3
|
Abidi MZ, Schold JD, Kaplan B, Weinberg A, Erlandson KM, Malamon JS. Patient years lost due to cytomegalovirus serostatus mismatching in the scientific registry of transplant recipients. Front Immunol 2024; 14:1292648. [PMID: 38264645 PMCID: PMC10803440 DOI: 10.3389/fimmu.2023.1292648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Background The cytomegalovirus (CMV) mismatch rate in deceased donor kidney transplant (DDKT) recipients in the US remains above 40%. Since CMV mismatching is common in DDKT recipients, the cumulative effects may be significant in the context of overall patient and graft survival. Our primary objective was to describe the short- and long-term risks associated with high-risk CMV donor positive/recipient negative (D+/R-) mismatching among DDKT recipients with the explicit goal of deriving a mathematical mismatching penalty. Methods We conducted a retrospective, secondary analysis of the Scientific Registry of Transplant Recipients (SRTR) database using donor-matched DDKT recipient pairs (N=105,608) transplanted between 2011-2022. All-cause mortality and graft failure hazard ratios were calculated from one year to ten years post-DDKT. All-cause graft failure included death events. Survival curves were calculated using the Kaplan-Meier estimation at 10 years post-DDKT and extrapolated to 20 years to provide the average graft days lost (aGDL) and average patient days lost (aPDL) due to CMV D+/R- serostatus mismatching. We also performed an age-based stratification analysis to compare the relative risk of CMV D+ mismatching by age. Results Among 31,518 CMV D+/R- recipients, at 1 year post-DDKT, the relative risk of death increased by 29% (p<0.001), and graft failure increased by 17% (p<0.001) as compared to matched CMV D+/R+ group (N=31,518). Age stratification demonstrated a significant increase in the risk associated with CMV mismatching in patients 40 years of age and greater. The aGDL per patient due to mismatching was 125 days and the aPDL per patient was 100 days. Conclusion The risks of CMV D+/R- mismatching are seen both at 1 year post-DDKT period and accumulated throughout the lifespan of the patient, with the average CMV D+/R- recipient losing more than three months of post-DDKT survival time. CMV D+/R- mismatching poses a more significant risk and a greater health burden than previously reported, thus obviating the need for better preventive strategies including CMV serodirected organ allocation to prolong lifespans and graft survival in high-risk patients.
Collapse
Affiliation(s)
- Maheen Z. Abidi
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
| | - Jesse D. Schold
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Bruce Kaplan
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Adriana Weinberg
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
- Department of Pediatrics and Pathology, University of Colorado, Aurora, CO, United States
| | - Kristine M. Erlandson
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
| | - John S. Malamon
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| |
Collapse
|
4
|
Satta S, Rockwood SJ, Wang K, Wang S, Mozneb M, Arzt M, Hsiai TK, Sharma A. Microfluidic Organ-Chips and Stem Cell Models in the Fight Against COVID-19. Circ Res 2023; 132:1405-1424. [PMID: 37167356 PMCID: PMC10171291 DOI: 10.1161/circresaha.122.321877] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
SARS-CoV-2, the virus underlying COVID-19, has now been recognized to cause multiorgan disease with a systemic effect on the host. To effectively combat SARS-CoV-2 and the subsequent development of COVID-19, it is critical to detect, monitor, and model viral pathogenesis. In this review, we discuss recent advancements in microfluidics, organ-on-a-chip, and human stem cell-derived models to study SARS-CoV-2 infection in the physiological organ microenvironment, together with their limitations. Microfluidic-based detection methods have greatly enhanced the rapidity, accessibility, and sensitivity of viral detection from patient samples. Engineered organ-on-a-chip models that recapitulate in vivo physiology have been developed for many organ systems to study viral pathology. Human stem cell-derived models have been utilized not only to model viral tropism and pathogenesis in a physiologically relevant context but also to screen for effective therapeutic compounds. The combination of all these platforms, along with future advancements, may aid to identify potential targets and develop novel strategies to counteract COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Sandro Satta
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Sarah J. Rockwood
- Stanford University Medical Scientist Training Program, Palo Alto, CA (S.J.R.)
| | - Kaidong Wang
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Shaolei Wang
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tzung K. Hsiai
- Division of Cardiology and Department of Bioengineering, School of Engineering (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Division of Cardiology, Department of Medicine, School of Medicine (S.S., K.W., S.W., T.K.H.), University of California, Los Angeles
- Department of Medicine, Greater Los Angeles VA Healthcare System, California (S.S., K.W., S.W., T.K.H.)
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Smidt Heart Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
- Cancer Institute (M.M., M.A., A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
5
|
Increased Risk to Develop Hypertension and Carotid Plaques in Patients with Long-Lasting Helicobacter pylori Gastritis. J Clin Med 2022; 11:jcm11092282. [PMID: 35566408 PMCID: PMC9104887 DOI: 10.3390/jcm11092282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori infection has been reported to be positively associated with hypertension, although with conflicting results. In this study, the relationship between H. pylori infection and hypertension, as well as atherosclerotic carotid lesions, was analyzed. Methods. Clinical records of patients referred to undergo upper endoscopy and gastric biopsy were retrieved. Information regarding the presence of H. pylori infection with atrophy/metaplasia/dysplasia (interpreted as a long-lasting infection), and current or past H. pylori infection was collected, as well as demographic variables, smoking habits, body mass index (BMI), dyslipidemia, diabetes, hypertension, presence of carotid lesions, and current treatment, and analyzed by multivariable regression models. Results. A total of 7152 clinical records from patients older than 30 years (63.4% women) were available for the study. Hypertension was present in 2039 (28.5%) patients and the risk was significantly increased in those with long-lasting H. pylori infection after adjusting for age decades, sex, BMI, cigarette smoking, diabetes, and dyslipidemia (OR 1.17, 95% CI 1.02–1.35). In addition, the long-lasting H. pylori infection was an independent risk for carotid plaques (OR 2.15, 95% CI 1.14–4.09). Conclusions. Our retrospective study demonstrated that long-lasting H. pylori infection is an independent risk factor for hypertension and the presence of carotid lesions after adjusting for potential confounders, although further validation our findings is needed from prospective studies.
Collapse
|
6
|
Chen TH, Ou SM, Tarng DC. Associations of high anti-CMV IgG titer with renal function decline and allograft rejection in kidney transplant patients. J Chin Med Assoc 2022; 85:183-189. [PMID: 34882099 DOI: 10.1097/jcma.0000000000000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND An anti-cytomegalovirus (CMV) immunoglobulin G (IgG) antibody is produced after primary CMV infection and generally persists after the primary infection. However, it is not well-known about the relationship between anti-CMV IgG titer and outcomes in kidney transplant recipients. We, therefore, aimed to explore the role of anti-CMV IgG titer on the risks of CMV disease development, allograft rejection, renal function decline, and mortality. METHODS In a hospital-based study, we identified 179 CMV-seropositive kidney transplant recipients between January 2013 and December 2017. These patients were divided into low and high anti-CMV IgG titer groups, respectively. The cutoff level of anti-CMV IgG titer was determined by receiver operating characteristic curve analysis. The outcomes evaluated included CMV disease, decrease of ≥15% in estimated glomerular filtration rate (eGFR), biopsy-proven allograft rejection, and all-cause mortality. RESULTS The high anti-CMV IgG titer group (≥846.2 AU/mL) exhibited a higher risk of CMV disease (adjusted hazard ratio [aHR], 3.77; 95% CI, 1.47-9.68; p = 0.006), eGFR decline ≥15% (aHR, 2.00; 95% CI, 1.19-3.35; p = 0.009), and renal allograft rejection (aHR, 2.95; 95% CI, 1.11-7.87; p = 0.030) than the low titer group (<846.2 AU/mL). CONCLUSION In kidney transplant recipients, a high anti-CMV IgG titer was associated with higher risks for developing CMV disease, undergoing allograft rejection, and eGFR decline.
Collapse
Affiliation(s)
- Tz-Heng Chen
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital Yuli Branch, Hualien, Taiwan, ROC
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shuo-Ming Ou
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department and Institute of Physiology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
7
|
A Comprehensive Review of Infections in Older Kidney Transplant Recipients. CURRENT TRANSPLANTATION REPORTS 2021. [DOI: 10.1007/s40472-021-00320-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
8
|
Cooke JP, Connor JH, Jain A. Acute and Chronic Cardiovascular Manifestations of COVID-19: Role for Endotheliopathy. Methodist Debakey Cardiovasc J 2021; 17:53-62. [PMID: 34992723 PMCID: PMC8680072 DOI: 10.14797/mdcvj.1044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
SARS-CoV-2, the virus that causes coronavirus disease 19 (COVID-19), is associated with a bewildering array of cardiovascular manifestations, including myocardial infarction and stroke, myocarditis and heart failure, atrial and ventricular arrhythmias, venous thromboembolism, and microvascular disease. Accumulating evidence indicates that a profound disturbance of endothelial homeostasis contributes to these conditions. Furthermore, the pulmonary infiltration and edema, and later pulmonary fibrosis, in patients with COVID-19 is promoted by endothelial alterations including the expression of endothelial adhesion molecules and chemokines, increased intercellular permeability, and endothelial-to-mesenchyme transitions. The cognitive disturbance occurring in this disease may also be due in part to an impairment of the blood-brain barrier. Venous thrombosis and pulmonary thromboembolism are most likely associated with an endothelial defect caused by circulating inflammatory cytokines and/or direct endothelial invasion by the virus. Endothelial-targeted therapies such as statins, nitric oxide donors, and antioxidants may be useful therapeutic adjuncts in COVID-19 by restoring endothelial homeostasis.
Collapse
Affiliation(s)
- John P Cooke
- Houston Methodist Research Institute, Houston Methodist, Houston, TX, US
| | - John H Connor
- Boston University Medical Center and National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, US
| | - Abhishek Jain
- Texas A&M University, College Station, TX, US.,Texas A&M Health Science Center, Bryan, TX, US
| |
Collapse
|
9
|
La Y, Kwon DE, Yoo SG, Lee KH, Han SH, Song YG. Human cytomegalovirus seroprevalence and titres in solid organ transplant recipients and transplant donors in Seoul, South Korea. BMC Infect Dis 2019; 19:948. [PMID: 31703564 PMCID: PMC6842252 DOI: 10.1186/s12879-019-4607-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) can cause poor outcomes in solid organ transplant (SOT) recipients; moreover, it is associated with cardiovascular diseases (CVD) in the general population. Accordingly, anti-HCMV immunoglobulin G (IgG) seroepidemiology may be useful in identifying the risk of post-SOT HCMV infection or disease as well as immunosenescence or CVD. However, HCMV seroprevalence and titre have not been fully evaluated with regard to age distribution or compared between SOT recipients and healthy individuals in South Korea. Methods We retrospectively retrieved all unduplicated anti-HCMV IgG results of individuals aged > 1 year evaluated between July 2006 and November 2017 at Severance Hospital in Seoul. The cohort, excluding haematopoietic stem cell transplant recipients and subjects with equivocal values, included 2184 SOT recipients and 3015 healthy transplant donors. All IgG results in the SOT recipients were measured during the pre-transplant period. Results The overall IgG seroprevalence and titres were significantly higher among SOT recipients than among healthy donors (98.7% vs. 88.6%, p < 0.001, and 64.7 ± 44.3 vs. 49.8 ± 20.6 arbitrary units/mL, p < 0.001, respectively). The lowest seropositive rate in the SOT group was observed in recipients aged between 11 and 15 years (70.6%). The frequency of seropositivity among adults aged ≥41 years increased to ≥90% in SOT recipients and healthy donors. Age was independently associated with higher HCMV seroprevalence (41–60 years, OR, 76.4, 95% CI, 24.5–238.9, p < 0.001; ≥ 61 years, OR, 4.4, 95% CI, 1.3–14.9, p < 0.001, compared to ≤40 years). The healthy donor group had an independently low HCMV seropositive rate (OR, 0.1, 95% CI, 0.1–0.2, p < 0.001). Conclusions HCMV seropositivity was the lowest among school-aged children and adolescents. IgG testing revealed an intermediate serostatus risk of post-transplant HCMV infection and disease for most adult SOT recipients in South Korea.
Collapse
Affiliation(s)
- Yeonju La
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Eun Kwon
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seul Gi Yoo
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Hwa Lee
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Han
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Yong Goo Song
- Department of Internal Medicine, Division of Infectious Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Abstract
Influenza viruses infect the upper respiratory system, causing usually a self-limited disease with mild respiratory symptoms. Acute lung injury, pulmonary microvascular leakage and cardiovascular collapse may occur in severe cases, usually in the elderly or in immunocompromised patients. Acute lung injury is a syndrome associated with pulmonary oedema, hypoxaemia and respiratory failure. Influenza virus primarily binds to the epithelium, interfering with the epithelial sodium channel function. However, the main clinical devastating effects are caused by endothelial dysfunction, thought to be the main mechanism leading to pulmonary oedema, respiratory failure and cardiovascular collapse. A significant association was found between influenza infection and acute myocardial infarction (AMI). The incidence of admission due to AMI during an acute viral infection was six times as high during the 7 days after laboratory confirmation of influenza infection as during the control interval (10-fold in influenza B, 5-fold in influenza A, 3.5-fold in respiratory syncytial virus and 2.7-fold for all other viruses). Our review will focus on the mechanisms responsible for endothelial dysfunction during influenza infection leading to cardiovascular collapse and death.
Collapse
Affiliation(s)
- A Peretz
- Clinical Microbiology Laboratory, Baruch Padeh Medical Center, Poriya, Tiberias, Israel
- The Research Institute, Baruch Padeh Medical Center
- Azrieli Faculty of Medicine
| | - M Azrad
- Clinical Microbiology Laboratory, Baruch Padeh Medical Center, Poriya, Tiberias, Israel
- The Research Institute, Baruch Padeh Medical Center
- Azrieli Faculty of Medicine
| | - A Blum
- The Research Institute, Baruch Padeh Medical Center
- Azrieli Faculty of Medicine
- Vascular and Regenerative Research Laboratory, Bar-Ilan University, Galilee, Safed, Israel
| |
Collapse
|
11
|
Feng Q, Hui J, Tang N, Liu YM, Zhong H, Li Z, Wang LM, Qu YY, Deng FM, He F. Unexpected role of the human cytomegalovirus contribute to essential hypertension in the Kazakh Chinese population of Xinjiang. Biosci Rep 2018; 38:BSR20171522. [PMID: 29752343 PMCID: PMC6019381 DOI: 10.1042/bsr20171522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/29/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection, chronic inflammation and oxidative stress, the renin-angiotensin system (RAS), endothelial function, and DNA methylation play roles in the pathogenesis of essential hypertension (EH); however, the mechanism by which HCMV predisposes patients to hypertension remain unclear. Our group previously demonstrated an association between EH and HCMV infection in Kazakh Chinese. Here, we investigated the relationship between HCMV infection and other clinicopathological features in 720 Kazakh individuals with or without hypertension (n=360 each; age: 18-80). Multiple linear and logistic regression analyses were used to determine the associations between HCMV infection, clinical characteristics, and EH. Notably, patients with EH, particularly those with HCMV infection, exhibited a marked increase in tumor necrosis factor-α (TNF-α) and 8-hydroxy-2-deoxyguanosine (8-OHDG) levels, but a decrease in endothelial nitric oxide synthase (eNOS) and renin levels. Similarly, elevated TNF-α and 8-OHDG levels were independent predictors of increased HCMV antibody titers, whereas eNOS and renin were negatively correlated with the latter. Moreover, serum angiotensin-converting enzyme (sACE, ACE) methylation was increased, whereas 11-β hydroxysteroid dehydrogenase 2 (HSD11β2; HSD3B2) methylation was decreased in patients with EH who were also infected with HCMV. A positive correlation between HSD3B2 methylation and HCMV IgG titer and blood pressure was additionally observed, whereas angiotensin-converting enzyme (ACE) methylation was inversely correlated with blood pressure. Collectively, these data indicate that HCMV may contribute to EH development in the Kazakh Chinese by increasing TNF-α and 8-OHDG levels, suppressing eNOS and renin, and manipulating HSD3B2 and ACE methylation.
Collapse
Affiliation(s)
- Qian Feng
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Jing Hui
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Na Tang
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Yong-Min Liu
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Hua Zhong
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Zhen Li
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - La-Mei Wang
- Centre of Medical Functional Experiments, Medical College of Shihezi University, Shihezi, China
| | - Yuan-Yuan Qu
- Department of Respiration Medicine, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xijiang, China
| | - Feng-Mei Deng
- Department of Pathophysiology, Chengdu Medical College, Sichuan, China
| | - Fang He
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| |
Collapse
|
12
|
Wang H, Peng G, Bai J, He B, Huang K, Hu X, Liu D. Cytomegalovirus Infection and Relative Risk of Cardiovascular Disease (Ischemic Heart Disease, Stroke, and Cardiovascular Death): A Meta-Analysis of Prospective Studies Up to 2016. J Am Heart Assoc 2017; 6:JAHA.116.005025. [PMID: 28684641 PMCID: PMC5586265 DOI: 10.1161/jaha.116.005025] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Several studies have suggested that cytomegalovirus infection is likely associated with an increased relative risk of cardiovascular disease (CVD); however, the results are inconsistent. We aimed to provide a systematic review and meta-analysis of community-based prospective studies assessing the association between cytomegalovirus infection and relative risk of CVD. METHODS AND RESULTS We searched Medline and EMBASE to retrieve prospective studies that reported risk estimates of the association between cytomegalovirus infection and relative risk of CVD. The search yielded 10 articles including a total of 34 564 participants and 4789 CVD patients. Overall, exposure to cytomegalovirus infection was associated with a 22% (relative risk: 1.22, 95% CI: 1.07-1.38, P=0.002) increased relative risk of future CVD. We estimated that 13.4% of CVD incidence could be attributable to cytomegalovirus infection. CONCLUSIONS In conclusion, cytomegalovirus infection is associated with a significantly increased relative risk of CVD.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Cardiology Ward 2, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Geng Peng
- Department of Cardiology Ward 2, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Jing Bai
- Department of Endocrinology and Diabetes, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Bing He
- Department of Joint Surgery, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Kecheng Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinrong Hu
- Department of Cardiology Ward 2, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Dongliang Liu
- Department of Cardiology Ward 2, Luohe Central Hospital, The First Affiliated Hospital of Luohe Medical College, Luohe, China
| |
Collapse
|
13
|
Human cytomegalovirus infection and vascular disease risk: A meta-analysis. Virus Res 2016; 227:124-134. [PMID: 27664838 DOI: 10.1016/j.virusres.2016.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Human cytomegalovirus (HCMV) infection has been associated with the acceleration of vascular disease. Numbers studies were conducted to analyze the association between HCMV infection and risk of vascular disease, but no clear consensus had been reached. The aim of this study was to confirm this relationship precisely by doing a systematic review and meta-analysis. METHODS We identified relevant studies through a search of PubMed and Embase. Studies were eligible for inclusion if they fulfilled all of the following selection criteria: (1) evaluating the association between HCMV infection and vascular disease; (2) case-control studies or nested case-control studies; (3) and supply the numbers (or percentage) of positivity for HCMV infection in cases and controls, respectively. Data were extracted and analyzed independently by two investigators. Ultimately, We included data from 68 studies, which altogether enrolled 12027 cases and 15386 controls from 24 countries. RESULTS HCMV IgG was detected 7376 in 10611 cases, HCMV IgM was detected 153 in 1486 cases and HCMV DNA was detected 654 in 2139 cases. Overall, people exposed to HCMV infection had higher risk than those not exposed for vascular disease (OR 1.70 [95% CI 1.43-2.03] IgG-based HCMV tests, 2.88 [95% CI 1.87-4.43] IgM-based HCMV tests and 2.56 [95% CI 1.46-4.49 PCR-based HCMV tests]). HCMV infection was clearly identified as a risk factor for vascular disease in Asian group, Caucasian group and other group, especially Asian group(OR 1.86 [95% CI 1.33-2.60] IgG-based HCMV tests, 3.57 [95% CI 1.94-6.60] IgM-based HCMV tests and 4.09 [95% CI 3.10-5.40 PCR-based HCMV tests]). CONCLUSION This meta-analysis suggested that HCMV infection is associated with an increased risk for vascular disease.
Collapse
|
14
|
Wang GC, Han C, Detrick B, Casolaro V, Levine DM, Fried LP, Walston JD. Herpesvirus Infections and Risk of Frailty and Mortality in Older Women: Women's Health and Aging Studies. J Am Geriatr Soc 2016; 64:998-1005. [PMID: 27131018 DOI: 10.1111/jgs.14090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To examine the relationship between herpesvirus infections and mortality and incident frailty risks in community-dwelling older women. DESIGN Nested prospective cohort study. SETTING Women's Health and Aging Studies I and II. PARTICIPANTS Community-dwelling older women aged 70 to 79 (n = 633). MEASUREMENTS Baseline serum antibody (immunoglobulin G) levels against four herpesviruses (herpes simplex virus types 1 (HSV-1) and 2 (HSV-2), varicella-zoster virus (VZV), 7 Epstein-Barr virus (EBV)), 3-year incident frailty rates, and 5-year mortality. RESULTS Women seropositive for HSV-1 and HSV-2, but not VZV and EBV, had higher risk of 3-year incident frailty (HSV-1: hazard ratio (HR) = 1.90, 95% confidence interval (CI) = 0.96-3.74; HSV-2: HR = 2.10, 95% CI = 1.05-4.37) and 5-year mortality (HR = 1.73, 95% CI = 0.93-3.20; HR = 1.80, 95% CI = 0.94-3.44, respectively) than seronegative women. Incremental increases in serum HSV-1 and HSV-2 antibody levels were associated with incrementally higher risks of incident frailty and mortality. After adjustment for potential confounders, only higher serum HSV-2 antibody level was independently predictive of higher risk of mortality in older women (for each unit increase in antibody index, HR = 1.47, 95% CI = 1.05-2.07). CONCLUSION HSV-1 and HSV-2 antibody levels are not independently associated with risk of incident frailty in older women. Only HSV-2 antibody level is independently predictive of 5-year mortality risk, with each incremental increase in the antibody level adding further risk.
Collapse
Affiliation(s)
- George C Wang
- Division of Geriatric Medicine and Gerontology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Christina Han
- Division of Geriatric Medicine and Gerontology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Barbara Detrick
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Vincenzo Casolaro
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy
| | - David M Levine
- Division of General Internal Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Linda P Fried
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
15
|
Liu S, Liu N, Ruan Y, Li X, Wen D, Chen L, Guo X, Guo L, Jiang C, Long D, Yu R, Tang R, Sang C, Bai Y, Zhang T, Bai R, Du X, Dong J, Ma X, Ma C. Plasma IgG antibody against cytomegalovirus but not herpes simplex virus is associated with recurrence of atrial fibrillation after catheter ablation. Eur Heart J Suppl 2016. [DOI: 10.1093/eurheartj/suw007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
16
|
Zhang J, Liu YY, Sun HL, Li S, Xiong HR, Yang ZQ, Xiang GD, Jiang XJ. High Human Cytomegalovirus IgG Level is Associated with Increased Incidence of Diabetic Atherosclerosis in Type 2 Diabetes Mellitus Patients. Med Sci Monit 2015; 21:4102-10. [PMID: 26717490 PMCID: PMC4699628 DOI: 10.12659/msm.896071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background At present, whether human cytomegalovirus (HCMV) infection is associated with type 2 diabetes mellitus (T2DM) is debatable. The effect of active HCMV infection on glucose regulation has been poorly studied. Although HCMV infection is correlated with atherosclerosis in cardiovascular disease, the role of HCMV infection in the development of diabetic atherosclerosis in T2DM is unclear and is usually neglected by endocrinologists. The aim of this study was to assess the effects of HCMV infection on glucose regulation and the development of diabetic atherosclerosis in T2DM patients. Material/Methods A total of 222 hospitalized T2DM patients were enrolled. Nested polymerase chain reactions were used to detect HCMV DNA extracted from peripheral blood leukocytes. Quantitative real-time PCR was used to determine viral load. HCMV IgG antibody concentrations were analyzed by chemiluminescence immunoassay. Results HCMV active infection, viral load, and HCMV IgG titers were not correlated with glucose regulation. Binary logistic regression demonstrated that the highest quartile of HCMV IgG concentration (>500 U/ml) was correlated with the incidence of diabetic atherosclerosis (OR: 8.0, 95%CI: 2.3–27.2), and that titer >127U/ml of HCMV IgG is an independent predictor for the development of diabetic atherosclerosis in T2DM patients (OR: 4.6, 95%CI: 1.9–11.3) after adjustment for all potential confounding factors. Conclusions Active HCMV infection is unlikely to influence glucose regulation in T2DM. However, HCMV IgG titers are associated with the incidence of diabetic atherosclerosis, and titer >127U/ml of HCMV IgG might be an independent risk factor for the development of diabetic atherosclerosis in T2DM patients.
Collapse
Affiliation(s)
- Jun Zhang
- Graduate School, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Yuan-yuan Liu
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Hui-ling Sun
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei, China (mainland)
| | - Shan Li
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Hai-rong Xiong
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Zhan-qiu Yang
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Guang-da Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei, China (mainland)
| | - Xiao-jing Jiang
- Department of Infectious Diseases, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei, China (mainland)
| |
Collapse
|
17
|
Barnes LL, Capuano AW, Aiello AE, Turner AD, Yolken RH, Torrey EF, Bennett DA. Cytomegalovirus infection and risk of Alzheimer disease in older black and white individuals. J Infect Dis 2015; 211:230-7. [PMID: 25108028 PMCID: PMC4326304 DOI: 10.1093/infdis/jiu437] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Human cytomegalovirus (CMV) is prevalent in older adults and has been implicated in many chronic diseases of aging. This study investigated the relation between CMV and the risk of Alzheimer disease (AD). METHODS Data come from 3 cohort studies that included 849 participants (mean age [±SD], 78.6 ± 7.2 years; mean education duration [±SD], 15.4 ± 3.3 years; 25% black). RESULTS A solid-phase enzyme-linked immunosorbent assay was used for detecting type-specific immunoglobulin G antibody responses to CMV and herpes simplex virus type 1 (HSV-1) measured in archived serum samples. Of 849 participants, 73.4% had serologic evidence of exposure to CMV (89.0% black and 68.2% white; P < .001). During an average of 5.0 years of follow-up, 93 persons developed AD. CMV seropositivity was associated with an increased risk of AD (relative risk, 2.15; 95% confidence interval, 1.42-3.27) and a faster rate of decline in global cognition (estimate [±standard error], -0.02 ± 0.01; P = .03) in models that controlled for age, sex, education duration, race, vascular risk factors, vascular diseases, and apolipoprotein ε4 level. Results were similar in black and white individuals for both incident AD and change in cognitive function and were independent of HSV-1 status. CONCLUSIONS These results suggest that CMV infection is associated with an increased risk of AD and a faster rate of cognitive decline in older diverse populations.
Collapse
Affiliation(s)
- Lisa L. Barnes
- Rush Alzheimer's Disease Center,Department of Neurological Sciences,Department of Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Ana W. Capuano
- Rush Alzheimer's Disease Center,Department of Neurological Sciences
| | | | | | | | | | - David A. Bennett
- Rush Alzheimer's Disease Center,Department of Neurological Sciences
| |
Collapse
|
18
|
Khoretonenko MV, Brunson JL, Senchenkov E, Leskov IL, Marks CR, Stokes KY. Platelets, acting in part via P-selectin, mediate cytomegalovirus-induced microvascular dysfunction. Am J Physiol Heart Circ Physiol 2014; 307:H1745-53. [PMID: 25326535 DOI: 10.1152/ajpheart.00201.2014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytomegalovirus (CMV) infects a majority of the population worldwide. It has been implicated in cardiovascular disease, induces microvascular dysfunction, and synergizes with hypercholesterolemia to promote leukocyte and platelet recruitment in venules. Although platelets and platelet-associated P-selectin contribute to cardiovascular disease inflammation, their role in CMV-induced vascular responses is unknown. We assessed the role of platelets in CMV-induced microvascular dysfunction by depleting platelets and developing bone marrow chimeric mice deficient in platelet P-selectin. Wild-type and chimeric mice received mock or murine (m)CMV intraperitoneally. Five weeks later, some mice were switched to a high-cholesterol diet (HC) to investigate the synergism between mCMV and HC. Arteriolar vasodilation and recruitment of leukocytes and donor platelets in venules were measured at 11wk. mCMV with or without HC caused significant endothelial dysfunction in arterioles. Platelet depletion restored normal vasodilation in mCMV-HC but not mCMV-ND mice, whereas protection was seen in both groups for platelet P-selectin chimeras. Only mCMV + HC elevated leukocyte and platelet recruitment in venules. Leukocyte adhesion was reduced to mock levels by acute platelet depletion but was only partially decreased in platelet P-selectin chimeras. Platelets from mCMV-HC mice and, to a lesser extent, mCMV-ND but not mock-HC mice showed significant adhesion in mCMV-HC recipients. Our findings implicate a role for platelets, acting through P-selectin, in CMV-induced arteriolar dysfunction and suggest that the addition of HC leads to a platelet-dependent, inflammatory infiltrate that is only partly platelet P-selectin dependent. CMV appeared to have a stronger activating influence than HC on platelets and may represent an additional therapeutic target in vulnerable patients.
Collapse
Affiliation(s)
- Mikhail V Khoretonenko
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Jerry L Brunson
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Evgeny Senchenkov
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Igor L Leskov
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Christian R Marks
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Karen Y Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| |
Collapse
|
19
|
Abstract
Although human cytomegalovirus (HCMV) primary infection is generally asymptomatic, in immune-compromised patients HCMV increases morbidity and mortality. As a member of the betaherpesvirus family, in vivo studies of HCMV are limited due to its species specificity. CMVs from other species are often used as surrogates to express HCMV genes/proteins or used as models for inferring HCMV protein function in humans. Using innovative experiments, these animal models have answered important questions about CMV's life cycle, dissemination, pathogenesis, immune evasion, and host immune response. This chapter provides CMV biologists with an overview of the insights gained using these animal models. Subsequent chapters will provide details of the specifics of the experimental methods developed for each of the animal models discussed here.
Collapse
Affiliation(s)
- Pranay Dogra
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | |
Collapse
|
20
|
Yaiw KC, Ovchinnikova O, Taher C, Mohammad AA, Davoudi B, Shlyakhto E, Rotar O, Konradi A, Wilhelmi V, Rahbar A, Butler L, Assinger A, Söderberg-Nauclér C. High prevalence of human cytomegalovirus in carotid atherosclerotic plaques obtained from Russian patients undergoing carotid endarterectomy. HERPESVIRIDAE 2013; 4:3. [PMID: 24229441 PMCID: PMC4177206 DOI: 10.1186/2042-4280-4-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 11/04/2013] [Indexed: 11/10/2022]
Abstract
Background Human cytomegalovirus (HCMV) infection is associated with cardiovascular disease (CVD) but the role of this virus in CVD progression remains unclear. We aimed to examine the HCMV serostatus in Russian patients (n = 90) who had undergone carotid endarterectomy (CEA) and controls (n = 82) as well as to determine the prevalence of HCMV immediate early (IE) and late (LA) antigens in carotid atherosclerotic plaques obtained from 89 patients. In addition, we sought to determine whether HCMV infection was associated with inflammatory activity in the plaque by quantifying infiltrating CD3 and CD68 positive cells and 5-LO immunoreactivity. Methods HCMV serology was assessed with ELISA and immunohistochemistry staining was performed to detect HCMV antigens, CD3, CD68 and 5-LO reactivity. The Fisher’s exact test was used to compare i) seroprevalence of HCMV IgG between patients and controls and ii) HCMV-positive or –negative to that of CD3, CD68 and 5-LO immunoreactive cells in plaque samples. The student-t test was performed to connote the significance level of mean optical density between patients and controls. Results The seroprevalence for HCMV IgG was high in both patients and controls (99% and 98%, respectively). Controls had significantly higher IgG titers for HCMV compared with patients (p = 0.0148). Strikingly, we found a high prevalence of HCMV antigens in atherosclerotic plaques; 57/89 (64%) and 47/87 (54%) were HCMV IE and LA positive, respectively. Most plaques had rather low HCMV reactivity with distinct areas of HCMV-positive cells mainly detected in shoulder regions of the plaques, but also in the area adjacent to the necrotic core and fibrous cap. In plaques, the cellular targets for HCMV infection appeared to be mainly macrophages/foam cells and smooth muscle cells. HCMV-positive plaques trended to be associated with increased numbers of CD68 positive macrophages and CD3 positive T cells, while 5-LO reactivity was high in both HCMV-positive and HCMV-negative plaques. Conclusions In Russian patients undergoing CEA, HCMV proteins are abundantly expressed in carotid plaques and may contribute to the inflammatory response in plaques via enhanced infiltration of CD68 and CD3 cells.
Collapse
Affiliation(s)
- Koon-Chu Yaiw
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Olga Ovchinnikova
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden.,Almazov Federal Center for Heart, Blood and Endocrinology, St. Petersburg, Russia
| | - Chato Taher
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Abdul-Aleem Mohammad
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Belghis Davoudi
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Eugene Shlyakhto
- Almazov Federal Center for Heart, Blood and Endocrinology, St. Petersburg, Russia
| | - Oxana Rotar
- Almazov Federal Center for Heart, Blood and Endocrinology, St. Petersburg, Russia
| | - Alexandra Konradi
- Almazov Federal Center for Heart, Blood and Endocrinology, St. Petersburg, Russia
| | - Vanessa Wilhelmi
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Afsar Rahbar
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Lynn Butler
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Alice Assinger
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| | - Cecilia Söderberg-Nauclér
- Department of Medicine, Center for Molecular Medicine, CMM L8:03, Karolinska Institutet, Solna, Stockholm SE-171 76, Sweden
| |
Collapse
|
21
|
Wall NA, Chue CD, Edwards NC, Pankhurst T, Harper L, Steeds RP, Lauder S, Townend JN, Moss P, Ferro CJ. Cytomegalovirus seropositivity is associated with increased arterial stiffness in patients with chronic kidney disease. PLoS One 2013; 8:e55686. [PMID: 23451030 PMCID: PMC3581505 DOI: 10.1371/journal.pone.0055686] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 12/28/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Patients with chronic kidney disease have an increased cardiovascular risk that is not fully explained by traditional risk factors but appears to be related to increased arterial stiffness. Cytomegalovirus (CMV) infection is associated with increased cardiovascular risk although the mechanisms for this are unknown. We examined whether CMV seropositivity was associated with increased arterial stiffness in patients with chronic kidney disease. METHODOLOGY AND PRINCIPAL FINDINGS In 215 non-diabetic patients with chronic kidney disease, CMV seropositivity was determined using an anti-CMV IgG ELISA. Pulse wave velocity was measured and aortic distensibility assessed in the ascending, proximal descending and distal descending thoracic aorta. Patients seropositive for CMV had a higher pulse wave velocity and lower aortic distensibility at all 3 levels. These differences (except for ascending aortic distensibility) persisted in a subcohort matched for age, gender and renal function, and when the whole cohort was divided into quartiles of age. In multivariable analyses, CMV seropositivity was an independent determinant of pulse wave velocity and proximal and distal descending aortic distensibility. CONCLUSIONS In patients with chronic kidney disease, CMV seropositivity is associated with increased arterial stiffness and decreased distensibility of the proximal descending and distal aorta. These findings suggest that further research is required to examine CMV as a possible cause of arterial disease and increased cardiovascular risk in patients with CKD and may be relevant more widely for CMV seropositive patients with normal renal function.
Collapse
Affiliation(s)
- Nadezhda A. Wall
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
- Department of Nephrology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Colin D. Chue
- School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Nicola C. Edwards
- School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Tanya Pankhurst
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Lorraine Harper
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
- Department of Nephrology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Richard P. Steeds
- School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Sarah Lauder
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Jonathan N. Townend
- School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Paul Moss
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Charles J. Ferro
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
- Department of Nephrology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Naing Z. Is cytomegalovirus infection causative for coronary heart disease? MICROBIOLOGY AUSTRALIA 2013. [DOI: 10.1071/ma13045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
23
|
Tang-Feldman YJ, Lochhead SR, Lochhead GR, Yu C, George M, Villablanca AC, Pomeroy C. Murine cytomegalovirus (MCMV) infection upregulates P38 MAP kinase in aortas of Apo E KO mice: a molecular mechanism for MCMV-induced acceleration of atherosclerosis. J Cardiovasc Transl Res 2012. [PMID: 23192592 DOI: 10.1007/s12265-012-9428-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Multiple studies suggest an association between cytomegalovirus (CMV) infection and atherogenesis; however, the molecular mechanisms by which viral infection might exacerbate atherosclerosis are not well understood. Aortas of MCMV-infected and uninfected Apo E knockout (KO) mice were analyzed for atherosclerotic lesion development and differential gene expression. Lesions in the infected mice were larger and showed more advanced disease compared to the uninfected mice. Sixty percent of the genes in the MAPK pathway were upregulated in the infected mice. p38 and ERK 1/2 MAPK genes were 5.6- and 2.0-fold higher, respectively, in aortas of infected vs. uninfected mice. Levels of VCAM-1, ICAM-1, and MCP-1 were ~2.0-2.6-fold higher in aortas of infected vs. uninfected mice. Inhibition of p38 with SB203580 resulted in lower levels of pro-atherogenic molecules and MCMV viral load in aortas of infected mice. MCMV-induced upregulation of p38 may drive the virus-induced acceleration of atherogenesis observed in our model.
Collapse
|
24
|
Infection of vascular endothelial cells with human cytomegalovirus under fluid shear stress reveals preferential entry and spread of virus in flow conditions simulating atheroprone regions of the artery. J Virol 2012; 86:13745-55. [PMID: 23055562 DOI: 10.1128/jvi.02244-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Atherosclerosis is a major pathogenic factor in cardiovascular diseases, which are the leading cause of mortality in developed countries. While risk factors for atherosclerosis tend to be systemic, the distribution of atherosclerotic plaques within the vasculature is preferentially located at branch points and curves where blood flow is disturbed and shear stress is low. It is now widely accepted that hemodynamic factors can modulate endothelial gene expression and function and influence the pathophysiological changes associated with atherosclerosis. Human cytomegalovirus (HCMV), a ubiquitous pathogen, has long been proposed as a risk factor for atherosclerosis. To date, the role of HCMV in atherogenesis has been explored only in static conditions, and it is not known how HCMV infection is influenced by the physiological context of flow. In this study, we utilized a parallel-plate flow system to simulate the effects of shear stresses in different regions of the vasculature in vitro. We found that endothelial cells cultured under low shear stress, which simulates the flow condition of atheroprone regions in vivo, are more permissive to HCMV infection than cells experiencing high shear stress or static conditions. Cells exposed to low shear stress show increased entry of HCMV compared to cells exposed to high shear stress or static conditions. Viral structural gene expression, viral titers, and viral spread are also enhanced in endothelial cells exposed to low shear stress. These results suggest that hemodynamic factors modulate HCMV infection of endothelial cells, thus providing new insights into the induction/acceleration of atherosclerosis by HCMV.
Collapse
|
25
|
Gkrania-Klotsas E, Langenberg C, Sharp SJ, Luben R, Khaw KT, Wareham NJ. Higher immunoglobulin G antibody levels against cytomegalovirus are associated with incident ischemic heart disease in the population-based EPIC-Norfolk cohort. J Infect Dis 2012; 206:1897-903. [PMID: 23045624 DOI: 10.1093/infdis/jis620] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) is associated with ischemic heart disease (IHD) among organ transplant recipients. The relationship between the levels of antibody for CMV with IHD in immunocompetent individuals is uncertain. METHODS We measured baseline CMV immunoglobulin G (IgG) levels in 12,574 participants without IHD from the population-based EPIC-Norfolk cohort, aged 40-70 years old at recruitment in 1993-1997. Underlying causes of death or hospitalization until 31 March 2008 were abstracted from death certificates and a database of hospital admissions, respectively. RESULTS Of the participants, 58% were seropositive for CMV. After a mean follow-up of 12 years (standard deviation, 2.2 years), 1356 first-time IHD events occurred. After adjustment for classic IHD risk factors, belonging to the highest antibody group was associated with an increased risk of incident IHD, compared with seronegativity (hazard ratio, 1.22; 95% confidence interval, 1.05-1.42). After additional adjustment for measures of social class, inflammation, and possible confounders, this association was unchanged (hazard ratio, 1.21; 95% confidence interval, 1.04-11.41). CONCLUSIONS This is the first population study to show that CMV IgG antibody levels are related to incident IHD compared to seronegativity. Studies correlating CMV antibody levels with direct measurements of active infection will be necessary.
Collapse
Affiliation(s)
- Effrossyni Gkrania-Klotsas
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom.
| | | | | | | | | | | |
Collapse
|
26
|
Pastano R, Dell'Agnola C, Bason C, Gigli F, Rabascio C, Puccetti A, Tinazzi E, Cetto G, Peccatori F, Martinelli G, Lunardi C. Antibodies against human cytomegalovirus late protein UL94 in the pathogenesis of scleroderma-like skin lesions in chronic graft-versus-host disease. Int Immunol 2012; 24:583-91. [PMID: 22773152 DOI: 10.1093/intimm/dxs061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (hCMV) infection and its reactivation correlate both with the increased risk and with the worsening of graft-versus-host disease (GVHD). Because scleroderma-like skin lesions can occur in chronic GVHD (cGVHD) in allogeneic stem-cell transplant (HCT) patients and hCMV is relevant in the pathogenesis of systemic sclerosis (SSc), we evaluated the possible pathogenetic link between hCMV and skin cGVHD. Plasma from 18 HCT patients was tested for anti-UL94 and/or anti-NAG-2 antibodies, identified in SSc patients, by direct ELISA assays. Both donors and recipients were anti-hCMV IgG positive, without autoimmune diseases. Patients' purified anti-UL94 and anti-NAG-2 IgG binding to human umbilical endothelial cells (HUVECs) and fibroblasts was performed by FACS analysis and ELISA test. HUVECs apoptosis and fibroblasts proliferation induced by patients' anti-NAG-2 antibodies were measured by DNA fragmentation and cell viability, respectively. About 11/18 patients developed cGVHD and all of them showed skin involvement, ranging from diffuse SSc-like lesions to limited erythema. Eight of eleven cGVHD patients were positive for anti-UL94 and/or anti-NAG-2 antibodies. Remarkably, 4/5 patients who developed diffuse or limited SSc-like lesions had antibodies directed against both UL94 and NAG-2; their anti-NAG-2 IgG-bound HUVECs and fibroblasts induce both endothelial cell apoptosis and fibroblasts proliferation, similar to that induced by purified anti-UL94 and anti-NAG-2 antibodies obtained from SSc patients. In conclusion, our data suggest a pathogenetic link between hCMV infection and scleroderma-like skin cGVHD in HCT patients through a mechanism of molecular mimicry between UL94 viral protein and NAG-2 molecule, as observed in patients with SSc.
Collapse
Affiliation(s)
- Rocco Pastano
- Hematoncology Division, Bone Marrow Transplant Unit, European Institute of Oncology, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ji YN, An L, Zhan P, Chen XH. Cytomegalovirus infection and coronary heart disease risk: a meta-analysis. Mol Biol Rep 2012; 39:6537-46. [PMID: 22311014 DOI: 10.1007/s11033-012-1482-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 01/24/2012] [Indexed: 12/11/2022]
Abstract
The chronic inflammatory process including cytomegalovirus (CMV) infection has been hypothesized to induce the progression of atherosclerosis in coronary heart disease (CHD). Numbers studies were conducted to analyze the association between CMV infection and risk of CHD, but no clear consensus had been reached. To assess this relationship more precisely, a meta-analysis was performed. The electronic databases PubMed, Embase, and CNKI were searched; data were extracted and analyzed independently by two investigators. Ultimately, 55 studies, involving 9,000 cases and 8,608 controls from six prospective studies (all with a nested case-control design) and 49 retrospective case-control studies were included. Overall, people exposed to CMV infection had an odds ratio (OR) of 1.67 (95% CI, 1.56-1.79) for CHD risk, relative to those not exposed. CMV infection was clearly identified as a risk factor for CHD in both prospective studies (OR, 1.31; 95% CI, 1.132-1.517) and retrospective studies (OR, 1.79; 95% CI, 1.659-1.939), and in both Asian group (OR, 2.69; 95% CI, 2.304-3.144) and non-Asian group (OR, 1.48; 95% CI, 1.371-1.600). Interestingly, in the subgroup analyses by detection methods of CMV, the increased risk (OR, 8.121) was greater among studies using polymerase chain reaction than the risk (OR, 1.561) among studies using enzyme-linked immunosorbent assay. In conclusion, this meta-analysis suggested that CMV infection is associated with an increased risk for CHD, especially among Asian populations.
Collapse
Affiliation(s)
- Ya-Nan Ji
- Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | | | | | | |
Collapse
|
28
|
Haarala A, Kähönen M, Lehtimäki T, Aittoniemi J, Jylhävä J, Hutri-Kähönen N, Taittonen L, Laitinen T, Juonala M, Viikari J, Raitakari OT, Hurme M. Relation of high cytomegalovirus antibody titres to blood pressure and brachial artery flow-mediated dilation in young men: the Cardiovascular Risk in Young Finns Study. Clin Exp Immunol 2012; 167:309-16. [PMID: 22236008 PMCID: PMC3278698 DOI: 10.1111/j.1365-2249.2011.04513.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2011] [Indexed: 12/18/2022] Open
Abstract
Human cytomegalovirus (CMV) infection is associated with a higher risk of cardiovascular disease in immunocompromised organ transplant patients. It has been linked with the pathogenesis of elevated arterial blood pressure. However, controversy exists as to whether CMV infection is associated with endothelial function, and little is known about its role as a potential risk factor for early atherosclerosis development at a young age. We aimed to discover if CMV antibody titres are associated with early vascular changes (carotid intima-media thickness, carotid artery distensibility and brachial artery flow-mediated dilation), blood pressure elevation or other traditional cardiovascular risk factors. CMV antibody titres were measured in 1074 women and 857 men (aged 24-39 years) taking part in the Cardiovascular Risk in Young Finns study. CMV antibody titres were significantly higher in women compared to men. In men, high CMV antibody titres were associated directly with age (P < 0·001) and systolic (P = 0·053) and diastolic (P = 0·002) blood pressure elevation, and associated inversely with flow-mediated dilation (P = 0·014). In women, CMV antibody titres did not associate with any of the analysed parameters. In a multivariate regression model, which included traditional atherosclerotic risk factors, CMV antibody titres were independent determinants for systolic (P = 0·029) and diastolic (P = 0·004) blood pressure elevation and flow-mediated dilation (P = 0·014) in men. High CMV antibody titres are associated independently with blood pressure and brachial artery flow-mediated dilation in young men. This association supports the hypothesis that common CMV infection and/or an immune response to CMV may lead to impaired vascular function at a young age.
Collapse
Affiliation(s)
- A Haarala
- Department of Microbiology and Immunology, University of Tampere, Medical School, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Leskov IL, Whitsett J, Vasquez-Vivar J, Stokes KY. NAD(P)H oxidase and eNOS play differential roles in cytomegalovirus infection-induced microvascular dysfunction. Free Radic Biol Med 2011; 51:2300-8. [PMID: 22033010 PMCID: PMC3272703 DOI: 10.1016/j.freeradbiomed.2011.09.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 09/26/2011] [Accepted: 09/28/2011] [Indexed: 12/22/2022]
Abstract
Primary cytomegalovirus (CMV) infection promotes oxidative stress and reduces nitric oxide (NO) bioavailability in endothelial cells. These events are among the earliest vascular responses to cardiovascular risk factors. We assessed the roles of NAD(P)H oxidase and NO bioavailability in microvascular responses to persistent CMV infection alone or with hypercholesterolemia. Wild-type (WT) or gp91(phox) (NAD(P)H oxidase subunit) knockout mice received mock inoculum or 3×10(4) PFU murine CMV (mCMV) ip 5 weeks before placement on a normal or high-cholesterol diet (HC) for 4 weeks before assessment of arteriolar function and venular blood cell recruitment using intravital microscopy. Some WT groups received sepiapterin (a precursor of the nitric oxide synthase cofactor tetrahydrobiopterin) or apocynin (NAD(P)H oxidase inhibitor/antioxidant). Endothelium-dependent vasodilation was impaired in mCMV vs mock WT, regardless of diet. This was not affected by sepiapterin, and pharmacological inhibition of nitric oxide synthase reduced dilation similarly in mock and mCMV mice. Apocynin or deficiency of total, but not blood cell or vascular wall only (tested using bone marrow chimeras), gp91(phox) protected against arteriolar dysfunction. Blood cell recruitment was induced by mCMV-HC. Sepiapterin, but not NAD(P)H oxidase deficiency/apocynin, reduced leukocyte accumulation, whereas platelet adhesion was reduced by sepiapterin, apocynin, or total, platelet-specific, or vascular wall gp91(phox) deficiency. These data implicate activation of both hematopoietic and vessel wall NAD(P)H oxidase in mCMV-induced arteriolar dysfunction and platelet and vascular NAD(P)H oxidase in the thrombogenic phenotype induced by mCMV-HC. In contrast, findings with sepiapterin suggest that eNOS dysfunction, perhaps uncoupling, mediates venular, but not arteriolar, responses to mCMV-HC, thus indicating that NAD(P)H oxidase and eNOS differentially regulate microvascular responses to mCMV.
Collapse
Affiliation(s)
- Igor L. Leskov
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130
| | - Jennifer Whitsett
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | - Karen Y. Stokes
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130
- Center for Molecular and Tumor Virology, LSU Health Sciences Center, Shreveport, LA 71130
| |
Collapse
|
30
|
|
31
|
Adler SP, Best AM, Marshall B, Vetrovec GW. Infection with cytomegalovirus is not associated with premature mortality. Infect Dis Rep 2011; 3:e17. [PMID: 24470914 PMCID: PMC3892592 DOI: 10.4081/idr.2011.e17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 10/30/2011] [Indexed: 11/23/2022] Open
Abstract
Over 90% of the world's population acquires a cytomegalovirus (CMV) infection. This infection, although asymptomatic or self-limiting, is a major burden to the immune system. For this reason, and because CMV immunization is possible, determining whether CMV can cause reduced longevity, particularly among those with coronary artery disease, is important and previous reports have been conflicting. Thus our objective was to assess the association between CMV infection as defined serologically and antibody levels against CMV and long-term survival (18 years). We completed a prospective observational cohort study of 915 consecutive patients (mean age 58 years) undergoing coronary angiography. CMV immunoglobulin levels were measured at baseline using either a whole cell CMV antigen or a purified protein antigen (gB). After adjustment for potentially confounding variables (age, race, gender, body mass index, the presence or absence of coronary artery disease, the number of diseased vessels, diabetes, renal disease, hypertension, dialysis, congestive heart failure, and the maximum percent reduction in luminal diameter), Cox's proportional hazards models showed no association between CMV seropositivity or levels of antibodies against CMV by either assay and longevity for both patients with or without coronary artery disease (CAD) nor for those under or over 70 years of age at baseline. Our observations suggest that universal immunization against CMV may not improve longevity.
Collapse
Affiliation(s)
- Stuart P Adler
- Division of Infectious Diseases, Department of Pediatrics
| | | | - Beth Marshall
- Division of Infectious Diseases, Department of Pediatrics
| | - George W Vetrovec
- Division of Cardiology, Department of Internal Medicine, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond VA, USA
| |
Collapse
|
32
|
Senchenkov E, Khoretonenko MV, Leskov IL, Ostanin DV, Stokes KY. P-selectin mediates the microvascular dysfunction associated with persistent cytomegalovirus infection in normocholesterolemic and hypercholesterolemic mice. Microcirculation 2011; 18:452-62. [PMID: 21457388 DOI: 10.1111/j.1549-8719.2011.00106.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Cytomegalovirus has been implicated in cardiovascular disease, possibly through the induction of inflammatory processes. P-selectin and L-selectin are adhesion molecules that mediate early microvascular responses to inflammatory stimuli. This study examined the role of these selectins in the microvascular dysfunction that occurs during persistent CMV infection. METHODS C57Bl/6, P- or L-selectin-deficient mice were mock-inoculated or infected with murine CMV, and five weeks later placed on normal diet or high cholesterol diet for six weeks. P-selectin expression was measured or intravital microscopy was performed to determine arteriolar vasodilation and venular blood cell recruitment. RESULTS P-selectin expression was significantly increased in the heart, lung, and spleen of mCMV-ND, but not mCMV-HC C57Bl/6. mCMV-ND and mCMV-HC exhibited impaired arteriolar function, which was reversed by treatment with an anti-P-selectin antibody, but not L-selectin deficiency. mCMV-HC also showed elevated leukocyte and platelet recruitment. P-selectin inhibition abrogated, whereas L-selectin deficiency partially reduced these responses. CONCLUSIONS We provide the first evidence for P-selectin upregulation by persistent mCMV infection and implicate this adhesion molecule in the associated arteriolar dysfunction. P-selectin, and to a lesser extent L-selectin, mediates the leukocyte and platelet recruitment induced by CMV infection combined with hypercholesterolemia.
Collapse
Affiliation(s)
- Evgeny Senchenkov
- Department of Molecular and Cellular Physiology Center for Molecular and Tumor Virology Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | | | | | | | | |
Collapse
|
33
|
Genome-wide association study does not reveal major genetic determinants for anti-cytomegalovirus antibody response. Genes Immun 2011; 13:184-90. [PMID: 21993531 DOI: 10.1038/gene.2011.71] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cytomegalovirus (CMV) causes an infection, which is followed by a lifelong latency. CMV has received much attention in clinical studies, but little is known about the genetic basis of this common infection. To identify genetic polymorphisms associated with the susceptibility to and strength of anti-CMV immunoglobulin G (IgG) response to CMV infection, we conducted a genome-wide association study (GWAS) using an Illumina BeadChip containing 670 000 probes and participants from the Cardiovascular Risk in Young Finns Study, including 1486 anti-CMV IgG seropositive and 648 seronegative individuals. Statistical analyses were performed using logistic (for susceptibility) and linear regression (for strength of antibody response). None of single-nucleotide polymorphisms (SNPs) was found to be associated with susceptibility to CMV infection at the level of genome-wide significance (P<5 × 10(-8)). Also, none of the association signals identified reached genome-wide levels of statistical significance in the study of the strength of the antibody response to CMV although five SNPs in AGBL1 gene region displayed a suggestive association (lowest P-value=1.86 × 10(-6)). The results indicate that there is no strong evidence of major host genetic factors involved in either susceptibility to or the strength of antibody response to human CMV infection.
Collapse
|
34
|
Khoretonenko MV, Leskov IL, Jennings SR, Yurochko AD, Stokes KY. Cytomegalovirus infection leads to microvascular dysfunction and exacerbates hypercholesterolemia-induced responses. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2134-44. [PMID: 20802174 DOI: 10.2353/ajpath.2010.100307] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cytomegalovirus (CMV) persistently infects more than 60% of the worldwide population. In immunocompetent hosts, it has been implicated in several diseases, including cardiovascular disease, possibly through the induction of inflammatory pathways. Cardiovascular risk factors promote an inflammatory phenotype in the microvasculature long before clinical disease is evident. This study determined whether CMV also impairs microvascular homeostasis and synergizes with hypercholesterolemia to exaggerate these responses. Intravital microscopy was used to assess endothelium-dependent and -independent arteriolar vasodilation and venular leukocyte and platelet adhesion in mice after injection with either mock inoculum or murine CMV (mCMV). Mice were fed a normal (ND) or high-cholesterol (HC) diet beginning at 5 weeks postinfection (p.i.), or a HC diet for the final 4 weeks of infection. mCMV-ND mice exhibited impaired endothelium-dependent vasodilation versus mock-ND at 9 and 12 weeks and endothelium-independent arteriolar dysfunction by 24 weeks. Transient mild leukocyte adhesion occurred in mCMV-ND venules at 7 and 21 weeks p.i. HC alone caused temporary arteriolar dysfunction and venular leukocyte and platelet recruitment, which were exaggerated and prolonged by mCMV infection. The time of introduction of HC after mCMV infection determined whether mCMV+HC led to worse venular inflammation than either factor alone. These findings reveal a proinflammatory influence of persistent mCMV on the microvasculature, and suggest that mCMV infection enhances microvasculature susceptibility to both inflammatory and thrombogenic responses caused by hypercholesterolemia.
Collapse
Affiliation(s)
- Mikhail V Khoretonenko
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | | | | | | | |
Collapse
|
35
|
Roberts ET, Haan MN, Dowd JB, Aiello AE. Cytomegalovirus antibody levels, inflammation, and mortality among elderly Latinos over 9 years of follow-up. Am J Epidemiol 2010; 172:363-71. [PMID: 20660122 DOI: 10.1093/aje/kwq177] [Citation(s) in RCA: 198] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study examined the relation between immune response to cytomegalovirus (CMV) and all-cause and cardiovascular disease (CVD) mortality, and possible mediating mechanisms. Data were derived from the Sacramento Area Latino Study on Aging, a population-based study of older Latinos (aged 60-101 years) in California followed in 1998-2008. CMV immunoglobulin G (IgG), tumor necrosis factor, and interleukin-6 were assayed from baseline blood draws. Data on all-cause and CVD mortality were abstracted from death certificates. Analyses included 1,468 of 1,789 participants. For individuals with CMV IgG antibody titers in the highest quartile compared with lower quartiles, fully adjusted models showed that all-cause mortality was 1.43 times (95% confidence interval: 1.14, 1.79) higher over 9 years. In fully adjusted models, the hazard of CVD mortality was also elevated (hazard ratio = 1.35, 95% confidence interval: 1.01, 1.80). A composite measure of tumor necrosis factor and interleukin-6 mediated a substantial proportion of the association between CMV and all-cause (18.9%, P < 0.001) and CVD (29.0%, P = 0.02) mortality. This study is the first known to show that high CMV IgG antibody levels are significantly related to mortality and that the relation is largely mediated by interleukin-6 and tumor necrosis factor. Further studies investigating methods for reducing IgG antibody response to CMV are warranted.
Collapse
Affiliation(s)
- Eric T Roberts
- Department of Epidemiology, 3659 SPH Tower, 109 Observatory, Ann Arbor, MI 48104-2548, USA
| | | | | | | |
Collapse
|
36
|
Wang GC, Kao WHL, Murakami P, Xue QL, Chiou RB, Detrick B, McDyer JF, Semba RD, Casolaro V, Walston JD, Fried LP. Cytomegalovirus infection and the risk of mortality and frailty in older women: a prospective observational cohort study. Am J Epidemiol 2010; 171:1144-52. [PMID: 20400465 DOI: 10.1093/aje/kwq062] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cytomegalovirus (CMV), a prevalent pathogen, causes severe disease in immunocompromised humans. However, present understanding is limited regarding the long-term clinical effect of persistent CMV infection in immunocompetent adults. The authors conducted a prospective observational cohort study (1992-2002) of 635 community-dwelling women in Baltimore, Maryland, aged 70-79 years in the Women's Health and Aging Studies to examine the effect of CMV infection on the risk of frailty, a common geriatric syndrome, and mortality in older women. The effect of baseline serum CMV antibody (immunoglobulin G) concentration on the risk of 3-year incident frailty, defined by using a 5-component measure, and 5-year mortality was examined with Cox proportional hazards models. Compared with those who were CMV seronegative, women in the highest quartile of CMV antibody concentration had a greater incidence of frailty (hazard ratio = 3.46, 95% confidence interval: 1.45, 8.27) and mortality (hazard ratio = 3.81, 95% confidence interval: 1.64, 8.83). After adjustment for potential confounders, CMV antibody concentration in the highest quartile independently increased the risk of 5-year mortality (hazard ratio = 2.79, 95% confidence interval: 1.22, 6.40). Better understanding of the long-term clinical consequences of CMV infection in immunocompetent humans is needed to guide public health efforts for this widely prevalent infection.
Collapse
Affiliation(s)
- George C Wang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ho RCM, Neo LF, Chua ANC, Cheak AAC, Mak A. Research on Psychoneuroimmunology: Does Stress Influence Immunity and Cause Coronary Artery Disease? ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2010. [DOI: 10.47102/annals-acadmedsg.v39n3p191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This review addresses the importance of psychoneuroimmunology (PNI) studies in understanding the role of acute and chronic psychological stressors on the immune system and development of coronary artery disease (CAD). Firstly, it illustrates how psychological stressors change endothelial function and lead to chemotaxis. Secondly, acute psychological stressors lead to leukocytosis, increased natural killer cell cytotoxicity and reduced proliferative response to mitogens while chronic psychological stressors may lead to adverse health effects. This will result in changes in cardiovascular function and development of CAD. Thirdly, acute and chronic psychological stressors will increase haemostatic factors and acute phase proteins, possibly leading to thrombus formation and myocardial infarction. The evidence for the effects of acute and chronic psychological stress on the onset and progression of CAD is consistent and convincing. This paper also highlights potential research areas and implications of early detection of immunological changes and cardiovascular risk in people under high psychological stress.
Key words: Cardiovascular, Inflammation, Psychoneuroimmunology, Stress
Collapse
Affiliation(s)
- Roger CM Ho
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Li Fang Neo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anna NC Chua
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alicia AC Cheak
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anselm Mak
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
38
|
Bicaudal D1-dependent trafficking of human cytomegalovirus tegument protein pp150 in virus-infected cells. J Virol 2010; 84:3162-77. [PMID: 20089649 DOI: 10.1128/jvi.01776-09] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) virion assembly takes place in the nucleus and cytoplasm of infected cells. The HCMV virion tegument protein pp150 (ppUL32) is an essential protein of HCMV and has been suggested to play a role in the cytoplasmic phase of HCMV assembly. To further define its role in viral assembly and to identify host cell proteins that interact with pp150 during viral assembly, we utilized yeast two-hybrid analyses to detect an interaction between pp150 and Bicaudal D1 (BicD1), a protein thought to play a role in trafficking within the secretory pathway. BicD1 is known to interact with the dynein motor complex and the Rab6 GTPase. The interaction between pp150 and BicD1 was confirmed by coimmunoprecipitation and fluorescence resonance energy transfer. Depletion of BicD1 with short hairpin RNA (shRNA) caused decreased virus yield and a defect in trafficking of pp150 to the cytoplasmic viral assembly compartment (AC), without altering trafficking to the AC of another essential tegument protein, pp28, or the viral glycoprotein complex gM/gN. The C terminus of BicD1 has been previously shown to interact with the GTPase Rab6, suggesting a potential role for Rab6-mediated vesicular trafficking in HCMV assembly. Finally, overexpression of the N terminus of truncated BicD1 acts in a dominant-negative manner and leads to disruption of the AC and a decrease in the assembly of infectious virus. This phenotype was similar to that observed following overexpression of dynamitin (p50) and provided additional evidence that morphogenesis of the AC and virus assembly were dynein dependent.
Collapse
|
39
|
Williamson CD, Colberg-Poley AM. Access of viral proteins to mitochondria via mitochondria-associated membranes. Rev Med Virol 2009; 19:147-64. [PMID: 19367604 DOI: 10.1002/rmv.611] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
By exploiting host cell machineries, viruses provide powerful tools for gaining insight into cellular pathways. Proteins from two unrelated viruses, human CMV (HCMV) and HCV, are documented to traffic sequentially from the ER into mitochondria, probably through the mitochondria-associated membrane (MAM) compartment. The MAM are sites of ER-mitochondrial contact enabling the direct transfer of membrane bound lipids and the generation of high calcium (Ca2+) microdomains for mitochondria signalling and responses to cellular stress. Both HCV core protein and HCMV UL37 proteins are associated with Ca2+ regulation and apoptotic signals. Trafficking of viral proteins to the MAM may allow viruses to manipulate a variety of fundamental cellular processes, which converge at the MAM, including Ca2+ signalling, lipid synthesis and transfer, bioenergetics, metabolic flow, and apoptosis. Because of their distinct topologies and targeted MAM sub-domains, mitochondrial trafficking (albeit it through the MAM) of the HCMV and HCV proteins predictably involves alternative pathways and, hence, distinct targeting signals. Indeed, we found that multiple cellular and viral proteins, which target the MAM, showed no apparent consensus primary targeting sequences. Nonetheless, these viral proteins provide us with valuable tools to access the poorly characterised MAM compartment, to define its cellular constituents and describe how virus infection alters these to its own end. Furthermore, because proper trafficking of viral proteins is necessary for their function, discovering the requirements for MAM to mitochondrial trafficking of essential viral proteins may provide novel targets for the rational design of anti-viral drugs.
Collapse
Affiliation(s)
- Chad D Williamson
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, 111 Michigan Ave, NW, Washington, DC 20010, USA.
| | | |
Collapse
|
40
|
Liu R, Moroi M, Yamamoto M, Kubota T, Ono T, Funatsu A, Komatsu H, Tsuji T, Hara H, Hara H, Nakamura M, Hirai H, Yamaguchi T. Presence and severity of Chlamydia pneumoniae and Cytomegalovirus infection in coronary plaques are associated with acute coronary syndromes. Int Heart J 2007; 47:511-9. [PMID: 16960406 DOI: 10.1536/ihj.47.511] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although an association between Chlamydia pneumoniae (Cpn) or Cytomegalovirus (CMV) infection and coronary atherosclerosis has been reported, such an association is less clear for acute coronary syndromes (ACS). The purpose of this study was to investigate the pathogenic roles of Cpn and CMV infection of coronary plaques in ACS. We divided 38 coronary plaque specimens obtained from 38 patients who underwent directional coronary atherectomy or thrombectomy into an ACS group (n = 21) and a non-ACS group (n = 17). Cpn and CMV in specimens were stained using immunohistochemical techniques and analyzed quantitatively. The detection rate for either Cpn- or CMV-positive cells in ACS patients was slightly higher compared with non-ACS patients. Detection rates for both Cpn- and CMV-positive cells were significantly higher in ACS patients than in non-ACS patients (P = 0.010). Furthermore, the density of Cpn- and CMV-positive cells in plaques was significantly higher in ACS patients than in non-ACS patients (P < 0.003). The results indicate that the presence and severity of Cpn and CMV infection in coronary plaques are greater in patients with ACS compared with non-ACS patients. We conclude that infection with Cpn and CMV in coronary plaques may be involved in the pathogenesis of ACS.
Collapse
Affiliation(s)
- Ruiqin Liu
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Ohashi, Meguro-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chow EKH, Razani B, Cheng G. Innate immune system regulation of nuclear hormone receptors in metabolic diseases. J Leukoc Biol 2007; 82:187-95. [PMID: 17314330 DOI: 10.1189/jlb.1206741] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The immune system modulates a number of biological processes to properly defend against pathogens. Here, we review how crosstalk between nuclear hormone receptors and the innate immune system may influence multiple biological functions during an immune response. Although nuclear hormone receptor repression of innate immune responses and inflammation has been well studied, a number of new studies have identified repression of nuclear hormone receptor signaling by various innate immune responses. IFN regulatory factor 3, a key transcription factor involved in the induction of antiviral genes, may play a role in mediating such crosstalk between the innate immune response and nuclear receptor-regulated metabolism. This crosstalk mechanism is now implicated in the pathogenesis of atherosclerosis and Reye's syndrome and could provide an explanation for other pathogen-associated metabolic and developmental disorders.
Collapse
Affiliation(s)
- Edward Kai-Hua Chow
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
42
|
Gibson FC, Yumoto H, Takahashi Y, Chou HH, Genco CA. Innate immune signaling and Porphyromonas gingivalis-accelerated atherosclerosis. J Dent Res 2006; 85:106-21. [PMID: 16434728 DOI: 10.1177/154405910608500202] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Periodontal diseases are a group of diseases that lead to erosion of the hard and soft tissues of the periodontium, which, in severe cases, can result in tooth loss. Anecdotal clinical observations have suggested that poor oral health may be associated with poor systemic health; however, only recently have appropriate epidemiological studies been initiated, with defined clinical endpoints of periodontal disease, to address the association of periodontal disease with increased risk for cardiovascular and cerebrovascular disease. Although conflicting reports exist, these epidemiological studies support this connection. Paralleling these epidemiological studies, emerging basic scientific studies also support that infection may represent a risk factor for atherosclerosis. With P. gingivalis as a model pathogen, in vitro studies support that this organism can activate host innate immune responses associated with atherosclerosis, and in vivo studies demonstrate that this organism can accelerate atheroma deposition in animal models. In this review, we focus primarily on the basic scientific studies performed to date which support that infection with bacteria, most notably P. gingivalis, accelerates atherosclerosis. Furthermore, we attempt to bring together these studies to provide an up-to-date framework of emerging theories into the mechanisms underlying periodontal disease and increased risk for atherosclerosis, as well as identify intervention strategies to reduce the incidence of periodontal disease in humans, in an attempt to decrease risk for systemic complications of periodontal disease such as atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- F C Gibson
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Evans Biomedical Research Center, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
43
|
Blans MCA, Visseren FLJ, Banga JD, Hoekstra JBL, van der Graaf Y, Diepersloot RJA, Bouter KP. Infection induced inflammation is associated with erectile dysfunction in men with diabetes. Eur J Clin Invest 2006; 36:497-502. [PMID: 16796607 DOI: 10.1111/j.1365-2362.2006.01653.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND In diabetic patients with erectile dysfunction, endothelial dysfunction is a major underlying cause. Infection-induced inflammation may be associated with endothelial dysfunction. The goal of this study was to determine whether erectile dysfunction in patients with diabetes is associated with infections of Chlamydia pneumoniae or cytomegalovirus and/or with low-grade inflammation. MATERIALS AND METHODS Diabetic patients, 57 with and 33 without erectile dysfunction, were enrolled in a case-control study. Both groups of patients consists of type 1 and type 2 diabetics. Serum antibodies against cytomegalovirus and C. pneumoniae and markers of inflammation, including high-sensitivity C-reactive protein and fibrinogen, were measured. RESULTS Adjusted odds ratios for erectile dysfunction in cytomegalovirus IgG, C. pneumoniae IgG and C. pneumoniae IgA seropositive men were 2.4 (95%CI; 1.0-6.0), 3.0 (95%CI; 1.2-8.1) and 1.8 (95%CI; 0.7-4.6), respectively. Odds ratios for the highest tertiles of high-sensitivity C-reactive protein and fibrinogen concentrations compared to the lowest tertile were 4.3 (95%CI; 1.4-13.1) and 6.6 (95%CI; 2.1-21.2), respectively. CONCLUSION Elevated high-sensitivity C-reactive protein or fibrinogen serum levels and infection with cytomegalovirus or C. pneumoniae were associated with erectile dysfunction in diabetes. The relation between cytomegalovirus and erectile dysfunction is markedly present in patients with elevated high-sensitivity C-reactive protein and fibrinogen levels, suggesting a modifying effect by the inflammation.
Collapse
Affiliation(s)
- M C A Blans
- Department of Medical Microbiology and Infectious Diseases, Gelre Hospitals, 7300 DS Apeldoorn, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
44
|
Stassen FR, Vega-Córdova X, Vliegen I, Bruggeman CA. Immune activation following cytomegalovirus infection: More important than direct viral effects in cardiovascular disease? J Clin Virol 2006; 35:349-53. [PMID: 16387544 DOI: 10.1016/j.jcv.2005.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 11/15/2005] [Accepted: 11/17/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Over the last 30 years multiple micro-organisms have been associated with different types of vascular disease, like atherosclerosis, restenosis or transplant arteriosclerosis. Nonetheless, it is still ambiguous which molecular mechanisms are exactly involved in the exacerbating effect of microbes in these disorders. OBJECTIVES AND STUDY DESIGN The present review summarizes sero-epidemiological, in vitro and animal data supporting the role of cytomegalovirus, a member of the herpes virus family, in vascular disease. Additionally, various ways by which the virus can potentially affect the disease will be discussed. RESULTS Rodent models as well as in vitro studies suggested that CMV might enhance lesion formation in various ways, like augmentation of the oxLDL uptake, altering monocyte adhesion or increasing the production of pro-inflammatory cytokines. Nevertheless, recent data from our lab and others suggest that alternative mechanisms also may contribute to CMV induced. Inspired by this, we will hypothesise alternative mechanisms by which CMV might affect atherosclerosis. CONCLUSIONS Although researchers have tried for many years to unravel the tactics by which micro-organisms, like CMV, aggravate atherosclerosis, so far most suggested mechanisms failed to fully explain both experimental and clinical observations. Therefore, new means to elucidate the observed effects are required.
Collapse
Affiliation(s)
- Frank R Stassen
- Department of Medical Microbiology and Maastricht Infection Center, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 5800, 6202AZ Maastricht, The Netherlands.
| | | | | | | |
Collapse
|
45
|
Söderberg-Nauclér C. Does cytomegalovirus play a causative role in the development of various inflammatory diseases and cancer? J Intern Med 2006; 259:219-46. [PMID: 16476101 DOI: 10.1111/j.1365-2796.2006.01618.x] [Citation(s) in RCA: 239] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human cytomegalovirus (HCMV) is a herpes virus that infects and is carried by 70-100% of the world's population. During its evolution, this virus has developed mechanisms that allow it to survive in an immunocompetent host. For many years, HCMV was not considered to be a major human pathogen, as it appeared to cause only rare cases of HCMV inclusion disease in neonates. However, HCMV is poorly adapted for survival in the immunosuppressed host and has emerged as an important human pathogen in AIDS patients and in patients undergoing immunosuppressive therapy following organ or bone marrow transplantation. HCMV-mediated disease in such patients has highlighted the possible role of this virus in the development of other diseases, in particular inflammatory diseases such as vascular diseases, autoimmune diseases and, more recently, with certain forms of cancers. Current research is focused on determining whether HCMV plays a causative role in these diseases or is merely an epiphenomenon of inflammation. Inflammation plays a central role in the pathogenesis of HCMV. This virus has developed a number of mechanisms that enable it to hide from the cells of the immune system and, at the same time, reactivation of a latent infection requires immune activation. Numerous products of the HCMV genome are devoted to control central functions of the innate and adaptive immune responses. By influencing the regulation of various cellular processes including the cell cycle, apoptosis and migration as well as tumour invasiveness and angiogenesis, HCMV may participate in disease development. Thus, the various drugs now available for treatment of HCMV disease (e.g. ganciclovir, acyclovir and foscarnet), may also prove to be useful in the treatment of other, more widespread diseases.
Collapse
Affiliation(s)
- C Söderberg-Nauclér
- Department of Medicine, Center for Molecular Medicine, L8:03, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
46
|
Singh M, Williams BA, Gersh BJ, McClelland RL, Ho KKL, Willerson JT, Penny WF, Cutlip DE, Holmes DR. Geographical Differences in the Rates of Angiographic Restenosis and Ischemia-Driven Target Vessel Revascularization After Percutaneous Coronary Interventions. J Am Coll Cardiol 2006; 47:34-9. [PMID: 16386661 DOI: 10.1016/j.jacc.2005.07.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 05/27/2005] [Accepted: 07/11/2005] [Indexed: 11/25/2022]
Abstract
UNLABELLED Many cardiologists consider it reasonable to assume in clinical practice that percutaneous coronary intervention using drug-eluting stents ought to be considered equivalent, if not superior, to bypass surgery. In the absence of a definitive clinical trial to support this view, how should the prudent, cutting-edge cardiologist proceed? OBJECTIVES This study assessed the geographical differences in target vessel revascularization (TVR) after percutaneous coronary intervention (PCI) in the Prevention of Restenosis With Tranilast and its Outcomes (PRESTO) trial. BACKGROUND An aggressive approach to PCI is more common in the U.S. than in other countries. The impact of this approach on restenosis outcomes has not been studied. METHODS Using the PRESTO trial, we compared nine-month ischemic TVR after PCI in U.S.-treated patients (n = 5,026) with rates in other countries (n = 6,458). We defined TVR as repeat intervention for chest pain/positive stress test. Additionally, angiographic restenosis (> or =50% narrowing or > or =50% loss of gain at nine-month follow-up) was compared between U.S. and non-U.S. patients within the prespecified angiographic subset (n = 2,823). Regression models were developed to adjust for clinical and lesion-related characteristics. RESULTS Higher rates of TVR (18% vs. 11%), and angiographic restenosis (65% vs. 48%) were observed in patients treated in the U.S. as compared with the other patients (p < 0.01 for both comparisons). Patients treated in the U.S. were more likely to be female, diabetic, not currently smoking, to have unstable angina, and to have a prior PCI. In U.S. patients, lesions tended to be longer, but less likely to be American College of Cardiology/American Heart Association class C. After adjusting for clinical and angiographic variables, PCI in the U.S. was still associated with increased angiographic restenosis and ischemic TVR. CONCLUSIONS Angiographic restenosis and ischemia-driven TVR rates were higher in patients treated in the U.S. The difference could only partially be explained by the higher prevalence of measured adverse clinical and angiographic features.
Collapse
Affiliation(s)
- Mandeep Singh
- Division of Internal Medicine and Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
DeMeritt IB, Podduturi JP, Tilley AM, Nogalski MT, Yurochko AD. Prolonged activation of NF-kappaB by human cytomegalovirus promotes efficient viral replication and late gene expression. Virology 2005; 346:15-31. [PMID: 16303162 PMCID: PMC2600890 DOI: 10.1016/j.virol.2005.09.065] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Revised: 08/12/2005] [Accepted: 09/23/2005] [Indexed: 01/04/2023]
Abstract
Infection of fibroblasts by human cytomegalovirus (HCMV) rapidly activates the NF-kappaB signaling pathway, which we documented promotes efficient transactivation of the major immediate-early promoter (DeMeritt, I.B., Milford, L.E., Yurochko, A.D. (2004). Activation of the NF-kappaB pathway in human cytomegalovirus-infected cells is necessary for efficient transactivation of the major immediate-early promoter. J. Virol. 78, 4498-4507). Because a second, sustained increase in NF-kappaB activity following the initial phase of NF-kappaB activation was also observed, we investigated the role that this prolonged NF-kappaB activation played in viral replication and late gene expression. We first investigated HCMV replication in cells in which NF-kappaB activation was blocked by pretreatment with NF-kappaB inhibitors: HCMV replication was significantly decreased in these cultures. A decrease in replication was also observed when NF-kappaB was inhibited up to 48 h post-infection, suggesting a previously unidentified role for NF-kappaB in the regulation of the later class of viral genes.
Collapse
Affiliation(s)
- Ian B. DeMeritt
- Department of Microbiology and Immunology and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
| | - Jagat P. Podduturi
- Department of Microbiology and Immunology and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
| | - A. Michael Tilley
- Department of Microbiology and Immunology and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
| | - Maciej T. Nogalski
- Department of Microbiology and Immunology and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
| | - Andrew D. Yurochko
- Department of Microbiology and Immunology and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932
- *Corresponding Author: Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, Phone: (318) 675-8332, Fax: (318) 675-5764, E-Mail:
| |
Collapse
|
48
|
Gómez E, Laurés A, Baltar JM, Melón S, Díez B, de Oña M. Cytomegalovirus Replication and “Herpesvirus Burden” as Risk Factor of Cardiovascular Events in the First Year After Renal Transplantation. Transplant Proc 2005; 37:3760-3. [PMID: 16386530 DOI: 10.1016/j.transproceed.2005.08.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cytomegalovirus (CMV) infection alone or in combination with other pathogens ("pathogen burden") has been postulated as a factor producing arteriosclerosis in some solid organ transplant recipients. The aim of this study was to assess whether the patients with CMV replication and/or "herpesvirus burden" experienced a greater incidence of cardiovascular events during the first year after kidney transplantation. One hundred twenty-one consecutive transplant recipients were prospectively studied for CMV replication using antigenemia and polymerase chain reaction (PCR) weekly during the 4 first months, and monthly thereafter for 1 year. Simultaneously, nested-PCR for human herpes virus (HHV)-6 and HHV-7 were performed to yield a herpesvirus burden (as determined by seropositivity), including CMV, herpes simplex virus (HSV), varicella-zoster virus (VZV), and Epstein-Barr virus (EBV). The following additional parameters were analyzed: gender, age, smoking, duration of dialysis, preexistent diabetes, and preexistent cardiovascular events. After 1 year posttransplantation cardiovascular events, body mass index, arterial hypertension, number of antihypertensive drugs, use of ACE and/or ARBs inhibitors, diabetes, anemia, homocysteine, creatinine, cholesterol, HDLc, LDLc, PTH-i, proteinuria, and immunosuppression with cyclosporine or tacrolimus. CMV replication was present in 79 (65.3%) patients. Among 121 renal transplant recipients, 13 presented cardiovascular events, all associated with CMV replication (P = .004). Neither HHV-6 or HHV-7 replication influenced this complication. All patients with these events were seropositive for CMV, HSV, VZV, and EBV, as opposed to 64.8% without them (P = .009). Other factors that showed differences between patients with versus without events were as follows: preexistent events (76.9% vs 14.8%; P = .000), age (60 +/- 10 vs 49 +/- 14; P = .002), serum triglyceride value (191 +/- 82 vs 135 +/- 72; P = .02), and anemia (23.1% vs 5.6%; P = .05). Multiple logistic regression analysis for statistically significant variables only showed that preexistent events influenced the development of posttransplantation events (odds ratio, 27; 95% confidence interval, 4.7-154; P = .0005). In conclusion, cardiovascular events within 1 year after transplantation were more frequent among patients with CMV replication and seropositivity for other herpesviruses. An important risk factor was the presence of preexistent events.
Collapse
Affiliation(s)
- E Gómez
- Nephrology Service, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | | | | | | |
Collapse
|
49
|
Sagedal S, Hartmann A, Rollag H. The impact of early cytomegalovirus infection and disease in renal transplant recipients. Clin Microbiol Infect 2005; 11:518-30. [PMID: 15966969 DOI: 10.1111/j.1469-0691.2005.01190.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human cytomegalovirus (HCMV) infection is the single most frequent infectious complication in the early period after kidney transplantation. The HCMV load in blood, measured by HCMV PCR or the HCMV pp65 antigen test, is a predictor of HCMV disease in seropositive recipients. However, plasma virus load measurements are of only modest value in predicting the risk of HCMV disease in seronegative recipients of kidneys from seropositive donors. HCMV infection is an independent risk-factor for acute kidney graft rejection. There is also evidence that HCMV is associated with an increased long-term mortality and post-transplant diabetes mellitus. Whether pre-emptive or prophylactic therapy should be the preferred strategy is not yet decided. Some studies indicate that HCMV prophylaxis may reduce the risk of acute rejection, and thereby increase long-term graft survival in seronegative recipients of kidneys from seropositive donors.
Collapse
Affiliation(s)
- S Sagedal
- Department of Internal Medicine, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | |
Collapse
|
50
|
Blum A, Hadas V, Burke M, Yust I, Kessler A. Viral load of the human immunodeficiency virus could be an independent risk factor for endothelial dysfunction. Clin Cardiol 2005; 28:149-53. [PMID: 15813624 PMCID: PMC6653956 DOI: 10.1002/clc.4960280311] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Recent reports of myocardial infarction in young persons infected with human immunodeficiency virus (HIV) who are receiving protease inhibitor therapy have raised concerns about premature coronary artery disease in this population. However, endothelial dysfunction, hypercoagulability, hypertriglyceridemia, and abnormal coronary artery pathology have been observed in association with HIV infection prior to the availability of protease inhibitor therapy. HYPOTHESIS The study was undertaken to determine the association between endothelial function, viral load, CD4+ count, and other well-established risk factors for atherosclerosis. METHODS This prospective, case-controlled study compared viral (HIV) load and the CD4+ T-lymphocyte count and endothelial function in 24 HIV-positive carriers. Brachial artery diameter, HIV viral load, and CD4 count were measured. RESULTS We found that viral load correlated inversely with endothelial function; the higher the viral load, the worse the endothelial dysfunction (p < 0.005). CONCLUSION High viral load appears to be associated with endothelial dysfunction in patients with HIV. This preliminary observation supports the infectious theory that viruses may play an important role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Arnon Blum
- Department of Internal Medicine A, Poria Medical Center, Lower Galilee, Israel.
| | | | | | | | | |
Collapse
|