1
|
Blanco R, Muñoz JP. Porphyromonas gingivalis and Human Cytomegalovirus Co-Infection: A Potential Link Between Periodontal Disease and Oral Cancer Development. Cancers (Basel) 2025; 17:1525. [PMID: 40361452 PMCID: PMC12071019 DOI: 10.3390/cancers17091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Periodontal disease (PD) is an inflammatory condition that can contribute to the development of oral cancer. Chronic inflammation from PD can lead to the release of inflammatory mediators and growth factors that promote tumorigenesis. Porphyromonas gingivalis (P. gingivalis) is one of several pathogens implicated in PD and its potential link to oral cancer. However, other viral infections, such as human cytomegalovirus (HCMV), can also contribute to chronic inflammation, creating a favorable environment for oral cancer development. OBJECTIVES The present literature review tries to investigate the possible influence of P. gingivalis and HCMV co-infection in fostering the development of oral cancer and chronic periodontitis. METHODS A comprehensive search was conducted in PubMed and Google Scholar, focusing on the relevance and significance of articles that examine the role of P. gingivalis and HCMV in periodontal disease and oral cancer. RESULTS The evidence suggests that P. gingivalis and HCMV may act synergistically to modulate host immunity, disrupt epithelial integrity, and interfere with key cellular pathways. These interactions may enhance tissue destruction and foster a microenvironment conducive to malignant transformation. However, most of these findings stem from in vitro models and small-scale clinical studies, limiting the generalizability and clinical relevance of current conclusions. CONCLUSIONS Although the proposed interaction between P. gingivalis and HCMV provides a compelling framework for understanding how microbial co-infections may influence oral cancer, the evidence remains preliminary and largely associative. To support these mechanistic hypotheses, future studies should give top priority to in vivo models, bigger patient cohorts, and longitudinal clinical studies.
Collapse
Affiliation(s)
- Rancés Blanco
- Independent Researcher, Av. Vicuña Mackenna Poniente 6315, La Florida 8240000, Chile
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| |
Collapse
|
2
|
Rosseels W, Godinas L, Jallah P, Vos R, Dupont L, Kuypers D, Vanhoutte T, Claes KJ. Thrombotic Microangiopathy: A Devastating Complication After Lung Transplantation. Transplant Direct 2025; 11:e1758. [PMID: 40078820 PMCID: PMC11896099 DOI: 10.1097/txd.0000000000001758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 03/14/2025] Open
Abstract
Background Thrombotic microangiopathy (TMA) following lung transplantation (LTx) is a rare but severe complication. The pathogenesis is poorly understood, and various risk factors have been suggested. In this study, we aimed to evaluate diagnostic accuracy, identify risk factors, and assess renal, pulmonary, and overall survival of TMA in this patient group. Methods We performed a case-control retrospective study of patients with TMA after LTX between January 1, 2000, and January 1, 2021. Controls were selected based on underlying lung disease, age, sex, cytomegalovirus risk, and immunosuppressive regimen. Overall survival data were collected for the whole lung transplant group. Results A total of 29 TMA cases (2.9%) were identified out of 1025 LTx. Median time to development of TMA was 5.9 mo, 76% occurred in the first 12 mo. In the TMA group a higher rate of HLA donor-specific antibodies (11% versus 1%; P = 0.05), a lower median time to onset of chronic lung allograft dysfunction (37 versus 91 mo; P = 0.0017), a higher rate of cytomegalovirus infection (45% versus 19%; P = 0.02), and a higher prevalence of end-stage renal disease (24% versus 6%; P = 0.03) and overall death (97% versus 44%; P < 0.0001) was found. Diagnostic assessment of TMA was complete in 48% of patients, with Coombs testing missing in 52% and a disintegrin and metalloproteinase with thrombospondin type 1 motif 13 activity not assessed in 59%. Conclusions TMA poses a significant risk of end-stage renal disease and mortality after LTx. Challenges remain in standardizing diagnostic criteria and understanding its pathogenesis, underscoring the need for unified protocols in diagnosis and standardized screening. This study identifies potential risk factors and temporal patterns for TMA occurrence, providing crucial insights for future treatment strategies.
Collapse
Affiliation(s)
- Wouter Rosseels
- Department of Nephrology and Renal Transplantation, UZ Leuven, Leuven, Belgium
| | - Laurent Godinas
- Department of Respiratory Diseases, UZ Leuven, Leuven, Belgium
| | - Papay Jallah
- Department of Nephrology and Renal Transplantation, UZ Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Respiratory Diseases, UZ Leuven, Leuven, Belgium
| | - Lieven Dupont
- Department of Respiratory Diseases, UZ Leuven, Leuven, Belgium
| | - Dirk Kuypers
- Department of Nephrology and Renal Transplantation, UZ Leuven, Leuven, Belgium
- Department of Microbiology, Immunology, and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Thomas Vanhoutte
- Department of Nephrology and Renal Transplantation, UZ Leuven, Leuven, Belgium
- Department of Microbiology, Immunology, and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Kathleen J. Claes
- Department of Nephrology and Renal Transplantation, UZ Leuven, Leuven, Belgium
- Department of Microbiology, Immunology, and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Chee YJ, Dalan R, Cheung C. The Interplay Between Immunity, Inflammation and Endothelial Dysfunction. Int J Mol Sci 2025; 26:1708. [PMID: 40004172 PMCID: PMC11855323 DOI: 10.3390/ijms26041708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The endothelium is pivotal in multiple physiological processes, such as maintaining vascular homeostasis, metabolism, platelet function, and oxidative stress. Emerging evidence in the past decade highlighted the immunomodulatory function of endothelium, serving as a link between innate, adaptive immunity and inflammation. This review examines the regulation of the immune-inflammatory axis by the endothelium, discusses physiological immune functions, and explores pathophysiological processes leading to endothelial dysfunction in various metabolic disturbances, including hyperglycemia, obesity, hypertension, and dyslipidaemia. The final section focuses on the novel, repurposed, and emerging therapeutic targets that address the immune-inflammatory axis in endothelial dysfunction.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| |
Collapse
|
4
|
Xu X, Silveira A, Lundman P, Rahbar A, Söderberg-Nauclér C. Enhanced levels of IL-6 and PAI-1 and decreased levels of MMP-3 in cytomegalovirus seropositive patients with prior myocardial infarction. IJC HEART & VASCULATURE 2025; 56:101570. [PMID: 39691830 PMCID: PMC11650320 DOI: 10.1016/j.ijcha.2024.101570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Background Efforts to understand atherosclerosis, a major cause of ischemic heart disease, have linked several lifestyle factors to increased risk for developing cardiovascular disease. Some studies suggest that cytomegalovirus (CMV), a widely prevalent herpesvirus, is reactivated in atherosclerotic plaques and associated with higher cardiovascular mortality risk. We aimed to explore whether CMV seropositivity and CMV-IgG antibody levels correlate with relevant biomarkers in a cohort of patients with myocardial infarction (MI) and matched controls. Methods and results We analyzed a dataset from 324 survivors of MI treated in Stockholm between 1996 and 2001. Blood samples collected three months after MI were used to measure protective Apo B100 autoantibodies, metabolic, and inflammatory biomarkers. CMV serology was performed on stored serum samples. Correlation analyses were conducted between biomarkers and CMV serostatus in 324 patients and age- and sex-matched controls. While CMV seroprevalence was equal, the CMV-IgG levels were higher in controls. Among various factors examined, CMV seropositive MI patients had elevated levels of plasminogen activator inhibitor-1 (PAI-1) and interleukin-6, along with lower levels of MMP-3, than CMV seronegative MI patients. CMV-IgG levels correlated positively with PAI-1 levels in patients. Although CMV seropositivity was associated with increased proinsulin levels, there was no correlation with diabetes diagnosis. Conclusions Our findings suggest an enhanced inflammatory and prothrombotic state in CMV seropositive patients after MI. Notably, patients had lower levels of CMV IgG than controls.
Collapse
Affiliation(s)
- Xinling Xu
- Department of Medicine Solna, Unit Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Angela Silveira
- Department of Medicine Solna, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pia Lundman
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Afsar Rahbar
- Department of Medicine Solna, Unit Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Söderberg-Nauclér
- Department of Medicine Solna, Unit Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Biosciences, InFLAMES Research Flagship Center, MediCity, University of Turku, Finland
| |
Collapse
|
5
|
Fernández S, Castro P, Azoulay E. What intensivists need to know about cytomegalovirus infection in immunocompromised ICU patients. Intensive Care Med 2025; 51:39-61. [PMID: 39774866 DOI: 10.1007/s00134-024-07737-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE Advances in therapeutic care are leading to an increase in the number of patients living with overt immunosuppression. These patients are at risk of cytomegalovirus (CMV) infection and disease that can lead to or develop during ICU admission. This manuscript aims to describe the clinical presentation, risk factors, and management of CMV infection and disease in this patient population. METHODS We conducted a literature search in PubMed up to April 2024, focusing on CMV infection and disease in patients with overt immunosuppression (hematopoietic stem cell and solid organ transplantation, solid or hematologic malignancies, HIV infection, immunosuppressive drugs, including corticosteroids, and primary immunodeficiencies) admitted to the intensive care unit (ICU). As there is limited ICU-specific data on CMV in immunosuppressed patients, many of the findings were extrapolated from the general literature. RESULTS CMV infection and disease in immunocompromised critically ill patients is associated with increased mortality and presents significant management challenges. Clinical manifestations are diverse, shaped by the underlying immune deficiency and primary disease. Pneumonia and encephalitis are among the most severe CMV end-organ diseases. CMV infection may also increase the risk of secondary infections and induce life-threatening conditions, such as thrombotic microangiopathy. Importantly, CMV reactivation is not synonymous with CMV disease, and qPCR testing of body fluids cannot reliably differentiate between viral shedding and tissue-invasive infection, which requires histopathological confirmation. Ganciclovir is commonly the first-line anti-viral, though maribavir shows potential for patients unresponsive to other antivirals. Identifying patients who require prophylactic or preemptive antiviral therapy is essential. CONCLUSIONS CMV infection and disease in critically ill immunocompromised patients pose a unique challenge for intensivists. The broad spectrum of clinical presentations and the difficulty in distinguishing CMV-related symptoms from other causes require a high level of clinical suspicion. Accurate interpretation of nucleic acid load levels and careful evaluation of CMV's pathogenic role when it is found are critical. Further studies focusing specifically on CMV infection and disease in critically ill immunocompromised patients are needed to optimize management strategies.
Collapse
Affiliation(s)
- Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain.
- Medical Intensive Care Unit, AP-HP, Saint-Louis University Hospital, Paris, France.
| | - Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Elie Azoulay
- Medical Intensive Care Unit, AP-HP, Saint-Louis University Hospital, Paris, France
| |
Collapse
|
6
|
Fattal I, Steinmetz T, Donin N, Tobar AF, Rozen-Zvi B, Rahamimov R, Nesher E, Shirazi I, Mor E, Babai I, Fishelson Z. Comprehensive Analysis of Thrombotic Microangiopathy Following Renal Transplantation. Int J Nephrol 2024; 2024:4396051. [PMID: 39742342 PMCID: PMC11688143 DOI: 10.1155/ijne/4396051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/21/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Thrombotic microangiopathy is a severe complication of renal transplantation. Little is known about risk factors, incidence of autoantibodies against complement components, and prognosis. Methods: Clinical and laboratory data were retrospectively collected for 13 patients diagnosed with post-transplant thrombotic microangiopathy (PT-TMA) in 2011-2018. Enzyme-linked immunosorbent assay (ELISA) results were compared to transplant recipients without PT-TMA and healthy controls. Results: Nine patients (69%) had potential PT-TMA risk factors other than exposure to calcineurin inhibitors (CNIs). Stratification by time to PT-TMA yielded two groups. Patients diagnosed within 6 months of transplantation (n = 6) were characterized by positive donor-specific antibody (DSA) test, complement-associated renal disease, and acute rejection. Two had IgG and IgA autoantibodies to complement Factors H and I, respectively. Patients diagnosed ≥ 3 years after transplantation (n = 7) had a high rate of infection. Renal biopsy yielded dense deposits in 6 patients, and only one with primary immune complex renal disease. Within 2 years, graft failure requiring dialysis occurred in 6 patients (46%). Three patients with early-onset PT-TMA showed improved renal function and remained stable under eculizumab treatment. Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disorder (EPTLD) developed in 3 patients, 2 of whom had received eculizumab for more than 5 years. Five patients (39%) died during follow-up. Conclusion: In this study, PT-TMA was associated with other risk factors besides CNI exposure, with differences by time of onset from transplantation. Prognosis was generally poor but better for early-onset PT-TMA managed with eculizumab. The development of late EPTLD in 3 patients raises concerns.
Collapse
Affiliation(s)
- Ittai Fattal
- Nephrology and Hypertension Unit, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Tali Steinmetz
- Nephrology and Hypertension Unit, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Natalie Donin
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ana Foigelman Tobar
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Pathology, Rabin Medical Center—Beilinson Hospital, Petach Tikva, Israel
| | - Benaya Rozen-Zvi
- Nephrology and Hypertension Unit, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ruth Rahamimov
- Nephrology and Hypertension Unit, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Transplantation, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Eviatar Nesher
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Transplantation, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Idit Shirazi
- Clinical Immunology Laboratory, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Eytan Mor
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Transplantation Unit, Department of Surgery B, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel
| | - Ilan Babai
- Clinical Immunology Laboratory, Rabin Medical Center—Beilinson Hospital, Petach Tikva 4941492, Israel
| | - Zvi Fishelson
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
7
|
George Pryzdial EL, Perrier JR, Rashid MU, West HE, Sutherland MR. Viral coagulation: pushing the envelope. J Thromb Haemost 2024; 22:3366-3382. [PMID: 39260743 DOI: 10.1016/j.jtha.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Many virus types affect the blood clotting system with correlations to pathology that range widely from thrombosis to hemorrhage linking to inflammation. Here we overview the intricate crosstalk induced by infection between proteins on the virus encoded by either the host or virus genomes, coagulation proteins, platelets, leukocytes, and endothelial cells. For blood-borne viruses with an outer covering acquired from the host cell, the envelope, a key player may be the cell-derived trigger of coagulation on the virus surface, tissue factor (TF). TF is a multifunctional transmembrane cofactor that accelerates factor (F)VIIa-dependent activation of FX to FXa, leading to clot formation. However, the nascent TF/FVIIa/FXa complex also facilitates G protein-coupled modulation of cells via protease-activated receptor 2. As a viral envelope constituent, TF can bypass the physiological modes of regulation, thereby initiating the activation of neighboring platelets, leukocytes, and endothelial cells. A thromboinflammatory environment is predicted due to feedback amplification in response to cellular release of cytokines, procoagulant proteins, neutrophil extracellular traps, and stimulus-induced accessibility of adhesive receptors, resulting in cellular aggregates. The pathobiological effects of thromboinflammation ultimately contribute to innate and adaptive immunity for viral clearance. In contrast, the preceding stages of viral infection may be enhanced via the TF-protease axis.
Collapse
Affiliation(s)
- Edward Louis George Pryzdial
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.
| | - John Ruggles Perrier
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mahamud-Ur Rashid
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Henry Euan West
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Michael Ross Sutherland
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Liu X, Shen J, Yan H, Hu J, Liao G, Liu D, Zhou S, Zhang J, Liao J, Guo Z, Li Y, Yang S, Li S, Chen H, Guo Y, Li M, Fan L, Li L, Luo P, Zhao M, Liu Y. Posttransplant complications: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2024; 5:e669. [PMID: 39224537 PMCID: PMC11366828 DOI: 10.1002/mco2.669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Posttransplantation complications pose a major challenge to the long-term survival and quality of life of organ transplant recipients. These complications encompass immune-mediated complications, infectious complications, metabolic complications, and malignancies, with each type influenced by various risk factors and pathological mechanisms. The molecular mechanisms underlying posttransplantation complications involve a complex interplay of immunological, metabolic, and oncogenic processes, including innate and adaptive immune activation, immunosuppressant side effects, and viral reactivation. Here, we provide a comprehensive overview of the clinical features, risk factors, and molecular mechanisms of major posttransplantation complications. We systematically summarize the current understanding of the immunological basis of allograft rejection and graft-versus-host disease, the metabolic dysregulation associated with immunosuppressive agents, and the role of oncogenic viruses in posttransplantation malignancies. Furthermore, we discuss potential prevention and intervention strategies based on these mechanistic insights, highlighting the importance of optimizing immunosuppressive regimens, enhancing infection prophylaxis, and implementing targeted therapies. We also emphasize the need for future research to develop individualized complication control strategies under the guidance of precision medicine, ultimately improving the prognosis and quality of life of transplant recipients.
Collapse
Affiliation(s)
- Xiaoyou Liu
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Junyi Shen
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hongyan Yan
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Jianmin Hu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guorong Liao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ding Liu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Song Zhou
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jie Zhang
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Jun Liao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zefeng Guo
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuzhu Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Siqiang Yang
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shichao Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hua Chen
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ying Guo
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Min Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Lipei Fan
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Liuyang Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Peng Luo
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ming Zhao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yongguang Liu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
9
|
Imanifard Z, Liguori L, Remuzzi G. TMA in Kidney Transplantation. Transplantation 2023; 107:2329-2340. [PMID: 36944606 DOI: 10.1097/tp.0000000000004585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Thrombotic microangiopathy (TMA) is a rare and devastating complication of kidney transplantation, which often leads to graft failure. Posttransplant TMA (PT-TMA) may occur either de novo or as a recurrence of the disease. De novo TMA can be triggered by immunosuppressant drugs, antibody-mediated rejection, viral infections, and ischemia/reperfusion injury in patients with no evidence of the disease before transplantation. Recurrent TMA may occur in the kidney grafts of patients with a history of atypical hemolytic uremic syndrome (aHUS) in the native kidneys. Studies have shown that some patients with aHUS carry genetic abnormalities that affect genes that code for complement regulators (CFH, MCP, CFI) and components (C3 and CFB), whereas in 10% of patients (mostly children), anti-FH autoantibodies have been reported. The incidence of aHUS recurrence is determined by the underlying genetic or acquired complement abnormality. Although treatment of the causative agents is usually the first line of treatment for de novo PT-TMA, this approach might be insufficient. Plasma exchange typically resolves hematologic abnormalities but does not improve kidney function. Targeted complement inhibition is an effective treatment for recurrent TMA and may be effective in de novo PT-TMA as well, but it is necessary to establish which patients can benefit from different therapeutic options and when and how these can be applied.
Collapse
Affiliation(s)
- Zahra Imanifard
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Clinical Research Center for Rare Diseases Aldo e Cele Daccò, Ranica, Italy
| | | | | |
Collapse
|
10
|
Bomfim GF, Priviero F, Poole E, Tostes RC, Sinclair JH, Stamou D, Uline MJ, Wills MR, Webb RC. Cytomegalovirus and Cardiovascular Disease: A Hypothetical Role for Viral G-Protein-Coupled Receptors in Hypertension. Am J Hypertens 2023; 36:471-480. [PMID: 37148218 PMCID: PMC10403975 DOI: 10.1093/ajh/hpad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023] Open
Abstract
Cytomegalovirus (CMV) is a member of the β-herpesviruses and is ubiquitous, infecting 50%-99% of the human population depending on ethnic and socioeconomic conditions. CMV establishes lifelong, latent infections in their host. Spontaneous reactivation of CMV is usually asymptomatic, but reactivation events in immunocompromised or immunosuppressed individuals can lead to severe morbidity and mortality. Moreover, herpesvirus infections have been associated with several cardiovascular and post-transplant diseases (stroke, atherosclerosis, post-transplant vasculopathy, and hypertension). Herpesviruses, including CMV, encode viral G-protein-coupled receptors (vGPCRs) that alter the host cell by hijacking signaling pathways that play important roles in the viral life cycle and these cardiovascular diseases. In this brief review, we discuss the pharmacology and signaling properties of these vGPCRs, and their contribution to hypertension. Overall, these vGPCRs can be considered attractive targets moving forward in the development of novel hypertensive therapies.
Collapse
Affiliation(s)
- Gisele F Bomfim
- Institute of Health Sciences, Federal University of Mato Grosso, campus Sinop (UFMT), Sinop, MT, Brazil
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, USA
| | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, USA
- Biomedical Engineering Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| | - Emma Poole
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Rita C Tostes
- Ribeirao Preto Medical School, University of Sao Paulo (FMRP-USP), Ribeirao Preto, SP, Brazil
| | - John H Sinclair
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Mark J Uline
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, USA
- Biomedical Engineering Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina, USA
| | - Mark R Wills
- Department of Pathology, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - R Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, USA
- Biomedical Engineering Program, University of South Carolina, Columbia, South Carolina, USA
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
11
|
Abstract
Sepsis is accompanied by thrombocytopenia and the severity of the thrombocytopenia is associated with mortality. This thrombocytopenia is characteristic of disseminated intravascular coagulation (DIC), the sepsis-associated coagulopathy. Many of the pathogens, both bacterial and viral, that cause sepsis also directly activate platelets, which suggests that pathogen-induced platelet activation leads to systemic thrombosis and drives the multi-organ failure of DIC. In this paper we review the mechanisms of platelet activation by pathogens and the evidence for a role for anti-platelet agents in the management of sepsis.
Collapse
Affiliation(s)
- Dermot Cox
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
12
|
Zerangian N, Erabi G, Poudineh M, Monajjem K, Diyanati M, Khanlari M, Khalaji A, Allafi D, Faridzadeh A, Amali A, Alizadeh N, Salimi Y, Ghane Ezabadi S, Abdi A, Hasanabadi Z, ShojaeiBaghini M, Deravi N. Venous thromboembolism in viral diseases: A comprehensive literature review. Health Sci Rep 2023; 6:e1085. [PMID: 36778773 PMCID: PMC9900357 DOI: 10.1002/hsr2.1085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/25/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Venous thromboembolism (VTE) is known to be a common respiratory and/or cardiovascular complication in hospitalized patients with viral infections. Numerous studies have proven human immunodeficiency virus infection to be a prothrombotic condition. An elevated VTE risk has been observed in critically ill H1N1 influenza patients. VTE risk is remarkably higher in patients infected with the Hepatitis C virus in contrast to uninfected subjects. The elevation of D-dimer levels supported the association between Chikungunya and the Zika virus and the rise of clinical VTE risk. Varicella-zoster virus is a risk factor for both cellulitis and the consequent invasive bacterial disease which may take part in thrombotic initiation. Eventually, hospitalized patients infected with the coronavirus disease of 2019 (COVID-19), the cause of the ongoing worldwide pandemic, could mainly suffer from an anomalous risk of coagulation activation with enhanced venous thrombosis events and poor quality clinical course. Although the risk of VTE in nonhospitalized COVID-19 patients is not known yet, there are a large number of guidelines and studies on thromboprophylaxis administration for COVID-19 cases. This study aims to take a detailed look at the effect of viral diseases on VTE, the epidemiology of VTE in viral diseases, and the diagnosis and treatment of VTE.
Collapse
Affiliation(s)
- Nasibeh Zerangian
- Health Education and Health Promotion, Department of Health Education and Health Promotion, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Gisou Erabi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | | | - Kosar Monajjem
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Maryam Diyanati
- Student Research CommitteeRafsanjan University of Medical SciencesRafsanjanIran
| | - Maryam Khanlari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | - Diba Allafi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Arian Amali
- Student Research Committee, Paramedical DepartmentIslamic Azad University, Mashhad BranchMashhadIran
| | - Nilufar Alizadeh
- Doctor of Medicine (MD), School of MedicineIran University of Medical SciencesTehranIran
| | - Yasaman Salimi
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Sajjad Ghane Ezabadi
- Student's Scientific Research Center, School of MedicineTehran University of Medical SciencesTehranIran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Zahra Hasanabadi
- Doctor of Medicine (MD), School of MedicineQazvin University of Medical ScienceQazvinIran
| | - Mahdie ShojaeiBaghini
- Medical Informatics Research Center, Institute for Futures Studies in HealthKerman University of Medical SciencesKermanIran
| | - Niloofar Deravi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
13
|
Guo M, Qi J, Hou Q, Li X, Han Y. Risk factors for transplant-associated thrombotic microangiopathy (TA-TMA): a systematic review and meta-analysis. Expert Rev Hematol 2023; 16:191-203. [PMID: 36588482 DOI: 10.1080/17474086.2023.2162501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Transplant-associated thrombotic microangiopathy (TA-TMA) is a severe hematopoietic stem cell transplantation complication with high mortality and a poor patient prognosis. The pathogenesis of TA-TMA is not yet clear. In previous studies, the conclusions of different centers remain controversial. We conducted a systematic review and meta-analysis of nine selected risk factors that might be associated with the onset of TA-TMA. MATERIALS AND METHODS PubMed databases were searched from their inception up to 15 September 2021, for relevant studies. The articles included unprocessed data related to one or more of the risk factors discussed in this meta-analysis, including recipient gender, donor type, graft source, pretreatment, infection, aGVHD, diagnosis, total body irradiation (TBI), and CMV infection. The outcome is the incidence rate (IR) of TA-TMA. RESULTS AND CONCLUSIONS According to the sixteen articles included, risk factors included in this Meta-analysis included gender, unrelated donor source (95% CI: 1.29-2.01), graft source from peripheral blood stem cell (PBSC)(95% CI: 0.48-0.97), RIC/NMA, class II-IV aGVHD (95% CI: 2.22-4.78), nonmalignant disease, TBI. However, inconsistent diagnostic criteria for TA-TMA and the limited number of studies have an impact on the results of the study. More prospective cohort studies and More accurate diagnostic criteria are needed.
Collapse
Affiliation(s)
- Mengting Guo
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jiaqian Qi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Qixiu Hou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xueqian Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Wu Q, Tian X, Gong N, Zheng J, Liang D, Li X, Lu X, Xue W, Tian P, Wen J. Early graft loss due to acute thrombotic microangiopathy accompanied by complement gene variants in living-related kidney transplantation: case series report. BMC Nephrol 2022; 23:249. [PMID: 35836191 PMCID: PMC9284761 DOI: 10.1186/s12882-022-02868-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background Recently, early graft loss has become very rare in living-related kidney transplantation (LKT) as a result of decreased risk of hyperacute rejection and improvements in immunosuppressive regimens. Post-transplant acute thrombotic microangiopathy (TMA) is a rare, multi-factorial disease that often occurs shortly after kidney transplantation and is usually resistant to treatment with dismal renal outcomes. The complement genetic variants may accelerate the development of TMA. However, the complement genetic test was seldom performed in unknown native kidney disease recipients scheduled for LKT. Case presentation We reported three cases of unknown native kidney diseases who had fulminant TMA in the allograft shortly after LKT. Both the donors and the recipients were noted to carry complement genetic variants, which were identified by genetic testing after transplantation. However, all recipients were refractory to treatment and had allograft loss within 3 months after LKT. Conclusion This case series highlights the suggestion to screen complement gene variants in both the donors and the recipients with unknown native kidney diseases scheduled for LKT. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02868-7.
Collapse
Affiliation(s)
- Qianqian Wu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xiaohui Tian
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin Zheng
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Dandan Liang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xue Li
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xia Lu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wujun Xue
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Puxun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China.
| | - Jiqiu Wen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
15
|
Li XL, Luan CY, Fan YJ, Lin XY, Jiang D, Su MX, Wang G, Yang X. A Rare Case of Acute Infectious Purpura Fulminans Caused by Klebsiella Pneumoniae and Human Herpesvirus Type 5. J Inflamm Res 2022; 15:4251-4260. [PMID: 35923907 PMCID: PMC9340174 DOI: 10.2147/jir.s369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Purpura fulminans (PF), a rare, life-threatening disorder, is a hematological emergency in which there is skin necrosis, disseminated intravascular coagulation (DIC), and protein C deficiency. In PF, the skin necrosis and DIC are secondary to protein C deficiency. This may progress rapidly to multiorgan failure caused by the thrombotic occlusion of small- and medium-sized blood vessels. Case Report This article presents the case of a 22-year-old male with fever as well as necrotic and purpuric skin lesions. The ultrasound and computed tomography scans revealed infections in the skin wounds as well as venous microthrombosis and thrombosis in multiple intracranial and pulmonary vessels. The laboratory tests showed signs of sepsis, thrombocytopenia, an abnormal decrease in protein C and antithrombin III, DIC, multiple organ and system failures, gastric varices, and gastrointestinal hemorrhage. The blood, sputum, and secretions under the skin lesions were cultured and were positive for Klebsiella pneumoniae. The results of the high-throughput genetic testing of the pathogenic microorganism DNA were consistent. In addition, human herpesvirus type 5 was detected. The histopathological examination of the skin lesions revealed pathological features consistent with PF. After successful treatment by the departments of Dermatology, Emergency Critical Care Medicine, and the Intensive Care Unit, the patient was discharged after 67 days of hospitalization. Conclusion Adults with acquired protein C and/or S deficiency states, including certain bacterial and viral infections, who drink alcohol and take varieties of non-steroidal anti-inflammatory analgesics at the same time, may develop acute infectious PF. Clinicians should be aware of this for early diagnosis and treatment.
Collapse
Affiliation(s)
- Xiao-Lan Li
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
- Correspondence: Xiao-Lan Li, Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, No. 374 of Dian-Mian Avenue, Wu-Hua District, Kunming, 650101, People’s Republic of China, Tel +86 871 63402212, Fax +86 871 65334416, Email
| | - Chun-Yan Luan
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Ying-Jun Fan
- Department of Rheumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Xiao-Ying Lin
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Dong Jiang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Mei-Xian Su
- Department of Emergency Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Gang Wang
- Department of Intensive Care Unit, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| | - Xu Yang
- Laboratory Bacteria Room, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, People’s Republic of China
| |
Collapse
|
16
|
Tan JS, Ren JM, Fan L, Wei Y, Hu S, Zhu SS, Yang Y, Cai J. Genetic Predisposition of Anti-Cytomegalovirus Immunoglobulin G Levels and the Risk of 9 Cardiovascular Diseases. Front Cell Infect Microbiol 2022; 12:884298. [PMID: 35832381 PMCID: PMC9272786 DOI: 10.3389/fcimb.2022.884298] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/23/2022] [Indexed: 12/20/2022] Open
Abstract
Background Accumulating evidence has indicated that persistent human cytomegalovirus (HCMV) infection is associated with several cardiovascular diseases including atherosclerosis and coronary artery disease. However, whether there is a causal association between the level of anti-HCMV immune response and the risk of cardiovascular diseases remains unknown. Methods Single-nucleotide polymorphisms associated with anti-cytomegalovirus immunoglobulin (Ig) G levels were used as instrumental variables to estimate the causal effect of anti-cytomegalovirus IgG levels on 9 cardiovascular diseases (including atrial fibrillation, coronary artery disease, hypertension, heart failure, peripheral artery disease, pulmonary embolism, deep vein thrombosis of the lower extremities, rheumatic valve diseases, and non-rheumatic valve diseases). For each cardiovascular disease, Mendelian randomization (MR) analyses were performed. Inverse variance-weighted meta-analysis (IVW) with a random-effects model was used as a principal analysis. In addition to this, the weighted median approach and MR-Egger method were used for further sensitivity analysis. Results In the IVW analysis, genetically predicted anti-cytomegalovirus IgG levels were suggestively associated with coronary artery disease with an odds ratio (OR) of 1.076 [95% CI, 1.009–1.147; p = 0.025], peripheral artery disease (OR 1.709; 95% CI, 1.039–2.812; p = 0.035), and deep vein thrombosis (OR 1.002; 95% CI, 1.000–1.004; p = 0.025). In the further analysis, similar causal associations were obtained from weighted median analysis and MR-Egger analysis with lower precision. No notable heterogeneities and horizontal pleiotropies were observed (p > 0.05). Conclusions/Interpretation Our findings first provide direct evidence that genetic predisposition of anti-cytomegalovirus IgG levels increases the risk of coronary artery disease, peripheral artery disease, and deep vein thrombosis.
Collapse
Affiliation(s)
- Jiang-Shan Tan
- Emergency Center, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Meng Ren
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education. Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Luyun Fan
- Hypertension Center, FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuhao Wei
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Song Hu
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Key Laboratory of Pulmonary Vascular Medicine, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng-Song Zhu
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Key Laboratory of Pulmonary Vascular Medicine, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmin Yang
- Emergency Center, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yangmin Yang, ; Jun Cai,
| | - Jun Cai
- Hypertension Center, FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Yangmin Yang, ; Jun Cai,
| |
Collapse
|
17
|
Blasco M, Guillén-Olmos E, Diaz-Ricart M, Palomo M. Complement Mediated Endothelial Damage in Thrombotic Microangiopathies. Front Med (Lausanne) 2022; 9:811504. [PMID: 35547236 PMCID: PMC9082680 DOI: 10.3389/fmed.2022.811504] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/14/2022] [Indexed: 11/24/2022] Open
Abstract
Thrombotic microangiopathies (TMA) constitute a group of different disorders that have a common underlying mechanism: the endothelial damage. These disorders may exhibit different mechanisms of endothelial injury depending on the pathological trigger. However, over the last decades, the potential role of the complement system (CS) has gained prominence in their pathogenesis. This is partly due to the great efficacy of complement-inhibitors in atypical hemolytic syndrome (aHUS), a TMA form where the primary defect is an alternative complement pathway dysregulation over endothelial cells (genetic and/or adquired). Complement involvement has also been demonstrated in other forms of TMA, such as thrombotic thrombocytopenic purpura (TTP) and in Shiga toxin-producing Escherichia coli hemolytic uremic syndrome (STEC-HUS), as well as in secondary TMAs, in which complement activation occurs in the context of other diseases. However, at present, there is scarce evidence about the efficacy of complement-targeted therapies in these entities. The relationship between complement dysregulation and endothelial damage as the main causes of TMA will be reviewed here. Moreover, the different clinical trials evaluating the use of complement-inhibitors for the treatment of patients suffering from different TMA-associated disorders are summarized, as a clear example of the entry into a new era of personalized medicine in its management.
Collapse
Affiliation(s)
- Miquel Blasco
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud (CSUR), University of Barcelona, Barcelona, Spain.,Institute of Biomedical Research August Pi i Sunyer (IDIPABS), Malalties Nefro-Urològiques i Trasplantament Renal, Barcelona, Spain
| | - Elena Guillén-Olmos
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud (CSUR), University of Barcelona, Barcelona, Spain
| | - Maribel Diaz-Ricart
- Hematopathology Unit, Department of Pathology, Hospital Clínic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Barcelona Endothelium Team, Barcelona, Spain
| | - Marta Palomo
- Hematopathology Unit, Department of Pathology, Hospital Clínic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Barcelona Endothelium Team, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Schrottmaier WC, Schmuckenschlager A, Pirabe A, Assinger A. Platelets in Viral Infections - Brave Soldiers or Trojan Horses. Front Immunol 2022; 13:856713. [PMID: 35419008 PMCID: PMC9001014 DOI: 10.3389/fimmu.2022.856713] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Viral infections are often associated with platelet activation and haemostatic complications. In line, low platelet counts represent a hallmark for poor prognosis in many infectious diseases. The underlying cause of platelet dysfunction in viral infections is multifaceted and complex. While some viruses directly interact with platelets and/or megakaryocytes to modulate their function, also immune and inflammatory responses directly and indirectly favour platelet activation. Platelet activation results in increased platelet consumption and degradation, which contributes to thrombocytopenia in these patients. The role of platelets is often bi-phasic. Initial platelet hyper-activation is followed by a state of platelet exhaustion and/or hypo-responsiveness, which together with low platelet counts promotes bleeding events. Thereby infectious diseases not only increase the thrombotic but also the bleeding risk or both, which represents a most dreaded clinical complication. Treatment options in these patients are limited and new therapeutic strategies are urgently needed to prevent adverse outcome. This review summarizes the current literature on platelet-virus interactions and their impact on viral pathologies and discusses potential intervention strategies. As pandemics and concomitant haemostatic dysregulations will remain a recurrent threat, understanding the role of platelets in viral infections represents a timely and pivotal challenge.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anna Schmuckenschlager
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
20
|
How Immunosenescence and Inflammaging May Contribute to Hyperinflammatory Syndrome in COVID-19. Int J Mol Sci 2021; 22:ijms222212539. [PMID: 34830421 PMCID: PMC8618618 DOI: 10.3390/ijms222212539] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/16/2022] Open
Abstract
Aging is characterized by the dynamic remodeling of the immune system designated “immunosenescence,” and is associated with altered hematopoiesis, thymic involution, and lifelong immune stimulation by multitudinous chronic stressors, including the cytomegalovirus (CMV). Such alterations may contribute to a lowered proportion of naïve T-cells and to reduced diversity of the T-cell repertoire. In the peripheral circulation, a shift occurs towards accumulations of T and B-cell populations with memory phenotypes, and to accumulation of putatively senescent and exhausted immune cells. The aging-related accumulations of functionally exhausted memory T lymphocytes, commonly secreting pro-inflammatory cytokines, together with mediators and factors of the innate immune system, are considered to contribute to the low-grade inflammation (inflammaging) often observed in elderly people. These senescent immune cells not only secrete inflammatory mediators, but are also able to negatively modulate their environments. In this review, we give a short summary of the ways that immunosenescence, inflammaging, and CMV infection may cause insufficient immune responses, contribute to the establishment of the hyperinflammatory syndrome and impact the severity of the coronavirus disease 2019 (COVID-19) in elderly people.
Collapse
|
21
|
Pickering H, Sen S, Arakawa-Hoyt J, Ishiyama K, Sun Y, Parmar R, Ahn RS, Sunga G, Llamas M, Hoffmann A, Deng M, Bunnapradist S, Schaenman JM, Gjertson DW, Rossetti M, Lanier LL, Reed EF. NK and CD8+ T cell phenotypes predict onset and control of CMV viremia after kidney transplant. JCI Insight 2021; 6:153175. [PMID: 34609965 PMCID: PMC8663544 DOI: 10.1172/jci.insight.153175] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
CMV causes mostly asymptomatic but lifelong infection. Primary infection or reactivation in immunocompromised individuals can be life-threatening. CMV viremia often occurs in solid organ transplant recipients and associates with decreased graft survival and higher mortality. Furthering understanding of impaired immunity that allows CMV reactivation is critical to guiding antiviral therapy and examining the effect of CMV on solid organ transplant outcomes. This study characterized longitudinal immune responses to CMV in 31 kidney transplant recipients with CMV viremia and matched, nonviremic recipients. Recipients were sampled 3 and 12 months after transplant, with additional samples 1 week and 1 month after viremia. PBMCs were stained for NK and T cell markers. PBMC transcriptomes were characterized by RNA-Seq. Plasma proteins were quantified by Luminex. CD8+ T cell transcriptomes were characterized by single-cell RNA-Seq. Before viremia, patients had high levels of IL-15 with concurrent expansion of immature CD56bright NK cells. After viremia, mature CD56dim NK cells and CD28–CD8+ T cells upregulating inhibitory and NK-associated receptors were expanded. Memory NK cells and NK-like CD28–CD8+ T cells were associated with control of viremia. These findings suggest that signatures of innate activation may be prognostic for CMV reactivation after transplant, while CD8+ T cell functionality is critical for effective control of CMV.
Collapse
Affiliation(s)
- Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, California, USA
| | - Kenichi Ishiyama
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, California, USA
| | - Yumeng Sun
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Richard S Ahn
- Microbiology, Immunology, and Molecular Genetics.,Institute for Quantitative and Computational Biosciences, and
| | - Gemalene Sunga
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Megan Llamas
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Alexander Hoffmann
- Institute for Quantitative and Computational Biosciences, and.,Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Mario Deng
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Suphamai Bunnapradist
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, California, USA
| | - Joanna M Schaenman
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - David W Gjertson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, California, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | | |
Collapse
|
22
|
Matucci-Cerinic M, Hughes M, Taliani G, Kahaleh B. Similarities between COVID-19 and systemic sclerosis early vasculopathy: A "viral" challenge for future research in scleroderma. Autoimmun Rev 2021; 20:102899. [PMID: 34274540 PMCID: PMC8280663 DOI: 10.1016/j.autrev.2021.102899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To review similarities between COVID-19 and systemic sclerosis (SSc) early vasculopathy to provide novel insights into both diseases. METHODS A narrative review of the literature supplemented with expert opinion. RESULTS There is clear evidence that the endothelium is at the centre stage in SSc and COVID-19, with endothelial cell activation/injury and dysfunction creating the crucial evolving step in the pathogenesis of both diseases. The angiotensin system has also been implicated in the early stages of both COVID-19 and SSc. Autoptic studies provide novel insights into the effects of SARS-CoV-2 on the endothelium. Normal endothelium and endothelial dysfunction in COVID-19 and SSc are discussed. It is debated whether SARS-CoV-2 infection triggers autoimmunity with production of autoantibodies which is of mechanistic interest because other viral illnesses are potentially involved in endothelial dysfunction and in SSc pathogenesis. CONCLUSION COVID-19 is due to a direct assault of SARS-CoV-2 on the vascular system as an acute infection, whereas SSc remains a chronic/sub-acute autoimmune disease of largely unknown etiology Further study and exploration of the SARS-CoV-2 pathogenic mechanisms might provide further useful milestones in the understanding of the early SSc pathogenesis.
Collapse
Affiliation(s)
- Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence & Division of Rheumatology AOUC, Florence, Italy; Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy.
| | - Michael Hughes
- Department of Rheumatology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Gloria Taliani
- Infectious Diseases Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Bashar Kahaleh
- Division of Rheumatology, Allergy and Immunology, University of Toledo Medical Center, Toledo, OH, USA
| |
Collapse
|
23
|
Gabr JB, Bilal H, Mirchia K, Perl A. The Use of Eculizumab in Tacrolimus-Induced Thrombotic Microangiopathy. J Investig Med High Impact Case Rep 2021; 8:2324709620947266. [PMID: 32757799 PMCID: PMC7412890 DOI: 10.1177/2324709620947266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Drug-induced thrombotic microangiopathy (DITMA) is a secondary cause of thrombotic microangiopathy and a potentially fatal inflammatory disease. DITMA has been attributed to a variety of drugs, particularly chemotherapeutic and immunosuppressive agents. Prompt diagnosis is critical for survival and treatment necessitates withdrawal of the offending drug; however, many cases require further treatment including plasmapheresis, immunosuppression, and anticoagulation. In this article, we report a cutaneous biopsy-proven case of tacrolimus-induced DITMA, which was successfully treated with eculizumab after failing the conventional standard of care.
Collapse
Affiliation(s)
| | - Hiba Bilal
- SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Andras Perl
- SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
24
|
Walter G, Richert Q, Ponnampalam A, Sharma A. Acute superior mesenteric vein thrombosis in the setting of cytomegalovirus mononucleosis: a case report and review of the literature. THE LANCET. INFECTIOUS DISEASES 2021; 21:e202-e207. [PMID: 34000241 DOI: 10.1016/s1473-3099(20)30782-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/27/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022]
Abstract
Cytomegalovirus is a viral genus of the overarching family Herpesviridae, and is of particular importance because of its relevance to human disease. This association is predominantly due to human cytomegalovirus, a well-studied pathogen. In addition to the mononucleosis syndrome that can occur during acute cytomegalovirus viraemia, this virion has been recurrently implicated as a provoking factor for thromboembolic disease in the published scientific literature. As physicians increasingly forgo extensive laboratory investigation in the setting of clinical hypercoagulability, it has also become evident that in some circumstances whether or not a particular investigation alters clinical management is not necessarily the only important question. Viraemia as a provoking factor for thrombosis stands as such an example. The aim of this Grand Round is to further explore the role of cytomegalovirus as it pertains to thromboembolic disease, especially in the present era of viral-associated thromboembolism.
Collapse
Affiliation(s)
- Graham Walter
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| | - Quinlan Richert
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Arjuna Ponnampalam
- Section of Hematology/Oncology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; CancerCare Manitoba, Manitoba, Winnipeg, Canada
| | - Aditya Sharma
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada; Section of General Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
25
|
Yousaf Z, Albaz N, Abdelmajid AA, Sabobeh T, Elzouki A. Reactivation cytomegalovirus leading to acute myocardial infarction-A first reported case in an immunocompetent patient. Clin Case Rep 2021; 9:1958-1963. [PMID: 33936622 PMCID: PMC8077325 DOI: 10.1002/ccr3.3914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/23/2021] [Indexed: 12/04/2022] Open
Abstract
Reactivation of cytomegalovirus (CMV) in immunocompetent patients may lead to increase morbidity and mortality. A clinical suspicion allows timely diagnosis, treatment, and favorable outcome. In a subject without apparent risk factors for acute myocardial infarction (AMI), CMV can be a possibility.
Collapse
Affiliation(s)
- Zohaib Yousaf
- Department of MedicineHamad Medical CorporationDohaQatar
| | - Nadeen Albaz
- Department of MedicineHamad Medical CorporationDohaQatar
| | | | - Taher Sabobeh
- Department of MedicineHamad Medical CorporationDohaQatar
| | - Abdel‐Naser Elzouki
- Department of MedicineHamad Medical CorporationDohaQatar
- College of MedicineQatar UniversityDohaQatar
- Weill Cornell Medical CollegeDohaQatar
| |
Collapse
|
26
|
Söderberg-Nauclér C. Does reactivation of cytomegalovirus contribute to severe COVID-19 disease? IMMUNITY & AGEING 2021; 18:12. [PMID: 33712035 PMCID: PMC7952506 DOI: 10.1186/s12979-021-00218-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
The majority of people infected with SARS-CoV-2 are asymptomatic or have mild to moderate symptoms. However, for unknown reasons, about 15 % have severe pneumonia requiring hospital care and oxygen support, and about 5 % develop acute respiratory distress syndrome, septic shock, and multiorgan failure that result in a high mortality rate. The risk of severe COVID-19 is highest among those who are over 70 years of age. Why severe COVID-19 develops in some people but not others is not understood. Could some cases involve reactivation of latent cytomegalovirus (CMV)?
Collapse
Affiliation(s)
- Cecilia Söderberg-Nauclér
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, and Division of Neurology, Karolinska University Hospital, Bioclinicum, Visionsgatan 4, 171 64, Solna, Stockholm, Sweden.
| |
Collapse
|
27
|
Jain A, Jafri F, Manglani R, Al-Mufti F, Aronow WS, Chandy D. Stroke in critical COVID-19 patients: a cautionary tale from the frontlines. Arch Med Sci Atheroscler Dis 2020; 5:e263-e270. [PMID: 33644484 PMCID: PMC7885816 DOI: 10.5114/amsad.2020.102423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Although Coronavirus Disease 2019 (COVID-19) is primarily a disease of the respiratory system in its transmission and clinical manifestations, physicians have also reported a tropism toward the nervous system. METHODS Neurological symptoms can occur as one of many systemic manifestations of a critical form of the disease or in isolation as the predominant presenting complaint. RESULTS We report a series of 6 patients who suffered significant cerebrovascular accidents while being treated for critical COVID-19 in the intensive care units of a quaternary care hospital in New York's Hudson valley. CONCLUSIONS This series demonstrates how a relatively rare but catastrophic neurological complication can occur in patients with COVID-19 while they are being managed for their more common problems such as respiratory and renal failure.
Collapse
Affiliation(s)
- Anant Jain
- Department of Medicine, Westchester Medical Center, Valhalla, NY, USA
| | - Firas Jafri
- Department of Medicine, Westchester Medical Center, Valhalla, NY, USA
| | - Ravi Manglani
- Division of Pulmonary, Critical Care and Sleep Medicine, Westchester Medical Center, Valhalla, NY, USA
| | - Fawaz Al-Mufti
- Department of Neurology, Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert S. Aronow
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY, USA
| | - Dipak Chandy
- Division of Pulmonary, Critical Care and Sleep Medicine, Westchester Medical Center, Valhalla, NY, USA
| |
Collapse
|
28
|
Chieosilapatham P, Prinyaroj N, Jamjanya S, Kiratikanon S, Thinrungroj N, Kaewpoowat Q, Kongkarnka S, Mahanupab P, Tovanabutra N, Chiewchanvit S, Chuamanochan M. Degos-like lesions as a cutaneous manifestation of cytomegalovirus infection: A rare and serious complication in a patient with drug-induced hypersensitivity syndrome. J Dermatol 2020; 48:533-536. [PMID: 33305840 DOI: 10.1111/1346-8138.15717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/31/2020] [Accepted: 11/08/2020] [Indexed: 12/17/2022]
Abstract
Cytomegalovirus causes a myriad of clinical features, potentially affecting any organ system, significantly increasing morbidity and even mortality. Vascular endothelial cell infection by cytomegalovirus has been implicated in the development of vasculopathy, possibly accounting for the clinical association between cytomegalovirus and vascular thrombosis. In contrast with visceral organ involvement, the cutaneous manifestations of cytomegalovirus are variable and rarely described. Malignant atrophic papulosis, commonly known as Degos disease, is an unusual small vessel arteriopathy with a pathognomonic clinical appearance of atrophic porcelain-white central papules surrounded by telangiectatic erythema. As with the arterial occlusive process, Degos disease may be idiopathic or secondary to autoimmune disorders or viral infection. All in all, cytomegalovirus-related Degos-like presentation has never been described. This report describes a case in which disseminated cytomegalovirus disease developed 4 weeks after the onset of drug-induced hypersensitivity syndrome with prominent Degos-like skin lesions. Our case highlights a rare example of Degos-like lesions occurring due to cytomegalovirus disease and emphasizes the importance of early recognition of the characteristic cutaneous eruption as a diagnostic clue leading to the prompt management of this life-threatening infection.
Collapse
Affiliation(s)
- Panjit Chieosilapatham
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nayanunt Prinyaroj
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinda Jamjanya
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Salin Kiratikanon
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nithi Thinrungroj
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Quanhathai Kaewpoowat
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sarawut Kongkarnka
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pongsak Mahanupab
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Napatra Tovanabutra
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siri Chiewchanvit
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Mati Chuamanochan
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
29
|
Duarte I, Gameiro J, Outerelo C, Nogueira E, Lopes JA. Atypical Hemolytic Uremic Syndrome and Nephrotic Syndrome Associated with Cytomegalovirus Infection. Nephron Clin Pract 2020; 145:188-191. [PMID: 33291103 DOI: 10.1159/000511833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/26/2020] [Indexed: 11/19/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare disease. It results from the dysregulation of the alternative complement pathway on the cell surface which causes endothelial damage. Increasing evidence links, these abnormalities to mutations in genes of complement regulators or with autoantibodies against complement factors. These mutations have an incomplete penetrance and variable phenotype. Cytomegalovirus (CMV) is endemic throughout the world, and the incidence of severe CMV disease in immunocompetent adults appears to be greater than previously thought. aHUS and nephrotic syndromes associated with CMV infection are rare. Identification of triggers for aHUS manifestation in a genetically susceptible patient is extremely important since this permits a faster initiation of treatment and clinical improvement. We report a case of a man with a homozygotic deletion of CFHR3-1 whose initial presentation was aHUS and nephrotic syndromes associated with CMV infection.
Collapse
Affiliation(s)
- Inês Duarte
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal,
| | - Joana Gameiro
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Cristina Outerelo
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Estela Nogueira
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - José António Lopes
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| |
Collapse
|
30
|
Delicate Role of PD-L1/PD-1 Axis in Blood Vessel Inflammatory Diseases: Current Insight and Future Significance. Int J Mol Sci 2020; 21:ijms21218159. [PMID: 33142805 PMCID: PMC7663405 DOI: 10.3390/ijms21218159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are the antigen-independent generator of secondary signals that aid in maintaining the homeostasis of the immune system. The programmed death ligand-1 (PD-L1)/PD-1 axis is one among the most extensively studied immune-inhibitory checkpoint molecules, which delivers a negative signal for T cell activation by binding to the PD-1 receptor. The general attributes of PD-L1's immune-suppressive qualities and novel mechanisms on the barrier functions of vascular endothelium to regulate blood vessel-related inflammatory diseases are concisely reviewed. Though targeting the PD-1/PD-L1 axis has received immense recognition-the Nobel Prize in clinical oncology was awarded in the year 2018 for this discovery-the use of therapeutic modulating strategies for the PD-L1/PD-1 pathway in chronic inflammatory blood vessel diseases is still limited to experimental models. However, studies using clinical specimens that support the role of PD-1 and PD-L1 in patients with underlying atherosclerosis are also detailed. Of note, delicate balances in the expression levels of PD-L1 that are needed to preserve T cell immunity and to curtail acute as well as chronic infections in underlying blood vessel diseases are discussed. A significant link exists between altered lipid and glucose metabolism in different cells and the expression of PD-1/PD-L1 molecules, and its possible implications on vascular inflammation are justified. This review summarizes the most recent insights concerning the role of the PD-L1/PD-1 axis in vascular inflammation and, in addition, provides an overview exploring the novel therapeutic approaches and challenges of manipulating these immune checkpoint proteins, PD-1 and PD-L1, for suppressing blood vessel inflammation.
Collapse
|
31
|
Braxton AM, Chalmin AL, Najarro KM, Brockhurst JK, Johnson KT, Lyons CE, Daly B, Cryer CG, Vijay S, Cyphers G, Guerrero-Martin SM, Aston SA, McGee K, Su YP, Arav-Boger R, Metcalf Pate KA. Platelet-endothelial associations may promote cytomegalovirus replication in the salivary gland in mice. Platelets 2020; 31:860-868. [PMID: 31726921 PMCID: PMC7220825 DOI: 10.1080/09537104.2019.1689383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/24/2019] [Accepted: 10/31/2019] [Indexed: 10/25/2022]
Abstract
Platelet decline is a feature of many acute viral infections, including cytomegalovirus (CMV) infection in humans and mice. Platelet sequestration in association with other cells, including endothelium and circulating leukocytes, can contribute to this decline and influence the immune response to and pathogenesis of viral infection. We sought to determine if platelet-endothelial associations (PEAs) contribute to platelet decline during acute murine CMV (mCMV) infection, and if these associations affect viral load and production. Male BALB/c mice were infected with mCMV (Smith strain), euthanized at timepoints throughout acute infection and compared to uninfected controls. An increase in PEA formation was confirmed in the salivary gland at all post-inoculation timepoints using immunohistochemistry for CD41+ platelets co-localizing with CD34+ vessels. Platelet depletion did not change amount of viral DNA or timecourse of infection, as measured by qPCR. However, platelet depletion reduced viral titer of mCMV in the salivary glands while undepleted controls demonstrated robust replication in the tissue by plaque assay. Thus, platelet associations with endothelium may enhance the ability of mCMV to replicate within the salivary gland. Further work is needed to determine the mechanisms behind this effect and if pharmacologic inhibition of PEAs may reduce CMV production in acutely infected patients.
Collapse
Affiliation(s)
- Alicia M. Braxton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alyssa L. Chalmin
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kevin M. Najarro
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jacqueline K. Brockhurst
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, USA
| | - Karl T. Johnson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Claire E. Lyons
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Brenna Daly
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, USA
| | - Catherine G. Cryer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, USA
| | - Shefali Vijay
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Griffin Cyphers
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Selena M. Guerrero-Martin
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - S. Andrew Aston
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Psychiatry of Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kirstin McGee
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yu-Pin Su
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ravit Arav-Boger
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA
| | - Kelly A. Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
32
|
Vasilieva E, Gianella S, Freeman ML. Novel Strategies to Combat CMV-Related Cardiovascular Disease. Pathog Immun 2020; 5:240-274. [PMID: 33089035 PMCID: PMC7556413 DOI: 10.20411/pai.v5i1.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV), a ubiquitous human pathogen that is never cleared from the host, has long been thought to be relatively innocuous in immunocompetent adults, but causes severe complications including blindness, end-organ disease, and death in newborns and in immuno-compromised individuals, such as organ transplant recipients and those suffering from AIDS. Yet even in persons with intact immunity, CMV infection is associated with profound stimulation of immune and inflammatory pathways. Carriers of CMV infection also have an elevated risk of developing cardiovascular complications. In this review, we define the proposed mechanisms of how CMV contributes to cardiovascular disease (CVD), describe current approaches to target CMV, and discuss how these strategies may or may not alleviate cardiovascular complications in those with CMV infection. In addition, we discuss the special situation of CMV coinfection in people with HIV infection receiving antiretroviral therapy, and describe how these 2 viral infections may interact to potentiate CVD in this especially vulnerable population.
Collapse
Affiliation(s)
- Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine; Department of Medicine; Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
33
|
Zhu W, Liu S. The role of human cytomegalovirus in atherosclerosis: a systematic review. Acta Biochim Biophys Sin (Shanghai) 2020; 52:339-353. [PMID: 32253424 DOI: 10.1093/abbs/gmaa005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/05/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a progressive vascular disease with increasing morbidity and mortality year by year in modern society. Human cytomegalovirus (HCMV) infection is closely associated with the development of atherosclerosis. HCMV infection may accelerate graft atherosclerosis and the development of transplant vasculopathy in organ transplantation. However, our current understanding of HCMV-associated atherosclerosis remains limited and is mainly based on clinical observations. The underlying mechanism of the involvement of HCMV infection in atherogenesis remains unclear. Here, we summarized current knowledge regarding the multiple influences of HCMV on a diverse range of infected cells, including vascular endothelial cells, vascular smooth muscle cells, monocytes, macrophages, and T cells. In addition, we described potential HCMV-induced molecular mechanisms, such as oxidative stress, endoplasmic reticulum stress, autophagy, lipid metabolism, and miRNA regulation, which are involved in the development of HCMV-associated atherogenesis. Gaining an improved understanding of these mechanisms will facilitate the development of novel and effective therapeutic strategies for the treatment of HCMV-related cardiovascular disease.
Collapse
Affiliation(s)
- Wenbo Zhu
- Clinical Research Institute, First Affiliated Hospital, University of South China, Hengyang 421001, China
| | - Shuangquan Liu
- Clinical Laboratory, First Affiliated Hospital, University of South China, Hengyang 421001, China
| |
Collapse
|
34
|
Schattner A, Dubin I, Glick Y. Cytomegalovirus-Associated Splenic Infarction. Am J Med 2020; 133:e104-e105. [PMID: 31622579 DOI: 10.1016/j.amjmed.2019.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Ami Schattner
- Department of Medicine, Laniado Hospital, Sanz Medical Center, Netanya, and the Faculty of Medicine, Hebrew University and Hadassah Medical School, Jerusalem, Israel.
| | - Ina Dubin
- Department of Medicine, Laniado Hospital, Sanz Medical Center, Netanya, Jerusalem, Israel
| | - Yair Glick
- Department of Imaging, Laniado Hospital, Sanz Medical Center, Netanya, Jerusalem, Israel
| |
Collapse
|
35
|
Lebedeva A, Maryukhnich E, Grivel JC, Vasilieva E, Margolis L, Shpektor A. Productive Cytomegalovirus Infection Is Associated With Impaired Endothelial Function in ST-Elevation Myocardial Infarction. Am J Med 2020; 133:133-142. [PMID: 31295440 PMCID: PMC6940528 DOI: 10.1016/j.amjmed.2019.06.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND An association between productive cytomegalovirus infection and atherosclerosis was shown recently in several trials, including a previous study of ours. However, the mechanism involved in this association is still under investigation. Here, we addressed the interaction between productive cytomegalovirus infection and endothelial function in patients with ST-elevation myocardial infarction (STEMI). METHODS We analyzed the presence of cytomegaloviral DNA in plasma and endothelial function in 33 patients with STEMI and 33 volunteers without cardiovascular diseases, using real-time polymerase chain reaction (PCR) and a noninvasive test of flow-mediated dilation. RESULTS Both the frequency of presence and the load of cytomegaloviral DNA were higher in plasma of patients with STEMI than those in controls. This difference was independent of other cardiovascular risk factors (7.38 [1.36-40.07]; P = 0.02). The results of the flow-mediated dilation test were lower in patients in STEMI than in controls (5.0% [2.65%-3.09%] vs 12. %5 [7.5%-15.15%]; P = 0.004) and correlated negatively with the cytomegaloviral DNA load (Spearman R = -0.407; P = 0.019) independently of other cardiovascular risk factors. CONCLUSIONS Productive cytomegalovirus infection in patients with STEMI correlated negatively with endothelial function independently of other cardiovascular risk factors. The impact of cytomegalovirus on endothelial function may explain the role of cytomegalovirus in cardiovascular prognosis.
Collapse
Affiliation(s)
- Anna Lebedeva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Elena Maryukhnich
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | | | - Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Md.
| | - Alexander Shpektor
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow, Russia
| |
Collapse
|
36
|
Martirosyan A, Aminov R, Manukyan G. Environmental Triggers of Autoreactive Responses: Induction of Antiphospholipid Antibody Formation. Front Immunol 2019; 10:1609. [PMID: 31354742 PMCID: PMC6635959 DOI: 10.3389/fimmu.2019.01609] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/27/2019] [Indexed: 12/19/2022] Open
Abstract
Antiphospholipid antibodies (aPLs) comprise a diverse family of autoantibodies targeted against proteins with the affinity toward negatively charged phospholipids or protein-phospholipid complexes. Their clinical significance, including prothrombotic potential of anti-cardiolipin antibodies (aCLs), anti-β2-glycoprotein I antibodies (aβ2-GPIs), and lupus anti-coagulant (LA), is well-established. However, the ontogeny of these pathogenic aPLs remains less clear. While transient appearance of aPLs could be induced by various environmental factors, in genetically predisposed individuals these factors may eventually lead to the development of the antiphospholipid syndrome (APS). Since the first description of APS, it has been found that a wide variety of microbial and viral agents influence aPLs production and contribute to clinical manifestations of APS. Many theories attempted to explain the pathogenic potential of different environmental factors as well as a phenomenon termed molecular mimicry between β2-GPI molecule and infection-relevant structures. In this review, we summarize and critically assess the pathogenic and non-pathogenic formation of aPLs and its contribution to the development of APS.
Collapse
Affiliation(s)
- Anush Martirosyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology, Yerevan, Armenia.,Russian-Armenian (Slavonic) University, Yerevan, Armenia
| | - Rustam Aminov
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Gayane Manukyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology, Yerevan, Armenia.,Russian-Armenian (Slavonic) University, Yerevan, Armenia
| |
Collapse
|
37
|
Denham C, Tissier G, Golding A. Antiphospholipid antibody syndrome with thrombotic splenic infarcts associated with acute cytomegalovirus infection. Access Microbiol 2019; 1:e000032. [PMID: 32974541 PMCID: PMC7481732 DOI: 10.1099/acmi.0.000032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/17/2019] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION We describe a case of acute cytomegalovirus (CMV) infection complicated by acquired antiphospholipid antibodies and splenic thrombi. We discuss the associations between CMV infection and thrombosis risk and correlation with antiphospolipid antibodies. CASE PRESENTATION A previously healthy 32-year-old woman is hospitalized for acute abdominal pain and fever and found to have multiple splenic infarcts on an abdominal computed tomography (CT) scan. An infectious work-up is negative except for acute CMV, and a hypercoagulable work-up is only positive for antiphospholipid antibodies. The patient is discharged and placed on anti-coagulation therapy for 6 months. CONCLUSION Co-incident thrombosis and antiphospholipid antibody syndrome can occur with acute viral infections, including CMV. We discuss the viral infection-associated increased risk of developing blood clots and antiphospholipid antibodies as being either correlative with or causative of viral-induced thrombosis.
Collapse
Affiliation(s)
- Christopher Denham
- Trinity School of Medicine, Kingstown, St.Vincent and the Grenadines, West Indies
| | - Ginger Tissier
- Trinity School of Medicine, Kingstown, St.Vincent and the Grenadines, West Indies
| | - Amit Golding
- Baltimore VA/VAMHCS and University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Anfasa F, Goeijenbier M, Widagdo W, Siegers JY, Mumtaz N, Okba N, van Riel D, Rockx B, Koopmans MPG, Meijers JCM, Martina BEE. Zika Virus Infection Induces Elevation of Tissue Factor Production and Apoptosis on Human Umbilical Vein Endothelial Cells. Front Microbiol 2019; 10:817. [PMID: 31068911 PMCID: PMC6491739 DOI: 10.3389/fmicb.2019.00817] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 04/01/2019] [Indexed: 12/22/2022] Open
Abstract
Zika virus (ZIKV) infection is typically characterized by a mild disease presenting with fever, maculopapular rash, headache, fatigue, myalgia, and arthralgia. A recent animal study found that ZIKV-infected pregnant Ifnar -/-mice developed vascular damage in the placenta and reduced amount of fetal capillaries. Moreover, ZIKV infection causes segmental thrombosis in the umbilical cord of pregnant rhesus macaques. Furthermore, several case reports suggest that ZIKV infection cause coagulation disorders. These results suggest that ZIKV could cause an alteration in the host hemostatic response, however, the mechanism has not been investigated thus far. This paper aims to determine whether ZIKV infection on HUVECs induces apoptosis and elevation of tissue factor (TF) that leads to activation of secondary hemostasis. We infected HUVECs with two ZIKV strains and performed virus titration, immunostaining, and flow cytometry to confirm and quantify infection. We measured TF concentrations with flow cytometry and performed thrombin generation test (TGT) as a functional assay to assess secondary hemostasis. Furthermore, we determined the amount of cell death using flow cytometry. We also performed enzyme-linked immunosorbent assay (ELISA) to determine interleukin (IL)-6 and IL-8 production and conducted blocking experiments to associate these cytokines with TF expression. Both ZIKV strains infected and replicated to high titers in HUVECs. We found that infection induced elevation of TF expressions. We also showed that increased TF expression led to shortened TGT time. Moreover, the data revealed that infection induced apoptosis. In addition, there was a significant increase of IL-6 and IL-8 production in infected cells. Here we provide in vitro evidence that infection of HUVECs with ZIKV induces apoptosis and elevation of TF expression that leads to activation of secondary hemostasis.
Collapse
Affiliation(s)
- Fatih Anfasa
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Marco Goeijenbier
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Widagdo Widagdo
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jurre Y Siegers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Noreen Mumtaz
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nisreen Okba
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joost C M Meijers
- Department of Plasma Proteins, Sanquin Research, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Byron E E Martina
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Artemis One Health Research Institute, Delft, Netherlands
| |
Collapse
|
39
|
Yokose T, Obara H, Shinoda M, Nakano Y, Kitago M, Yagi H, Abe Y, Yamada Y, Matsubara K, Oshima G, Hori S, Ibuki S, Higashi H, Masuda Y, Hayashi M, Mori T, Kawaida M, Fujimura T, Hoshino K, Kameyama K, Kuroda T, Kitagawa Y. Colon perforation due to antigenemia-negative cytomegalovirus gastroenteritis after liver transplantation: A case report and review of literature. World J Gastroenterol 2019; 25:1899-1906. [PMID: 31057303 PMCID: PMC6478612 DOI: 10.3748/wjg.v25.i15.1899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/03/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) remains a critical complication after solid-organ transplantation. The CMV antigenemia (AG) test is useful for monitoring CMV infection. Although the AG-positivity rate in CMV gastroenteritis is known to be low at onset, almost all cases become positive during the disease course. We treated a patient with transverse colon perforation due to AG-negative CMV gastroenteritis, following a living donor liver transplantation (LDLT).
CASE SUMMARY The patient was a 52-year-old woman with decompensated liver cirrhosis as a result of autoimmune hepatitis who underwent a blood-type compatible LDLT with her second son as the donor. On day 20 after surgery, upper and lower gastrointestinal endoscopy (GE) revealed multiple gastric ulcers and transverse colon ulcers. The biopsy tissue immunostaining confirmed a diagnosis of CMV gastroenteritis. On day 28 after surgery, an abdominal computed tomography revealed transverse colon perforation, and simple lavage and drainage were performed along with an urgent ileostomy. Although the repeated remission and aggravation of CMV gastroenteritis and acute cellular rejection made the control of immunosuppression difficult, the upper GE eventually revealed an improvement in the gastric ulcers, and the biopsy samples were negative for CMV. The CMV-AG test remained negative, therefore, we had to evaluate the status of the CMV infection on the basis of the clinical symptoms and GE.
CONCLUSION This case report suggests a monitoring method that could be useful for AG-negative CMV gastroenteritis after a solid-organ transplantation.
Collapse
Affiliation(s)
- Takahiro Yokose
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Hideaki Obara
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Yutaka Nakano
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Yohei Yamada
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Kentaro Matsubara
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Go Oshima
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Shutaro Hori
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Sho Ibuki
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Hisanobu Higashi
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Yuki Masuda
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Masanori Hayashi
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Takehiko Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Miho Kawaida
- Department of Pathology, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Takumi Fujimura
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Ken Hoshino
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Kaori Kameyama
- Department of Pathology, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Tatsuo Kuroda
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo 1608582, Japan
| |
Collapse
|
40
|
Lebedeva AM, Shpektor AV, Vasilieva EY, Margolis LB. Cytomegalovirus Infection in Cardiovascular Diseases. BIOCHEMISTRY (MOSCOW) 2019; 83:1437-1447. [PMID: 30878019 DOI: 10.1134/s0006297918120027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Atherosclerosis underlies the development of many cardiovascular diseases that continue to hold a leading place among the causes of death in developed countries. The role of activated immune cells in atherosclerosis progression has been convincingly demonstrated, but the mechanism of their action remains poorly investigated. Since atherosclerosis is associated with chronic inflammatory response, involvement of viral and bacterial infections in atherogenesis has been examined. A special place among the infectious agents is held by human herpesviruses as the most common persistent viruses in human population coupled to chronic inflammation during atherosclerosis. We found that activation of cytomegalovirus (CMV, human herpesvirus 5) infection is associated with the emergence of acute coronary syndrome, which is in a good agreement with the data on productive CMV infection published elsewhere. In this review, we discuss the data obtained by us and other researchers regarding the role of cytomegalovirus infection and related potential mechanisms resulting in the expansion of atherosclerotic plaques during ischemic heart disease and stroke, including virus transfer to immune and endothelial cells via extracellular vesicles. In particular, the data presented in the review demonstrate that virus spreading in the vascular wall triggers immune system activation in atherosclerotic plaques and causes endothelial dysfunction. Moreover, productive CMV infection in patients with acute myocardial infarction correlates with the extent of endothelial dysfunction. The mechanisms described by us and other researchers may explain the role of CMV infection in atherosclerosis and development of ischemic heart disease.
Collapse
Affiliation(s)
- A M Lebedeva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia.
| | - A V Shpektor
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - E Yu Vasilieva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - L B Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
41
|
Konradt C, Hunter CA. Pathogen interactions with endothelial cells and the induction of innate and adaptive immunity. Eur J Immunol 2018; 48:1607-1620. [PMID: 30160302 DOI: 10.1002/eji.201646789] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 07/24/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022]
Abstract
There are over 10 trillion endothelial cells (EC) that line the vasculature of the human body. These cells not only have specialized functions in the maintenance of homeostasis within the circulation and various tissues but they also have a major role in immune function. EC also represent an important replicative niche for a subset of viral, bacterial, and parasitic organisms that are present in the blood or lymph; however, there are major gaps in our knowledge regarding how pathogens interact with EC and how this influences disease outcome. In this article, we review the literature on EC-pathogen interactions and their role in innate and adaptive mechanisms of resistance to infection and highlight opportunities to address prominent knowledge gaps.
Collapse
Affiliation(s)
- Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Koupenova M, Mick E, Corkrey HA, Huan T, Clancy L, Shah R, Benjamin EJ, Levy D, Kurt-Jones EA, Tanriverdi K, Freedman JE. Micro RNAs from DNA Viruses are Found Widely in Plasma in a Large Observational Human Population. Sci Rep 2018; 8:6397. [PMID: 29686252 PMCID: PMC5913337 DOI: 10.1038/s41598-018-24765-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/10/2018] [Indexed: 12/19/2022] Open
Abstract
Viral infections associate with disease risk and select families of viruses encode miRNAs that control an efficient viral cycle. The association of viral miRNA expression with disease in a large human population has not been previously explored. We sequenced plasma RNA from 40 participants of the Framingham Heart Study (FHS, Offspring Cohort, Visit 8) and identified 3 viral miRNAs from 3 different human Herpesviridae. These miRNAs were mostly related to viral latency and have not been previously detected in human plasma. Viral miRNA expression was then screened in the plasma of 2763 participants of the remaining cohort utilizing high-throughput RT-qPCR. All 3 viral miRNAs associated with combinations of inflammatory or prothrombotic circulating biomarkers (sTNFRII, IL-6, sICAM1, OPG, P-selectin) but did not associate with hypertension, coronary heart disease or cancer. Using a large observational population, we demonstrate that the presence of select viral miRNAs in the human circulation associate with inflammatory biomarkers and possibly immune response, but fail to associate with overt disease. This study greatly extends smaller singular observations of viral miRNAs in the human circulation and suggests that select viral miRNAs, such as those for latency, may not impact disease manifestation.
Collapse
Affiliation(s)
- Milka Koupenova
- University of Massachusetts Medical School, Department of Medicine, Division of Cardiovascular Medicine, Worcester, MA, 01605, USA.
| | - Eric Mick
- University of Massachusetts Medical School, Department of Quantitative Health Sciences, Worcester, MA, 01605, USA
| | - Heather A Corkrey
- University of Massachusetts Medical School, Department of Medicine, Division of Cardiovascular Medicine, Worcester, MA, 01605, USA
| | - Tianxiao Huan
- National Heart, Lung, and Blood Institute, National Institutes of Health (NHLBI) and Boston University's Framingham Heart Institute, Framingham, MA, 01702, USA
- Population Sciences Branch, NHLBI, Bethesda, Maryland, 20824, USA
| | - Lauren Clancy
- University of Massachusetts Medical School, Department of Medicine, Division of Cardiovascular Medicine, Worcester, MA, 01605, USA
| | - Ravi Shah
- Beth Israel Deaconess Medical Center, Cardiovascular Institute, Boston, MA, 02215, USA
| | - Emelia J Benjamin
- Boston University School of Medicine, Department of Medicine, Boston, MA, 02118, USA
- Boston University School of Public Health, Department of Epidemiology, Boston, MA, 02118, USA
- National Heart, Lung, and Blood Institute, National Institutes of Health (NHLBI) and Boston University's Framingham Heart Institute, Framingham, MA, 01702, USA
| | - Daniel Levy
- National Heart, Lung, and Blood Institute, National Institutes of Health (NHLBI) and Boston University's Framingham Heart Institute, Framingham, MA, 01702, USA
- Population Sciences Branch, NHLBI, Bethesda, Maryland, 20824, USA
| | - Evelyn A Kurt-Jones
- University of Massachusetts Medical School, Department of Medicine, Division of Infectious Disease and Immunology, Worcester, MA, 01605, USA
| | - Kahraman Tanriverdi
- University of Massachusetts Medical School, Department of Medicine, Division of Cardiovascular Medicine, Worcester, MA, 01605, USA
| | - Jane E Freedman
- University of Massachusetts Medical School, Department of Medicine, Division of Cardiovascular Medicine, Worcester, MA, 01605, USA
| |
Collapse
|
43
|
Tang ER, Chapman T, Finn LS, Leger KJ. Perforated jejunitis in a child with acute lymphoblastic leukemia treated with pegaspargase. Radiol Case Rep 2018; 13:568-572. [PMID: 29988797 PMCID: PMC6030564 DOI: 10.1016/j.radcr.2018.02.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/07/2018] [Indexed: 01/19/2023] Open
Abstract
Survival rates of children with acute lymphoblastic leukemia have improved since the incorporation of asparaginase in the treatment protocol, but the medication has potential serious complications, including vascular thrombosis. Here, we describe the case of a 13-year-old boy with pre-T-cell acute lymphoblastic leukemia whose treatment course was complicated by perforated jejunitis requiring resection of a portion of his small bowel. Pathologic assessment showed transmural ischemia, mesenteric venous and arterial thrombi, and scattered cytomegalovirus inclusion bodies. Pediatric mesenteric ischemia is rare, and its consideration in patients treated with asparaginase is discussed.
Collapse
Affiliation(s)
- Elizabeth R Tang
- Department of Radiology, Seattle Children's Hospital, University of Washington School of Medicine, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - Teresa Chapman
- Department of Radiology, Seattle Children's Hospital, University of Washington School of Medicine, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - Laura S Finn
- Department of Pathology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, USA
| | - Kasey J Leger
- Department of Pediatrics, Division of Hematology-Oncology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
44
|
Du Y, Zhang G, Liu Z. Human cytomegalovirus infection and coronary heart disease: a systematic review. Virol J 2018; 15:31. [PMID: 29409508 PMCID: PMC5801777 DOI: 10.1186/s12985-018-0937-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) infection is closely associated with coronary heart disease. Main body of the abstract In 1987, Adam et al. were the first to report an association between HCMV infection and atherosclerosis (AS), and later, many serum epidemiology and molecular biology studies showed that HCMV-infected endothelial cells play an important role in the development of AS. As patients with HCMV are generally susceptible to coronary heart disease, and with the increasing elderly population, a review of recent studies focusing on the relationships of HCMV infection and coronary heart disease is timely and necessary. Short conclusion The role of HCMV infection in the development of AS needs further study, since many remaining issues need to be explored and resolved. For example, whether HCMV promotes the development of coronary AS, and what the independent factors that lead to coronary artery AS by viral infection are. A comprehensive understanding of HCMV infection is needed in order to develop better strategies for preventing AS.
Collapse
Affiliation(s)
- Yu Du
- Department of Microbiology, Weifang Medical University, Weifang, 261053, China
| | - Guangxue Zhang
- Department of Clinical Laboratory, Shandong Qingzhou Rongjun Hospital, Qingzhou, 262500, China
| | - Zhijun Liu
- Department of Microbiology, Weifang Medical University, Weifang, 261053, China.
| |
Collapse
|
45
|
Seaby EG, Gilbert RD. Thrombotic microangiopathy following haematopoietic stem cell transplant. Pediatr Nephrol 2018; 33:1489-1500. [PMID: 28993886 PMCID: PMC6061668 DOI: 10.1007/s00467-017-3803-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/23/2017] [Accepted: 09/01/2017] [Indexed: 12/19/2022]
Abstract
Thrombotic microangiopathy is a potentially lethal complication of haematopoietic stem cell (bone marrow) transplantation. The pathophysiology is incompletely understood, although endothelial damage appears to be central. Platelet activation, neutrophil extracellular traps and complement activation appear to play key roles. Diagnosis may be difficult and universally accepted diagnostic criteria are not available. Treatment remains controversial. In some cases, withdrawal of calcineurin inhibitors is adequate. Rituximab and defibrotide also appear to have been used successfully. In severe cases, complement inhibitors such as eculizumab may play a valuable role. Further research is required to define the pathophysiology and determine both robust diagnostic criteria and the optimal treatment.
Collapse
Affiliation(s)
- Eleanor G. Seaby
- Human Genetics and Genomics Department, University of Southampton, Southampton, UK
| | - Rodney D. Gilbert
- Southampton Children’s Hospital and Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD UK
| |
Collapse
|
46
|
Guo N, Zhang N, Yan L, Cao X, Lv F, Wang J, Wang Y, Cong H. Down-regulation of single-stranded DNA-binding protein 1 expression induced by HCMV infection promotes lipid accumulation in cells. ACTA ACUST UNITED AC 2017; 50:e6389. [PMID: 28902926 PMCID: PMC5597284 DOI: 10.1590/1414-431x20176389] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/22/2017] [Indexed: 01/07/2023]
Abstract
The objective of this study was to observe the infection of human cytomegalovirus (HCMV) to human umbilical vein endothelial cells, and its effect on the expression of single-stranded DNA-binding protein (SSBP1) and on lipid metabolism in endothelial cells. We screened the differential expression of mRNAs after HCMV infection by suppression subtractive hybridization and the expression levels of SSBP1 mRNA and protein after HCMV infection by real-time PCR and western blot. After verification of successful infection by indirect immunofluorescent staining and RT-PCR, we found a differential expression of lipid metabolism-related genes including LDLR, SCARB, CETP, HMGCR, ApoB and LPL induced by HCMV infection. The expression levels of SSBP1 mRNA and protein after HCMV infection were significantly down-regulated. Furthermore, we found that upregulation of SSBP1 inhibited the expression of atherosclerosis-associated LDLR, SCARB, HMGCR, CETP as well as the accumulation of lipids in the cells. The results showed that the inhibition of SSBP1 by HCMV infection promotes lipid accumulation in the cells.
Collapse
Affiliation(s)
- N Guo
- Department of Cardiology, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China.,Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - N Zhang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - L Yan
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - X Cao
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - F Lv
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - J Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Y Wang
- Department of Cardiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - H Cong
- Department of Cardiology, Tianjin Chest Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
47
|
Fleck-Derderian S, McClellan W, Wojcicki JM. The association between cytomegalovirus infection, obesity, and metabolic syndrome in U.S. adult females. Obesity (Silver Spring) 2017; 25:626-633. [PMID: 28229547 DOI: 10.1002/oby.21764] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 11/17/2016] [Accepted: 12/05/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The purpose of this analysis was to determine whether cytomegalovirus (CMV) infection is associated with the prevalence of metabolic syndrome (MetS) and whether this relationship differs by BMI. METHODS Data from the 1999-2004 National Health and Nutrition Examination Survey (NHANES) were pooled (N = 2,532). Logistic regression was used for assessing the association between CMV and MetS, stratified by gender and BMI, categorized as normal weight, overweight, obesity, and extreme obesity, and adjusted for age, race/ethnicity, and poverty level. RESULTS In unadjusted analyses, CMV infection was significantly associated with MetS in females (OR: 1.50; 95% CI: 1.1-2.1) but not males. After adjusting for confounders, the odds of MetS were higher in CMV+ normal-weight females (aOR: 65.31; 95% CI: 6.8-625.6) but lower in CMV+ females with extreme obesity (aOR: 0.25; 95% CI: 0.1-0.9). CMV infection was associated with higher high-density lipoprotein cholesterol (HDL-C) and lower triglycerides in females with extreme obesity but lower HDL-C in normal-weight females. CONCLUSIONS CMV infection was found to be associated with unique MetS phenotypes that differ between BMI categories and gender. Seropositive normal-weight females had a higher prevalence of MetS and dyslipidemia, while infection in females with extreme obesity was associated with a more metabolically benign profile.
Collapse
Affiliation(s)
- Shannon Fleck-Derderian
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - William McClellan
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Janet M Wojcicki
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
48
|
Salehi N, Choi ED, Garrison RC. A Case of Miller Fisher Syndrome, Thromboembolic Disease, and Angioedema: Association or Coincidence? AMERICAN JOURNAL OF CASE REPORTS 2017; 18:52-59. [PMID: 28090073 PMCID: PMC5260666 DOI: 10.12659/ajcr.901940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Miller Fisher Syndrome is characterized by the clinical triad of ophthalmoplegia, ataxia, and areflexia, and is considered to be a variant of Guillain-Barre Syndrome. Miller Fisher Syndrome is observed in approximately 1-5% of all Guillain-Barre cases in Western countries. Patients with Miller Fisher Syndrome usually have good recovery without residual deficits. Venous thromboembolism is a common complication of Guillain-Barre Syndrome and has also been reported in Miller Fisher Syndrome, but it has generally been reported in the presence of at least one prothrombotic risk factor such as immobility. A direct correlation between venous thromboembolism and Miller Fisher Syndrome or Guillain-Barre Syndrome has not been previously described. CASE REPORT We report the case of a 32-year-old Hispanic male who presented with acute, severe thromboembolic disease and concurrently demonstrated characteristic clinical features of Miller Fisher Syndrome including ophthalmoplegia, ataxia, and areflexia. Past medical and family history were negative for thromboembolic disease, and subsequent hypercoagulability workup was unremarkable. During the course of hospitalization, the patient also developed angioedema. CONCLUSIONS We describe a possible association between Miller Fisher Syndrome, thromboembolic disease, and angioedema.
Collapse
Affiliation(s)
- Nooshin Salehi
- Department of Medicine, Riverside University Health System, Moreno Valley, CA, USA
| | - Eric D Choi
- School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Roger C Garrison
- Department of Medicine, Riverside University Health System, Moreno Valley, CA, USA.,School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
49
|
Kaminski H, Fishman JA. The Cell Biology of Cytomegalovirus: Implications for Transplantation. Am J Transplant 2016; 16:2254-69. [PMID: 26991039 DOI: 10.1111/ajt.13791] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/17/2016] [Accepted: 03/07/2016] [Indexed: 01/25/2023]
Abstract
Interpretation of clinical data regarding the impact of cytomegalovirus (CMV) infection on allograft function is complicated by the diversity of viral strains and substantial variability of cellular receptors and viral gene expression in different tissues. Variation also exists in nonspecific (monocytes and dendritic cells) and specific (NK cells, antibodies) responses that augment T cell antiviral activities. Innate immune signaling pathways and expanded pools of memory NK cells and γδ T cells also serve to amplify host responses to infection. The clinical impact of specific memory T cell anti-CMV responses that cross-react with graft antigens and alloantigens is uncertain but appears to contribute to graft injury and to the abrogation of allograft tolerance. These responses are modified by diverse immunosuppressive regimens and by underlying host immune deficits. The impact of CMV infection on the transplant recipient reflects cellular changes and corresponding host responses, the convergence of which has been termed the "indirect effects" of CMV infection. Future studies will clarify interactions between CMV infection and allograft injury and will guide interventions that may enhance clinical outcomes in transplantation.
Collapse
Affiliation(s)
- H Kaminski
- Kidney Transplant Unit, CHU Bordeaux Pellegrin, Place Raba Léon, Bordeaux, France
| | - J A Fishman
- Transplant Infectious Disease and Immunocompromised Host Program and MGH Transplant Center, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
50
|
Chou JW, Cheng KS. Pulmonary embolism in an immunocompetent patient with acute cytomegalovirus colitis. Intest Res 2016; 14:187-90. [PMID: 27175121 PMCID: PMC4863054 DOI: 10.5217/ir.2016.14.2.187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/01/2016] [Accepted: 01/08/2016] [Indexed: 11/29/2022] Open
Abstract
Acute cytomegalovirus (CMV) infection occurs commonly in immunocompromised and immunocompetent patients, but is usually asymptomatic in the latter. Vascular events associated with acute CMV infection have been described, but are rare. Hence, such events are rarely reported in the literature. We report a case of pulmonary embolism secondary to acute CMV colitis in an immunocompetent 78-year-old man. The patient presented with fever and diarrhea. Colonic ulcers were diagnosed based on colonoscopy findings, and CMV was the proven etiology on pathological examination. The patient subsequently experienced acute respiratory failure. Pulmonary embolism was diagnosed based on the chest radiography and computed tomography findings. A diagnosis of acute CMV colitis complicated by pulmonary embolism was made. The patient was successfully treated with intravenous administration of unfractionated heparin and intravenous ganciclovir.
Collapse
Affiliation(s)
- Jen-Wei Chou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung, Taiwan, Republic of China
| | - Ken-Sheng Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|