1
|
Onisor D, Brusnic O, Mocan S, Stoian M, Avram C, Boicean A, Dobru D. Cytomegalovirus in Ulcerative Colitis: An Unwanted "Guest". Pathogens 2024; 13:650. [PMID: 39204250 PMCID: PMC11356953 DOI: 10.3390/pathogens13080650] [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: 06/22/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
The role of cytomegalovirus (CMV) in the flare-up of ulcerative colitis (UC) is not clearly understood. CMV can cause similar symptoms in different clinical contexts, which may be attributed to the natural evolution of the viral infection, the patient's immune status, or its association with inflammatory bowel disease (IBD). This study aims to delineate the diverse manifestations of CMV-related lesions from clinical, endoscopic, and histopathological perspectives, alongside a brief narrative review of the literature. In managing IBD patients, it is crucial to be vigilant for signs of CMV reactivation, especially before the initiation of more intensive therapies.
Collapse
Affiliation(s)
- Danusia Onisor
- Department of Internal Medicine VII, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Street No. 38, 540139 Targu Mures, Romania; (D.O.); (D.D.)
- Gastroenterology Department, Mureș County Clinical Hospital, 540103 Targu Mures, Romania
| | - Olga Brusnic
- Department of Internal Medicine VII, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Street No. 38, 540139 Targu Mures, Romania; (D.O.); (D.D.)
- Gastroenterology Department, Mureș County Clinical Hospital, 540103 Targu Mures, Romania
| | - Simona Mocan
- Pathology Department, Emergency County Hospital, 540136 Targu Mures, Romania;
| | - Mircea Stoian
- Department of Anesthesiology and Intensive Care, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Calin Avram
- Department of Medical Informatics and Biostatistics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Street No. 38, 540139 Targu Mures, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania;
| | - Daniela Dobru
- Department of Internal Medicine VII, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Street No. 38, 540139 Targu Mures, Romania; (D.O.); (D.D.)
- Gastroenterology Department, Mureș County Clinical Hospital, 540103 Targu Mures, Romania
| |
Collapse
|
2
|
Momayaz Sanat Z, Siami Z, Alatab S, Vahedi H, Fanni Z. Cytomegalovirus Infection in Adult Patients with Inflammatory Bowel Disease: A Literature Review. ARCHIVES OF IRANIAN MEDICINE 2024; 27:277-286. [PMID: 38690795 PMCID: PMC11097322 DOI: 10.34172/aim.2024.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/13/2024] [Indexed: 05/03/2024]
Abstract
Human cytomegalovirus (HCMV) is classified within the Herpesvirales order and is prevalent in 50%‒80% of the general population. Most carriers experience this infection without noticeable clinical symptoms. HCMV causes a lifelong latent infection that can be reactivated due to immune disorders and inflammation. The reactivation of HCMV becomes particularly significant when it coincides with inflammatory bowel disease (IBD). While cytomegalovirus (CMV) colitis in IBD patients was identified years ago, the role of CMV in triggering flare-ups, acute severe colitis, treatment resistance, and other outcomes in IBD patients experiencing CMV reactivation remains a subject of ongoing debate. In this review, we aim to address an updated insight into aspects related to the CMV colitis in IBD patients including epidemiology, risk factors, clinical features, diagnostic tests, histology, place of immunosuppressants and indications for antiviral treatment. We suggest for personalized and thorough assessment based on the disease phase and colitis severity when prescribing drugs to these patients. Furthermore, we emphasize the importance of regular patient follow-up to monitor drug side effects, ensuring treatment success, and minimizing the risk of colectomy.
Collapse
Affiliation(s)
- Zahra Momayaz Sanat
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Siami
- Department of Infectious Disease, School of Medicine, Ziaeian hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sudabeh Alatab
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Homayoon Vahedi
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Fanni
- Ziaeian Hospital, Tehran university of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Gocho K, Yamashita A, Iizuka N, Sato K, Imasaka K, Hamanaka N, Kimura T. Primary Cytomegalovirus Pneumonia Successfully Treated with Corticosteroid Therapy and Valganciclovir. Intern Med 2024; 63:271-276. [PMID: 37225488 PMCID: PMC10864076 DOI: 10.2169/internalmedicine.1638-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/23/2023] [Indexed: 05/26/2023] Open
Abstract
Cytomegalovirus infection is typically asymptomatic in immunocompetent individuals. A 26-year-old woman was admitted to our hospital with a fever and breathlessness. Chest computed tomography (CT) revealed bilateral diffuse reticulation and nodules. Laboratory investigations showed atypical lymphocytosis and increased transaminases. She was treated with corticosteroid pulse therapy because of acute lung injury, and her clinical condition improved. Based on the presence of cytomegalovirus antibodies, antigen, and polymerase chain reaction findings, she was diagnosed with primary cytomegalovirus pneumonia and treated with valganciclovir. Primary cytomegalovirus pneumonia is very rare in immunocompetent individuals. The efficacy of corticosteroid and valganciclovir against cytomegalovirus pneumonia in this patient is noteworthy.
Collapse
Affiliation(s)
- Kyoko Gocho
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Aya Yamashita
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Noboru Iizuka
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Kenya Sato
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Keisuke Imasaka
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Nobuyuki Hamanaka
- Department of Respiratory Medicine, Saiseikai Yokohamashi Tobu Hospital, Japan
| | - Tokuhiro Kimura
- Department of Diagnostic Pathology, Saiseikai Yokohamashi Tobu Hospital, Japan
| |
Collapse
|
4
|
Stern L, McGuire HM, Avdic S, Blyth E, Gottlieb D, Patrick E, Abendroth A, Slobedman B. Circulating cytokine and chemokine patterns associated with cytomegalovirus reactivation after stem cell transplantation. Clin Transl Immunology 2023; 12:e16815. [PMID: 38034080 PMCID: PMC10684332 DOI: 10.1002/cti2.1473] [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: 05/01/2023] [Revised: 08/31/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Objectives Human cytomegalovirus (HCMV) reactivation is the leading viral complication after allogeneic haematopoietic stem cell transplantation (allo-HSCT). Understanding of circulating cytokine/chemokine patterns which accompany HCMV reactivation and correlate with HCMV DNAemia magnitude is limited. We aimed to characterise plasma cytokine/chemokine profiles in 36 allo-HSCT patients (21 with HCMV reactivation and 15 without HCMV reactivation) at four time-points in the first 100-day post-transplant. Methods The concentrations of 31 cytokines/chemokines in plasma samples were analysed using a multiplex bead-based immunoassay. Cytokine/chemokine concentrations were compared in patients with high-level HCMV DNAemia, low-level HCMV DNAemia or no HCMV reactivation, and correlated with immune cell frequencies measured using mass cytometry. Results Increased plasma levels of T helper 1-type cytokines/chemokines (TNF, IL-18, IP-10, MIG) were detected in patients with HCMV reactivation at the peak of HCMV DNAemia, relative to non-reactivators. Stem cell factor (SCF) levels were significantly higher before the detection of HCMV reactivation in patients who went on to develop high-level HCMV DNAemia (810-52 740 copies/mL) vs. low-level HCMV DNAemia (< 250 copies/mL). High-level HCMV reactivators, but not low-level reactivators, developed an elevated inflammatory cytokine/chemokine profile (MIP-1α, MIP-1β, TNF, LT-α, IL-13, IL-9, SCF, HGF) at the peak of reactivation. Plasma cytokine concentrations displayed unique correlations with circulating immune cell frequencies in patients with HCMV reactivation. Conclusion This study identifies distinct circulating cytokine/chemokine signatures associated with the magnitude of HCMV DNAemia and the progression of HCMV reactivation after allo-HSCT, providing important insight into immune recovery patterns associated with HCMV reactivation and viral control.
Collapse
Affiliation(s)
- Lauren Stern
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| | - Helen M McGuire
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| | - Selmir Avdic
- Westmead Institute for Medical ResearchThe University of SydneySydneyNSWAustralia
| | - Emily Blyth
- Westmead Institute for Medical ResearchThe University of SydneySydneyNSWAustralia
- Blood Transplant and Cell Therapies Program, Department of HaematologyWestmead HospitalSydneyNSWAustralia
- Faculty of Medicine and Health, Sydney Medical SchoolThe University of SydneySydneyNSWAustralia
| | - David Gottlieb
- Westmead Institute for Medical ResearchThe University of SydneySydneyNSWAustralia
- Blood Transplant and Cell Therapies Program, Department of HaematologyWestmead HospitalSydneyNSWAustralia
- Faculty of Medicine and Health, Sydney Medical SchoolThe University of SydneySydneyNSWAustralia
| | - Ellis Patrick
- Westmead Institute for Medical ResearchThe University of SydneySydneyNSWAustralia
- School of Mathematics and StatisticsThe University of SydneySydneyNSWAustralia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
| |
Collapse
|
5
|
Hatayama Y, Watanabe K, Ichikawa H, Kawamura K, Fukuda T, Motokura T. Differential Reactivation of Cytomegalovirus and Epstein-Barr Virus in Patients with B Cell Lymphoma. Viral Immunol 2023; 36:520-525. [PMID: 37440168 DOI: 10.1089/vim.2023.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023] Open
Abstract
Although cytomegalovirus (CMV) and Epstein-Barr virus (EBV) are considered latent viruses, their reactivation occurs in immunosuppressed conditions. We previously reported that CMV and EBV are reactivated in patients receiving immunosuppressive therapy and/or chemotherapy. This retrospective, single-center study aimed to determine the frequency of viral reactivation and clinical characteristics of patients with B cell lymphoma (B-ML) receiving chemotherapy. Twenty-four patients (mean age 73 years, range 40-87 years; male-to-female ratio, 15:9) with diffuse large B cell lymphoma (n = 15), follicular lymphoma (n = 8), or mantle cell lymphoma (n = 1) were enrolled. Serum CMV and EBV DNA levels were analyzed using quantitative real-time polymerase chain reaction in patients with B-ML receiving chemotherapy. We determined the cumulative reactivation of each virus and analyzed the relationship between viral reactivation and clinical characteristics. Three patients experienced relapse or refractory (R/R) disease and the others had de novo lymphomas. The frequencies of CMV and EBV reactivations were 54.2% and 37.5%, respectively. CMV reactivation occurred significantly earlier during chemotherapy courses in R/R patients than in de novo patients (p = 0.0038), while EBV reactivation was frequently found before treatment. Baseline serum levels of soluble interleukin-2 receptor were higher (4318.0 vs. 981.1 U/mL, p = 0.010) and hemoglobin levels were lower (11.1 vs. 13.0 g/dL, p = 0.0038) in patients with EBV reactivation than in those without reactivation. These findings were not observed in patients with CMV reactivation. CMV reactivation was associated with iatrogenic immunosuppression, whereas EBV reactivation was related to immunosuppression by lymphoma, indicating that the mechanisms of these viral reactivations differed.
Collapse
Affiliation(s)
- Yuki Hatayama
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Japan
| | - Kanako Watanabe
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Japan
| | - Hitomi Ichikawa
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Japan
| | - Koji Kawamura
- Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, School of Medicine, Tottori University Faculty of Medicine, Yonago, Japan
| | - Tetsuya Fukuda
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Japan
| | - Toru Motokura
- Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, School of Medicine, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
6
|
Zhu F, Hu Z, Yu W, Dai F, Jing D, Zhou G. Ulcerative Colitis Concomitant with Cytomegalovirus Infection, Bullous Sweet's Syndrome, and Acute Myeloid Leukemia: A Case Report and Literature Review. J Inflamm Res 2023; 16:3715-3723. [PMID: 37663756 PMCID: PMC10473406 DOI: 10.2147/jir.s422057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023] Open
Abstract
Background Ulcerative colitis (UC) is a chronic, relapsing progressive inflammatory immune disease. There is still no cure for it. Even worse, UC may predispose patients to opportunistic infections, and several extra-intestinal manifestations (EIMs) and comorbidities may antedate, occur with, or postdate the onset of UC, which may increase the mortality risk. But case reports of UC patients simultaneously concomitant with opportunistic infection, EIM, and comorbidity are extremely rare. Case Presentation We report a case of 51-year-old male patient with incipient UC accompanied by cytomegalovirus (CMV) infection and bullous Sweet's syndrome (bSS, a cutaneous EIM of UC) after treatment with oral mesalazine and prednisolone for 3 weeks. After clearance of the CMV infection by using ganciclovir, the patient was administered two cycles of infliximab to cure UC and bSS; however, he developed acute myeloid leukemia (AML) a month later and died after two cycles of chemotherapy. Conclusion Based on this rare case of UC concomitant with CMV infection, bSS and AML, we recommend that it is important to distinguish between an acute UC flare and opportunistic infections, especially in patients receiving immunosuppressive therapy, and monitor EIMs and comorbidities timely. Particular attention should be paid to cancer surveillance. Clinicians should be mindful of these facts to adopt optimal therapeutic options to address all aspects of UC. Early initiation of biological therapy may be of benefit to patients with newly diagnosed severe UC.
Collapse
Affiliation(s)
- Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250355, People’s Republic of China
| | - Zongjing Hu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Wei Yu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Fengxian Dai
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| |
Collapse
|
7
|
Pinana M, Rapoport C, Champtiaux N, Lescaille G, Allenbach Y, Rochefort J. Cytomegalovirus-induced oral ulcers: A case report and literature review. Clin Case Rep 2023; 11:e7459. [PMID: 37305877 PMCID: PMC10248210 DOI: 10.1002/ccr3.7459] [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: 12/27/2022] [Revised: 02/22/2023] [Accepted: 05/19/2023] [Indexed: 06/13/2023] Open
Abstract
Cytomegalovirus (CMV) ulcerations are rare clinical entities, but their occurrence is favored in immunocompromised patients who present a favorable environment for opportunistic infections. We describe the case of a patient treated for a systemic lupus erythematosus suffering from deep oral ulcerations. The case illustrates the complexity of establishing a precise etiological diagnosis of CMV lesions, as the diagnostic hypothesis can be varied: related to an immunodeficiency disorder or drug-induced toxidermia.
Collapse
Affiliation(s)
- Margaux Pinana
- Department of Oral Mucosal Pathology, AP‐HP Groupe Hospitalier Pitié‐SalpêtrièreUniversity of ParisParisFrance
| | - Camille Rapoport
- Department of Anatomopathology, AP‐HP Groupe Hospitalier Pitié‐SalpêtrièreSorbonne UniversityParisFrance
| | - Nicolas Champtiaux
- Department of Internal MedicineGroupe Hospitalier Pitié‐SalpêtrièreParisFrance
| | - Géraldine Lescaille
- Department of Oral Mucosal Pathology, AP‐HP Groupe Hospitalier Pitié‐SalpêtrièreUniversity of ParisParisFrance
| | - Yves Allenbach
- Department of Internal MedicineGroupe Hospitalier Pitié‐SalpêtrièreParisFrance
| | - Juliette Rochefort
- Department of Oral Mucosal Pathology, AP‐HP Groupe Hospitalier Pitié‐SalpêtrièreUniversity of ParisParisFrance
| |
Collapse
|
8
|
Parhizgari N, Zarei Ghobadi M, Rezaei F, Maraashi SM, Khatami MR, Mokhtari-Azad T. Transcriptomic analysis of human cytomegalovirus to survey the indirect effects on renal transplant recipients. Transpl Immunol 2023; 78:101746. [PMID: 36796459 DOI: 10.1016/j.trim.2022.101746] [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: 07/01/2022] [Revised: 10/02/2022] [Accepted: 11/13/2022] [Indexed: 02/17/2023]
Abstract
Post-transplant human cytomegalovirus (HCMV) viremia has been linked to adverse "indirect effects" among transplant patients. HCMV-created immunomodulatory mechanisms could be associated with the indirect effects. OBJECTIVE In the present study, the RNA-Seq whole transcriptome of renal transplant (RT) patients was analyzed to seek the underlying pathobiologic pathways associated with the long-term indirect effects of HCMV. METHODS To investigate the activated biological pathways in HCMV infection, total RNA was extracted from PBMCs of 2 RT patients with active HCMV and 2 RT patients without infection and then were sequenced using RNA-Seq. The resulted raw data were analyzed by conventional RNA-Seq software to determine the Differentially Expressed Genes (DEGs). Afterward, Gene Ontology (GO) and pathway enrichment analyses were conducted to determine the enriched pathways and biological processes by DEGs. Eventually, the relative expressions of some significant genes were validated in the twenty external RT patients. RESULT The analysis of RNA-Seq data related to RT patients with HCMV active viremia led to the identification of 140 up-regulated and 100 down-regulated DEGs. KEGG pathway analysis revealed the enrichment of DEGs in IL18 signaling, AGE-RAGE signaling pathway in diabetic complications, signaling by GPCR, Platelet activation, signaling and aggregation, Estrogen signaling pathway and signaling by Wnt due to HCMV infection. The expression levels of six genes involved in enriched pathways including F3, PTX3, ADRA2B, GNG11, GP9, HBEGF were then verified using RT-qPCR. The results were in consistent with RNA-Seq resultsoutcomes. CONCLUSION This study specifies some pathobiological pathways which are activated in HCMV active infection and could be linked to the adverse indirect effects caused by HCMV infection in transplant patients.
Collapse
Affiliation(s)
- Najmeh Parhizgari
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Zarei Ghobadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahdi Maraashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Zhang J, Kamoi K, Zong Y, Yang M, Ohno-Matsui K. Cytomegalovirus Anterior Uveitis: Clinical Manifestations, Diagnosis, Treatment, and Immunological Mechanisms. Viruses 2023; 15:185. [PMID: 36680225 PMCID: PMC9867175 DOI: 10.3390/v15010185] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
Little is known regarding anterior uveitis (AU), the most common ocular disease associated with cytomegalovirus (CMV) infection in immunocompetent populations. CMV AU is highly prevalent in Asia, with a higher incidence in men. Clinically, it manifests mainly as anterior chamber inflammation and elevated intraocular pressure (IOP). Acute CMV AU may resemble Posner-Schlossman syndrome with its recurrent hypertensive iritis, while chronic CMV AU may resemble Fuchs uveitis because of its elevated IOP. Without prompt treatment, it may progress to glaucoma; therefore, early diagnosis is critical to prognosis. Knowledge regarding clinical features and aqueous humor analyses can facilitate accurate diagnoses; so, we compared and summarized these aspects. Early antiviral treatment reduces the risk of a glaucoma surgery requirement, and therapeutic effects vary based on drug delivery. Both oral valganciclovir and topical ganciclovir can produce positive clinical outcomes, and higher concentration and frequency are beneficial in chronic CMV retinitis. An extended antiviral course could prevent relapses, but should be limited to 6 months to prevent drug resistance and side effects. In this review, we have systematically summarized the pathogenesis, clinical features, diagnostic and therapeutic aspects, and immunological mechanisms of CMV AU with the goal of providing a theoretical foundation for early clinical diagnosis and treatment.
Collapse
Affiliation(s)
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | | | | | | |
Collapse
|
10
|
Shen HC, Feng JY, Sun CY, Huang JR, Chen YM, Chen WC, Yang KY. Analysis of the effect of cytomegalovirus infection in clinical outcomes and prolonged duration of SARS-CoV-2 shedding in intensive care unit patients with COVID-19 pneumonia. Ther Adv Respir Dis 2023; 17:17534666231209150. [PMID: 37949827 PMCID: PMC10640799 DOI: 10.1177/17534666231209150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a global outbreak disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Cytomegalovirus (CMV) infection can occur in critical COVID-19 patients and is associated with adverse clinical outcomes. OBJECTIVE The aim of this study was to explore the clinical characteristics and outcome of CMV infection in critical COVID-19 patients. DESIGN This was a retrospective cohort study. METHODS From May to September 2021, SARS-CoV-2 reverse transcription polymerase chain reaction (RT-PCR)-confirmed COVID-19 patients with intensive care unit (ICU) admission were enrolled. CMV infection was confirmed by PCR. Baseline characteristics, critical illness data and clinical outcomes were recorded and analyzed. RESULTS Seventy-two RT-PCR-confirmed COVID-19 patients with ICU admission were included during the study period and 48 (66.7%) patients required mechanical ventilation (MV). Overall, in-hospital mortality was 19.4%. Twenty-one (29.2%) patients developed CMV infection. Patients with CMV infection had a higher likelihood of diabetes, higher lactate dehydrogenase and lactate levels, and higher proportions of MV, anticoagulant, and steroid use. Patients with CMV infection were associated with longer duration of SARS-CoV-2 shedding, longer ICU and hospital stay, and fewer ventilator-free days. The independent risk factor for development of CMV infection was a higher accumulative steroid dose. CONCLUSION CMV infection adversely impacted the outcomes of critical COVID-19 patients, resulting in longer ICU stays, longer mechanical ventilation uses and prolonged shedding of SARS-CoV-2.
Collapse
Affiliation(s)
- Hsiao-Chin Shen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
- Department of Medical Education, Taipei Veterans General Hospital, Taipei
| | - Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
| | - Chuan-Yen Sun
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
| | - Jhong-Ru Huang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
| | - Wei-Chih Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
| | - Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital, Institute of Emergency and Critical Care Medicine and School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, #201, Sec. Shih-Pai Road, Taipei 11217
| |
Collapse
|
11
|
Maslinska M, Kostyra-Grabczak K. The role of virus infections in Sjögren’s syndrome. Front Immunol 2022; 13:823659. [PMID: 36148238 PMCID: PMC9488556 DOI: 10.3389/fimmu.2022.823659] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
Primary Sjögren’s syndrome (pSS) is an autoimmune disease with a clinical picture of not only mainly exocrine gland involvement, with dryness symptoms, but also internal organ and systems involvement. The epithelial damage and releasing of antigens, which, in some circumstances, become autoantigens, underlay the pathogenesis of pSS. The activation of autoimmune processes in pSS leads to the hyperactivation of B cells with autoantibody production and other immunological phenomena such as hypergammaglobulinemia, production of cryoglobulins, or formation of extra-nodal lymphoid tissue. Among the risk factors for the development of this disease are viral infections, which themselves can activate autoimmune reactions and influence the host’s immune response. It is known that viruses, through various mechanisms, can influence the immune system and initiate autoimmune reactions. These mechanisms include molecular mimicry, bystander activation, production of superantigens—proteins encoded by viruses—or a programming to produce viral cytokines similar to host cytokines such as, e.g., interleukin-10. Of particular importance for pSS are viruses which not only, as expected, activate the interferon pathway but also play a particular role, directly or indirectly, in B cell activation or present tropism to organs also targeted in the course of pSS. This article is an attempt to present the current knowledge of the influence specific viruses have on the development and course of pSS.
Collapse
|
12
|
Taherifard E, Mortazavi R, Mokhtari M, Taherifard A, Kiani Salmi S, Taherifard E. Cytomegalovirus gastritis in a patient with severe acute respiratory syndrome coronavirus 2 infection: A case report and literature review. Respir Med Case Rep 2022; 37:101644. [PMID: 35392550 PMCID: PMC8975752 DOI: 10.1016/j.rmcr.2022.101644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/17/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022] Open
Abstract
In this study, we reported a previously immunocompetent patient who developed cytomegalovirus-induced gastric ulcers after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. A 33-year-old man was referred to our center with complaints of persistent dysphagia and odynophagia, and epigastric pain and discomfort after ingesting solids or liquids, a few days after his hospital discharge following admission to treat coronavirus disease 2019 (Covid-19). Endoscopy revealed inflammation and a whitish exudate in the esophagus, and multiple large active ulcers in the stomach. Histopathological and immunohistochemical findings were strongly suggestive of cytomegalovirus infection.
Collapse
Affiliation(s)
- Ehsan Taherifard
- Internal Medicine Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roozbeh Mortazavi
- Internal Medicine Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maral Mokhtari
- Pathology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Taherifard
- Radiology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sima Kiani Salmi
- Radiology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Erfan Taherifard
- Internal Medicine Department, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Royo-Rubio E, Martín-Cañadilla V, Rusnati M, Milanesi M, Lozano-Cruz T, Gómez R, Jiménez JL, Muñoz-Fernández MÁ. Prevention of Herpesviridae Infections by Cationic PEGylated Carbosilane Dendrimers. Pharmaceutics 2022; 14:pharmaceutics14030536. [PMID: 35335912 PMCID: PMC8950866 DOI: 10.3390/pharmaceutics14030536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/28/2022] Open
Abstract
Infections caused by viruses from the Herpesviridae family produce some of the most prevalent transmitted diseases in the world, constituting a serious global public health issue. Some of the virus properties such as latency and the appearance of resistance to antiviral treatments complicate the development of effective therapies capable of facing the infection. In this context, dendrimers present themselves as promising alternatives to current treatments. In this study, we propose the use of PEGylated cationic carbosilane dendrimers as inhibitors of herpes simplex virus 2 (HSV-2) and human cytomegalovirus (HCMV)infections. Studies of mitochondrial toxicity, membrane integrity, internalization and viral infection inhibition indicated that G2-SN15-PEG, G3-SN31-PEG, G2-SN15-PEG fluorescein isothiocyanate (FITC) labeled and G3-SN31-PEG-FITC dendrimers are valid candidates to target HSV-2 and HCMV infections since they are biocompatible, can be effectively internalized and are able to significantly inhibit both infections. Later studies (including viral inactivation, binding inhibition, heparan sulphate proteoglycans (HSPG)binding and surface plasmon resonance assays) confirmed that inhibition takes place at first infection stages. More precisely, these studies established that their attachment to cell membrane heparan sulphate proteoglycans impede the interaction between viral glycoproteins and these cell receptors, thus preventing infection. Altogether, our research confirmed the high capacity of these PEGylated carbosilane dendrimers to prevent HSV-2 and HCMV infections, making them valid candidates as antiviral agents against Herpesviridae infections.
Collapse
Affiliation(s)
- Elena Royo-Rubio
- Laboratorio InmunoBiologia Molecular, Instituto Investigacion Sanitaria Gregorio Maranon (IiSGM), Hospital General Universitario Gregorio Maranon (HGUGM), 28009 Madrid, Spain; (E.R.-R.); (V.M.-C.)
- Plataforma de Laboratorio (Inmunologia), HGUGM, IiSGM, Spanish HIV HGM BioBank, 28009 Madrid, Spain;
| | - Vanessa Martín-Cañadilla
- Laboratorio InmunoBiologia Molecular, Instituto Investigacion Sanitaria Gregorio Maranon (IiSGM), Hospital General Universitario Gregorio Maranon (HGUGM), 28009 Madrid, Spain; (E.R.-R.); (V.M.-C.)
- Plataforma de Laboratorio (Inmunologia), HGUGM, IiSGM, Spanish HIV HGM BioBank, 28009 Madrid, Spain;
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.R.); (M.M.)
| | - Maria Milanesi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.R.); (M.M.)
| | - Tania Lozano-Cruz
- Departmento Quimica Organica y Quimica Inorganica, Instituto de Investigacion Quimica “Andres M. del Rio″ (IQAR), Universidad de Alcalá (IRYCIS), Campus Universitario, 28871 Madrid, Spain; (T.L.-C.); (R.G.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rafael Gómez
- Departmento Quimica Organica y Quimica Inorganica, Instituto de Investigacion Quimica “Andres M. del Rio″ (IQAR), Universidad de Alcalá (IRYCIS), Campus Universitario, 28871 Madrid, Spain; (T.L.-C.); (R.G.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - José Luís Jiménez
- Plataforma de Laboratorio (Inmunologia), HGUGM, IiSGM, Spanish HIV HGM BioBank, 28009 Madrid, Spain;
| | - Maria Ángeles Muñoz-Fernández
- Laboratorio InmunoBiologia Molecular, Instituto Investigacion Sanitaria Gregorio Maranon (IiSGM), Hospital General Universitario Gregorio Maranon (HGUGM), 28009 Madrid, Spain; (E.R.-R.); (V.M.-C.)
- Correspondence: or
| |
Collapse
|
14
|
Jena A, Mishra S, Singh AK, Sekar A, Sharma V. Cytomegalovirus in ulcerative colitis: an evidence-based approach to diagnosis and treatment. Expert Rev Gastroenterol Hepatol 2022; 16:109-120. [PMID: 35057693 DOI: 10.1080/17474124.2022.2032662] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The detection of cytomegalovirus (CMV) in the setting of inflammatory bowel disease often creates confusion whether CMV is a 'bystander' or 'disease.' AREAS COVERED This review discusses the clinical conundrum of CMV in ulcerative colitis, approach to discriminate infection from disease, and therapeutic considerations (immunosuppressive and anti-CMV treatment). CMV disease should be considered in corticosteroid refractory- dependent and thiopurine refractory disease. Endoscopy may reveal deep punched out ulcers, irregular ulcers, or cobble-stoning. The diagnosis rests on the presence and abundance of viral inclusion bodies on hematoxylin and eosin stain, positive immunohistochemistry, and/or positive tissue polymerase chain reaction. CMV disease is associated with worse outcomes including increased colectomy rates. EXPERT OPINION The timing and duration of antiviral drugs in CMV disease is debatable but depends on the load of CMV in tissue. In high-grade infection, CMV needs to be treated while increasing immunosuppression may work in the setting of low-grade infection. Ganciclovir is the drug of choice for treatment of CMV disease. Tumor necrosis factor inhibitors may be useful for treating underlying disease activity in the setting of CMV. Other emerging therapies include fecal microbiota transplantation. Randomized studies are necessary to define the best timing and duration of anti-CMV therapy.
Collapse
Affiliation(s)
- Anuraag Jena
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shubhra Mishra
- Department of Gastroenterology, AIG Hospitals, Hyderabad, India
| | - Anupam Kumar Singh
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aravind Sekar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishal Sharma
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
15
|
Gröschel M, Voigt S, Schwitzer S, Ernst A, Basta D. Cytomegalovirus Seropositivity as a Potential Risk Factor for Increased Noise Trauma Susceptibility. Noise Health 2022; 24:1-6. [PMID: 35645133 PMCID: PMC9239143 DOI: 10.4103/nah.nah_4_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
CONTEXT Cytomegalovirus (CMV) represents the leading congenital viral infection in humans. Although congenital CMV due to vertically transmitted infections is the main cause of CMV-related diseases, adult CMV infections might still be of clinical significance. It is still discussed how far CMV seropositivity, due to horizontal infection in immunocompetent adults, is able to induce significant dysfunction. The present study investigates in how far CMV seropositivity is an additional risk factor for an increasing susceptibility to sensorineural hearing loss induced by acoustic injury during adulthood in a guinea pig CMV (GPCMV) model of noise-induced hearing loss (NIHL). METHODS Two groups (GPCMV seropositive vs. seronegative) of normal hearing adult guinea pigs were exposed to a broadband noise (5-20 kHz) for 2 hours at 115 dB sound pressure level. Frequency-specific auditory brainstem response recordings for determination of auditory threshold shift were carried out and the number of missing outer hair cells was counted 2 weeks after the noise exposure. RESULTS The data show a slightly increased shift in auditory thresholds in seropositive animals compared to the seronegative control group in response to noise trauma. However, the observed difference was significant at least at high frequencies. The differences in threshold shift are not correlated with outer hair cell loss between the experimental groups. CONCLUSION The results point to potential additional pathologies in a guinea pig NIHL model in correlation to GPCMV seropositivity, which should be taken into account when assessing risks of latent/reactivated CMV infection. Due to the relatively slight effect in the present data, the aim of future studies should be a more detailed consideration (e.g., larger sample size) and to localize possible target structures as well as the significance of the infection route.
Collapse
Affiliation(s)
- Moritz Gröschel
- Department of Otolaryngology at UKB, University of Berlin, Charité Medical School, Berlin, Germany,Address for correspondence: Dr Moritz Gröschel, Department of Otolaryngology, Unfallkrankenhaus Berlin, Warener Str 7, 12683 Berlin, Germany. E-mail:
| | - Stefan Voigt
- Department of Otolaryngology at UKB, University of Berlin, Charité Medical School, Berlin, Germany
| | - Susanne Schwitzer
- Department of Otolaryngology at UKB, University of Berlin, Charité Medical School, Berlin, Germany
| | - Arne Ernst
- Department of Otolaryngology at UKB, University of Berlin, Charité Medical School, Berlin, Germany
| | - Dietmar Basta
- Department of Otolaryngology at UKB, University of Berlin, Charité Medical School, Berlin, Germany
| |
Collapse
|
16
|
Beyaz MO, Bozkurt E, Onalan MA, Oztas DM, Meric M, Ulukan MO, Coskun U, Ugurlucan M. Evaluation of chronic inflammation in the aetiology of venous insufficiency by investigating cytomegalovirus DNA. Arch Med Sci 2022; 18:129-132. [PMID: 35154533 PMCID: PMC8826984 DOI: 10.5114/aoms.2019.85232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/08/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Lower extremity venous insufficiency is a significant health problem with economic and sociological consequences, lowering the quality of life, and sometimes leading to serious complications. The aim of this study is to evaluate the cytomegalovirus (CMV) effect on chronic inflammation in the aetiology of chronic venous insufficiency. MATERIAL AND METHODS Between November 2017 and August 2018, 468 patients who underwent radio-frequency ablation therapy and phlebectomy were included in the study. PCR analyses for CMV DNA were performed on the venous tissue samples. Patients with post-thrombotic syndrome were excluded from the study. After ethical approval, the relationship between the presence of CMV DNA, gender, body mass index, and bilaterality of chronic venous insufficiency were investigated. RESULTS When the relationship between CMV DNA and gender or body mass index was examined, a significant relationship was not detected. But when the patients with bilateral chronic venous insufficiency and patients with unilateral chronic venous insufficiency were compared regarding CMV DNA positivity, the patients with bilateral chronic venous insufficiency had significantly higher CMV DNA positivity (p = 0.002). Also, the incidence of venous ulcers in the CMV DNA exposed group was significantly higher. CONCLUSIONS In the literature there are many studies showing that CMV triggers atherosclerosis, but there is no study in which CMV directly produces chronic venous insufficiency. The high rates of positivity suggest that CMV, which is the basis of chronic inflammation, may be a significant factor in the aetiology of chronic venous insufficiency.
Collapse
Affiliation(s)
- Metin O Beyaz
- Cardiovascular Surgery Clinic, Umraniye Education and Research Hospital, Istanbul, Turkey
| | - Emre Bozkurt
- General Surgery Clinic, Gumushane Kelkit Hospital, Gumushane, Turkey
| | - Mehmet A Onalan
- Department of Cardiovascular Surgery, Medical Faculty, Acibadem University, Istanbul, Turkey
| | - Didem M Oztas
- Cardiovascular Surgery Clinic, Bagcilar Education and Research Hospital, Istanbul, Turkey
| | - Mert Meric
- Department of Cardiovascular Surgery, Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Mustafa Ozer Ulukan
- Department of Cardiovascular Surgery, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Ugur Coskun
- Cardiology Department, School of Medicine Karakum, Kyrenia University, Kyrenia, Cyprus
| | - Murat Ugurlucan
- Department of Cardiovascular Surgery, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
17
|
Heldman MR, Ma J, Gauthier J, O’Hara RA, Cowan AJ, Yoke LM, So L, Gulleen E, Duke ER, Liu C, Turtle CJ, Hill JA. CMV and HSV Pneumonia After Immunosuppressive Agents for Treatment of Cytokine Release Syndrome Due to Chimeric Antigen Receptor-modified T (CAR-T)-Cell Immunotherapy. J Immunother 2021; 44:351-354. [PMID: 34369454 PMCID: PMC8497421 DOI: 10.1097/cji.0000000000000388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/15/2021] [Indexed: 11/25/2022]
Abstract
Pneumonia due to cytomegalovirus and herpes simplex virus-1 caused substantial morbidity after hematopoietic cell transplantation before the institution of preventative approaches. End-organ disease from herpesviruses is poorly described after chimeric antigen receptor-modified T-cell immunotherapy. We report 2 cases of cytomegalovirus pneumonia and 1 case of herpes simplex virus-1 gingivostomatitis, esophagitis, and pneumonia after chimeric antigen receptor-modified T-cell immunotherapy for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Madeleine R. Heldman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jimmy Ma
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
| | - Jordan Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Riley A. O’Hara
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Andrew J. Cowan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Leah M. Yoke
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lisa So
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Elizabeth Gulleen
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Elizabeth R. Duke
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Catherine Liu
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Cameron J. Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Joshua A. Hill
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
18
|
Yamamoto Y, Shiroyama T, Hirata H, Kuge T, Matsumoto K, Yoneda M, Yamamoto M, Uchiyama A, Takeda Y, Kumanogoh A. Prolonged corticosteroid therapy and cytomegalovirus infection in patients with severe COVID-19. J Med Virol 2021; 94:1067-1073. [PMID: 34708883 PMCID: PMC8661974 DOI: 10.1002/jmv.27421] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/25/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023]
Abstract
Systemic corticosteroid therapy is frequently used to treat coronavirus disease 2019 (COVID‐19). However, its maximum duration without secondary infections remains unclear. We aimed to evaluate the utility of monitoring cytomegalovirus (CMV) infection in patients with COVID‐19 and estimate the maximum duration of systemic corticosteroid therapy without secondary infections. We included 59 patients with severe COVID‐19 without CMV infection on admission to the intensive care unit (ICU). All patients received systemic corticosteroid therapy under invasive mechanical ventilation, with examination for plasma CMV‐deoxyribonucleic acid (DNA) levels during the ICU stay. We analyzed the correlations among patient characteristics, CMV infection, diseases, and patient mortality. CMV infections were newly identified in 15 (25.4%) patients; moreover, anti‐CMV treatment was administered to six (10.2%) patients during the ICU stay. Four (6.8%) patients had secondary infection‐related mortality. The cumulative incidences of CMV infection and anti‐CMV treatment during the ICU stay were 26.8% (95% confidence interval [CI], 15.8%–39.0%) and 12.3% (95% CI, 4.8%–23.4%), respectively. Furthermore, the median duration of systemic corticosteroid therapy without CMV infection was 15 days (95% CI, 13–16 days). The presence of CMV infection was associated with mortality during the ICU stay (p = 0.003). Monitoring plasma CMV‐DNA levels could facilitate the detection of secondary CMV infection due to prolonged systemic corticosteroid therapy. The duration of systemic corticosteroid therapy for COVID‐19 should be limited.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayuki Shiroyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoki Kuge
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kinnosuke Matsumoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Midori Yoneda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Yamamoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akinori Uchiyama
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, Suita, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.,Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
19
|
Kostoff RN, Briggs MB, Kanduc D, Shores DR, Kovatsi L, Vardavas AI, Porter AL. Common contributing factors to COVID-19 and inflammatory bowel disease. Toxicol Rep 2021; 8:1616-1637. [PMID: 34485092 PMCID: PMC8406546 DOI: 10.1016/j.toxrep.2021.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/17/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022] Open
Abstract
The devastating complications of coronavirus disease 2019 (COVID-19) result from an individual's dysfunctional immune response following the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple toxic stressors and behaviors contribute to underlying immune system dysfunction. SARS-CoV-2 exploits the dysfunctional immune system to trigger a chain of events ultimately leading to COVID-19. We have previously identified many contributing factors (CFs) (representing toxic exposure, lifestyle factors and psychosocial stressors) common to myriad chronic diseases. We hypothesized significant overlap between CFs associated with COVID-19 and inflammatory bowel disease (IBD), because of the strong role immune dysfunction plays in each disease. A streamlined dot-product approach was used to identify potential CFs to COVID-19 and IBD. Of the fifty CFs to COVID-19 that were validated for demonstration purposes, approximately half had direct impact on COVID-19 (the CF and COVID-19 were mentioned in the same record; i.e., CF---→COVID-19), and the other half had indirect impact. The nascent character of the COVID-19 core literature (∼ one year old) did not allow sufficient time for the direct impacts of many CFs on COVID-19 to be identified. Therefore, an immune system dysfunction (ID) literature directly related to the COVID-19 core literature was used to augment the COVID-19 core literature and provide the remaining CFs that impacted COVID-19 indirectly (i.e., CF---→immune system dysfunction---→COVID-19). Approximately 13000 potential CFs for myriad diseases (obtained from government and university toxic substance lists) served as the starting point for the dot-product identification process. These phrases were intersected (dot-product) with phrases extracted from a PubMed-derived IBD core literature, a nascent COVID-19 core literature, and the COVID-19-related immune system dysfunction (ID) core literature to identify common ID/COVID-19 and IBD CFs. Approximately 3000 potential CFs common to both ID and IBD, almost 2300 potential CFs common to ID and COVID-19, and over 1900 potential CFs common to IBD and COVID-19 were identified. As proof of concept, we validated fifty of these ∼3000 overlapping ID/IBD candidate CFs with biologic plausibility. We further validated 24 of the fifty as common CFs in the IBD and nascent COVID-19 core literatures. This significant finding demonstrated that the CFs indirectly related to COVID-19 -- identified with use of the immune system dysfunction literature -- are strong candidates to emerge eventually as CFs directly related to COVID-19. As discussed in the main text, many more CFs common to all these core literatures could be identified and validated. ID and IBD share many common risk/contributing factors, including behaviors and toxic exposures that impair immune function. A key component to immune system health is removal of those factors that contribute to immune system dysfunction in the first place. This requires a paradigm shift from traditional Western medicine, which often focuses on treatment, rather than prevention.
Collapse
Affiliation(s)
- Ronald Neil Kostoff
- School of Public Policy, Georgia Institute of Technology, Gainesville, VA, 20155, United States
| | | | - Darja Kanduc
- Dept. of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Via Orabona 4, Bari, 70125, Italy
| | - Darla Roye Shores
- Department of Pediatrics, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54124, Greece
| | - Alexander I. Vardavas
- Laboratory of Toxicology & Forensic Sciences, Faculty of Medicine, University of Crete, Greece
| | - Alan L. Porter
- R&D, Search Technology, Inc., Peachtree Corners, GA, 30092, United States
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA, 30332, United States
| |
Collapse
|
20
|
Evaluation of Role of Herpes Simplex Virus Types 1 and 2 and Cytomegalovirus in Alzheimer's Disease. MEDICAL LABORATORY JOURNAL 2021. [DOI: 10.52547/mlj.15.4.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
21
|
Jorgenson MR, Marka N, Leverson GE, Smith JA, Odorico JS. Valganciclovir prophylaxis extension from 3 to 6 months in high-risk pancreas-transplant recipients does not impact incidence of cytomegalovirus infection at 12 months. Clin Transplant 2021; 35:e14379. [PMID: 34075624 DOI: 10.1111/ctr.14379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/11/2021] [Accepted: 05/28/2021] [Indexed: 01/10/2023]
Abstract
PROBLEM Incidence and impact of CMV infection in pancreas-transplant recipients (PTRs) in the valganciclovir prophylaxis era has not been completely elucidated. METHODS Adult D+/R- PTRs were divided into a current era (1/1/2011-12/31/17; 6-month PPX) and a historic era (1/1/2003-12/31/09; 3-month PPX). PRIMARY OBJECTIVE effect of prophylaxis extension on the incidence of CMV infection. SECONDARY OBJECTIVE impact of extension on valganciclovir-related toxicity (leukopenia) and transplant outcomes. RESULTS There were 177 D+/R- PTRs in the study period (historic:98, current:79). Prophylaxis extension resulted in significant reduction of CMV infection from 25.4% to 10.9% at 6 months, (57% reduction, p = .021). However, 1-year rates of CMV infection (historic:31% vs current:36%) and end-organ disease (historic:7.7% vs current:6.9%) were not different (p = .93). Prophylaxis extension significantly increased leukopenia (white blood cell count<3 K/uL) at 6 months (historic:9.5% vs current:28.6%, p = .018). On multivariable analysis prophylaxis extension was not associated with reduced rates of CMV infection (p = .99) or CMV end-organ disease (p = .3). Additionally, there was no significant difference in rejection (p = .2), graft survival (p = .08), death-censored graft survival(p = .07) or patient survival (p = .6). CONCLUSIONS Prophylaxis extension in D+/R- PTRs appears to delay time to first CMV but not reduce overall incidence. These findings suggest a hybrid approach, incorporating antiviral withdrawal and protocolized monitoring, may be needed to improve CMV-related outcomes.
Collapse
Affiliation(s)
- Margaret R Jorgenson
- Department of Pharmacy, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Nicholas Marka
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Glen E Leverson
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Jeannina A Smith
- Department of Medicine, Division of Infectious Diseases, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Jon S Odorico
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
22
|
Cytomegalovirus Latency Exacerbated small-for-size Liver Graft Injury through Activation of CCL19/CCR7 in Hepatic Stellate Cells. Transplantation 2021; 106:519-530. [PMID: 34156186 DOI: 10.1097/tp.0000000000003846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The interplay between cytomegalovirus (CMV) latency and graft malfunction after living donor liver transplantation (LDLT) remains poorly defined due to the complexity of clinical confounding factors. Here, we aimed to investigate the effects of CMV latency on small-for-size graft injury and to get further insight on the pathogenic role of hepatic stellate cells (HSCs) in this process. METHODS Rat orthotopic liver transplantation with small-for-size grafts was performed in a CMV latent model developed in immunocompetent Sprague Dawley (SD) rats using Priscott strain. Post-transplant graft injury including hepatocyte damage, stellate cell activation and fibrogenesis were evaluated. Differential gene expression of HSCs in response to CMV latency was screened by cDNA microarray. Clinical validation was further conducted in human biopsies. RESULTS CMV latency aggravated hepatocyte apoptosis/necrosis in the early phase, enhanced HSC expansion and graft fibrosis during the middle-late phase in small-for-size liver grafts of the rat model. cDNA microarray mining revealed CCL19/CCR7 as one of the most noteworthy pathways bridging HSC activation and liver graft injury in the presence of CMV latency. Together with CCL19 upregulation, coherent overexpression of CCR7 in accumulated HSCs was confirmed in both rat and human CMV latent recipients. Moreover, addition of CCL19 in vitro promoted HSC migration by increasing the level of matrix metalloproteinase-2 (MMP2). CONCLUSION Our data demonstrated that CMV latency aggravated early/late phase liver graft damage and fibrogenesis via CCL19/CCR7/HSCs axis. Blockade of CMV latency-related stellate cell activation may shed light on the strategy of graft protection clinically.
Collapse
|
23
|
Maksimowicz-McKinnon K, Zhou J, Hudy J, Hegab S, McKinnon JE. Subclinical CMV viremia is associated with increased nosocomial infections and prolonged hospitalization in patients with systemic autoimmune diseases. J Clin Virol 2021; 140:104849. [PMID: 34023574 DOI: 10.1016/j.jcv.2021.104849] [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: 11/18/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Subclinical cytomegalovirus (CMV) viremia has been associated with other infections, prolonged hospitalization, and mortality in select immunosuppressed populations. We examined the incidence and outcomes of subclinical CMV viremia in hospitalized patients with systemic autoimmune diseases (AD) [systemic lupus erythematosus (SLE) or anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV)] using a highly sensitive CMV assay. METHODS Prospectively collected samples were obtained from AD hospitalized patients at study entry with a second sample collected 1 week later or at hospital discharge. Controls included age- and gender- matched inpatients without AD and outpatients with AD. All samples were tested in batch using the Abbott RealTime CMV for investigational use assay (RT assay), with a LLOD (LLOQ) at 21 IU/mL (32 IU/mL). RESULTS Twenty-three inpatients (10 SLE, 8 AAV, 5 controls), and 31 outpatient controls were recruited. Subclinical CMV viremia was found in 61% (11/18) of inpatient AD subjects, 3% (1/31) of outpatient AD subjects, and in none of the five inpatient controls (p < 0.001). CMV viremia was associated with increased median length of ICU stay (13 vs. 4 days, p = 0.033), hospital stay (17 vs. 9 days, p = 0.014) and increased nosocomial infections (7 vs. 1, p = 0.007). CMV viremia was not associated with overall severity of illness nor with disease-specific activity or damage. CONCLUSION Over one-half of hospitalized AD patients in our cohort had detectable CMV viremia, which was associated with increased length of hospital stay and nosocomial infections. These data suggest that further study of the immunomodulatory effects of subclinical CMV viremia in AD is warranted.
Collapse
Affiliation(s)
| | - Junying Zhou
- Henry Ford Hospital, 2799 W. Grand Blvd, Detroit, MI, USA.
| | - Jenna Hudy
- Henry Ford Hospital, 3031 W. Grand Blvd. Suite 800, Detroit, MI, USA.
| | - Sara Hegab
- Henry Ford Hospital, 2799 W. Grand Blvd, Detroit, MI, USA.
| | - John E McKinnon
- Medicine, Wayne State University, Henry Ford Hospital, 2799 W. Grand Blvd, Detroit, MI, USA.
| |
Collapse
|
24
|
Jorgenson MR, Wong C, Descourouez JL, Saddler CM, Smith JA, Mandelbrot DA. Conversion from cytomegalovirus universal prophylaxis with valganciclovir to the preemptive monitoring approach to manage leukopenia after kidney or pancreas transplantation. Transpl Infect Dis 2021; 23:e13617. [PMID: 33866643 DOI: 10.1111/tid.13617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/05/2021] [Accepted: 04/05/2021] [Indexed: 01/31/2023]
Abstract
PURPOSE In clinical practice, conversion from universal cytomegalovirus prophylaxis (CMV PPX) with valganciclovir (VGC) to targeted therapy (preemptive monitoring, PEM) is often pursued in the setting of leukopenia. It is unknown if this is an effective strategy. METHODS Adult patients receiving a kidney and/or pancreas transplant were included if converted from PPX to PEM between 9/1/19 and 3/1/20 due to leukopenia. A positive CMV viral load (VL) was defined as CMV PCR greater than the lower limit of quantification (LLOQ) based on local lab testing. A CMV VL of >500 IU/mL was chosen as the preemptive treatment (PET) threshold. Primary objective was to describe the impact of conversion on resolution of leukopenia. Secondary objectives were to assess PEM associated outcomes. RESULTS There were 49 patients converted from PPX to PEM due to leukopenia in the study period; 88% were KTRs and 96% received lymphocyte-depleting induction; 84% were seropositive at transplant (R+) and 16% were high-risk (D+/R-). Mean WBC at time of enrollment was 1.4 ± 0.4. After PEM conversion, WBC recovered to >3 in 87.8% of the population in a mean of 26.8 ± 24.5 days. Immunosuppression was modified in 96% of the population and GCSF was required in 46.9% of patients. CMV viremia occurred in 36.7% of the population; 78% were KTRs and 94% were R+. Time from PEM enrollment to PET was 64 ± 34 days. Median VL at first detection was 587 IU/mL, median peak was 1920 IU/mL. Five patients (27.8%) presented with symptoms consistent with CMV syndrome, none had end organ disease. Six patients (33%) presented with a VL <500 IU/mL at first detection, but all subsequently surpassed the threshold and required PET. Mean duration of PET was 25 ± 11 days. Mean change in WBC in response to PET was -0.4 ± 1.3. Immunosuppression required further adjustment in 61% of patients. There were no deaths or graft loss due to CMV at last follow-up. CONCLUSION In kidney and pancreas transplant recipients who undergo PEM conversion due to leukopenia, withholding of VGC can improve leukopenia, but other concomitant measures are necessary. This population should be considered fairly high risk, with a threshold of treatment of first quantifiable replication. Our findings suggest lack of harm from this approach but highlight the importance of close monitoring to prevent symptomatic replication. Larger studies with longer follow-up are needed to better evaluate the impact of PEM conversion on late-onset CMV and patient and graft outcomes.
Collapse
Affiliation(s)
- Margaret R Jorgenson
- Department of Pharmacy, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Cynthia Wong
- Department of Pharmacy, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Jillian L Descourouez
- Department of Pharmacy, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Christopher M Saddler
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Jeannina A Smith
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Didier A Mandelbrot
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| |
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
|
Kuo CW, Wang SY, Tsai HP, Su PL, Cia CT, Lai CH, Chen CW, Shieh CC, Lin SH. Invasive pulmonary aspergillosis is associated with cytomegalovirus viremia in critically ill patients - A retrospective cohort study. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 55:291-299. [PMID: 33840605 DOI: 10.1016/j.jmii.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/23/2021] [Accepted: 03/08/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND/PURPOSE Cytomegalovirus (CMV) viremia is associated with a higher mortality rate and prolonged intensive care unit (ICU) stay for critically ill patients. CMV infection causes transient but substantial immunosuppression for transplant recipients, increasing risk of fungal infection. The association between CMV viremia and invasive pulmonary aspergillosis (IPA) for critically ill patients is still unknown. METHODS We retrospectively analyzed patients received bronchoalveolar lavage (BAL), galactomannan test, influenza survey and blood CMV viral load test in ICUs of a university hospital between April 2017 and May 2020. Independent risks for IPA were analyzed by multivariable logistic regression. RESULTS A total of 136 patients were included. Twenty-one patients had IPA, 48 patients had CMV viremia and 22 patients had influenza. In a multivariable logistic regression model, patients with CMV viremia or influenza had higher IPA risk (adjusted odds ratio, 3.98 and 8.72; 95% CI, 1.26-12.60 and 2.64-28.82; p value = 0.019 and <0.001, respectively.). Patients with detectable CMV in BAL fluid did not have higher IPA risk (crude odds ratio, 0.95; 95% CI, 0.33-2.79; p value = 0.933). After stratifying patients by CMV viral load, the IPA risk is higher for patients with higher viral loads. There is an additive synergistic effect on IPA risk between CMV viremia and influenza infection. CONCLUSION For critically ill patients, CMV viremia is an independent risk factor of IPA. Patients with higher blood CMV viral loads have a higher risk of IPA. CMV viremia and influenza have an additive synergistic effect for IPA risk in critically ill patients.
Collapse
Affiliation(s)
- Chin-Wei Kuo
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Critical Care Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Yuan Wang
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lan Su
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cong-Tat Cia
- Division of Critical Care Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center for Infection Control, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Han Lai
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chang-Wen Chen
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Critical Care Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Biostatistics Consulting Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
27
|
Potential Impact of Human Cytomegalovirus Infection on Immunity to Ovarian Tumours and Cancer Progression. Biomedicines 2021; 9:biomedicines9040351. [PMID: 33808294 PMCID: PMC8065684 DOI: 10.3390/biomedicines9040351] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common, and life-threatening gynaecological cancer affecting females. Almost 75% of all OC cases are diagnosed at late stages, where the 5-year survival rate is less than 30%. The aetiology of the disease is still unclear, and there are currently no screening method nor effective treatment strategies for the advanced disease. A growing body of evidence shows that human cytomegalovirus (HCMV) infecting more than 50% of the world population, may play a role in inducing carcinogenesis through its immunomodulatory activities. In healthy subjects, the primary HCMV infection is essentially asymptomatic. The virus then establishes a life-long chronic latency primarily in the hematopoietic progenitor cells in the bone marrow, with periodic reactivation from latency that is often characterized by high levels of circulating pro-inflammatory cytokines. Currently, infection-induced chronic inflammation is considered as an essential process for OC progression and metastasis. In line with this observation, few recent studies have identified high expressions of HCMV proteins on OC tissue biopsies that were associated with poor survival outcomes. Active HCMV infection in the OC tumour microenvironment may thus directly contribute to OC progression. In this review, we highlight the potential impact of HCMV infection-induced immunomodulatory effects on host immune responses to OC that may promote OC progression.
Collapse
|
28
|
Gozzi-Silva SC, Benard G, Alberca RW, Yendo TM, Teixeira FME, Oliveira LDM, Beserra DR, Pietrobon AJ, de Oliveira EA, Branco ACCC, Andrade MMDS, Fernandes IG, Pereira NZ, Ramos YÁL, Lima JC, Provenci B, Mangini S, Duarte AJDS, Sato MN. SARS-CoV-2 Infection and CMV Dissemination in Transplant Recipients as a Treatment for Chagas Cardiomyopathy: A Case Report. Trop Med Infect Dis 2021; 6:22. [PMID: 33579042 PMCID: PMC7985779 DOI: 10.3390/tropicalmed6010022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 has infected over 90 million people worldwide, therefore it is considered a pandemic. SARS-CoV-2 infection can lead to severe pneumonia, acute respiratory distress syndrome (ARDS), septic shock, and/or organ failure. Individuals receiving a heart transplantation (HT) may be at higher risk of adverse outcomes attributable to COVID-19 due to immunosuppressives, as well as concomitant infections that may also influence the prognoses. Herein, we describe the first report of two cases of HT recipients with concomitant infections by SARS-CoV-2, Trypanosoma cruzi, and cytomegalovirus (CMV) dissemination, from the first day of hospitalization due to COVID-19 in the intensive care unit (ICU) until the death of the patients.
Collapse
Affiliation(s)
- Sarah Cristina Gozzi-Silva
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| | - Gil Benard
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Ricardo Wesley Alberca
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Tatiana Mina Yendo
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Franciane Mouradian Emidio Teixeira
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| | - Luana de Mendonça Oliveira
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Danielle Rosa Beserra
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Anna Julia Pietrobon
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| | - Emily Araujo de Oliveira
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| | - Milena Mary de Souza Andrade
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Iara Grigoletto Fernandes
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Nátalli Zanete Pereira
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Yasmim Álefe Leuzzi Ramos
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Julia Cataldo Lima
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Bruna Provenci
- Instituto do Coração (Incor), Hospital das Clínicas, School of Medicine of University of São Paulo (HCFMUSP), 05403-900 São Paulo, Brazil; (B.P.); (S.M.)
| | - Sandrigo Mangini
- Instituto do Coração (Incor), Hospital das Clínicas, School of Medicine of University of São Paulo (HCFMUSP), 05403-900 São Paulo, Brazil; (B.P.); (S.M.)
| | - Alberto José da Silva Duarte
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Institute of Tropical Medicine of School of Medicine of São Paulo (FMUSP), 05403-000 São Paulo, Brazil; (G.B.); (R.W.A.); (T.M.Y.); (F.M.E.T.); (L.d.M.O.); (D.R.B.); (A.J.P.); (E.A.d.O.); (A.C.C.C.B.); (M.M.d.S.A.); (I.G.F.); (N.Z.P.); (Y.Á.L.R.); (J.C.L.); (A.J.d.S.D.); (M.N.S.)
- Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil
| |
Collapse
|
29
|
Gugliesi F, Pasquero S, Griffante G, Scutera S, Albano C, Pacheco SFC, Riva G, Dell’Oste V, Biolatti M. Human Cytomegalovirus and Autoimmune Diseases: Where Are We? Viruses 2021; 13:260. [PMID: 33567734 PMCID: PMC7914970 DOI: 10.3390/v13020260] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous double-stranded DNA virus belonging to the β-subgroup of the herpesvirus family. After the initial infection, the virus establishes latency in poorly differentiated myeloid precursors from where it can reactivate at later times to cause recurrences. In immunocompetent subjects, primary HCMV infection is usually asymptomatic, while in immunocompromised patients, HCMV infection can lead to severe, life-threatening diseases, whose clinical severity parallels the degree of immunosuppression. The existence of a strict interplay between HCMV and the immune system has led many to hypothesize that HCMV could also be involved in autoimmune diseases (ADs). Indeed, signs of active viral infection were later found in a variety of different ADs, such as rheumatological, neurological, enteric disorders, and metabolic diseases. In addition, HCMV infection has been frequently linked to increased production of autoantibodies, which play a driving role in AD progression, as observed in systemic lupus erythematosus (SLE) patients. Documented mechanisms of HCMV-associated autoimmunity include molecular mimicry, inflammation, and nonspecific B-cell activation. In this review, we summarize the available literature on the various ADs arising from or exacerbating upon HCMV infection, focusing on the potential role of HCMV-mediated immune activation at disease onset.
Collapse
Affiliation(s)
- Francesca Gugliesi
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Selina Pasquero
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Gloria Griffante
- Department of Translational Medicine, Molecular Virology Unit, University of Piemonte Orientale Medical School, 28100 Novara, Italy;
| | - Sara Scutera
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Camilla Albano
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Sergio Fernando Castillo Pacheco
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Giuseppe Riva
- Otorhinolaryngology Division, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy;
| | - Valentina Dell’Oste
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| | - Matteo Biolatti
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (F.G.); (S.P.); (S.S.); (C.A.); (S.F.C.P.); (V.D.)
| |
Collapse
|
30
|
Schaenman J, Phonphok K, Spanuchart I, Duong T, Sievers TM, Lum E, Reed EF, Bunnapradist S. Early cytomegalovirus DNAemia and antiviral dose adjustment in high vs intermediate risk kidney transplant recipients. Transpl Infect Dis 2021; 23:e13457. [PMID: 32892447 PMCID: PMC7962879 DOI: 10.1111/tid.13457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/24/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection continues to negatively affect outcomes for solid organ transplant recipients, despite the advent of strategies for preemptive surveillance and prophylaxis. The impact is especially great for CMV seronegative recipients of donor seropositive organs, who typically lack the ability to control CMV infection at the time of transplantation. METHODS We reviewed episodes of CMV DNAemia in a modern cohort of kidney transplant recipients over a 3-year period at a high-volume transplant center to investigate the frequency of DNAemia during antiviral prophylaxis. RESULTS Despite receipt of antiviral prophylaxis per current guidelines, 75 cases of CMV DNAemia were observed in the first 100 days after transplantation. For high risk patients, median time to DNAemia was 75 days after transplantation, and the majority of patients had experienced dose-reduction of valganciclovir due to renal insufficiency. Review of CMV seropositive intermediate risk patients demonstrated DNAemia occurring earlier after transplantation compared with high risk patients with a median time of 64 days (P = .029). The impact of valganciclovir dose adjustment was less notable in the intermediate risk group. CONCLUSIONS Guidelines recommend beginning routine surveillance for CMV after the completion of antiviral prophylaxis. Our findings suggest that closer monitoring may be beneficial, especially for high risk patients at risk for DNAemia. Patients requiring dose adjustment of valganciclovir due to renal insufficiency may be at increased risk for CMV DNAemia. Improved methods for CMV prophylaxis and evaluation of immunologic risk for CMV DNAemia and disease are needed to improve patient outcomes after kidney transplantation.
Collapse
Affiliation(s)
- Joanna Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Korntip Phonphok
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Ittikorn Spanuchart
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Tin Duong
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Theodore M. Sievers
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Erik Lum
- Division of Nephrology, David Geffen School of Medicine, Los Angeles, CA 90095
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, CA 90095
| | | |
Collapse
|
31
|
Selby PR, Shakib S, Peake SL, Warner MS, Yeung D, Hahn U, Roberts JA. A Systematic Review of the Clinical Pharmacokinetics, Pharmacodynamics and Toxicodynamics of Ganciclovir/Valganciclovir in Allogeneic Haematopoietic Stem Cell Transplant Patients. Clin Pharmacokinet 2021; 60:727-739. [PMID: 33515202 DOI: 10.1007/s40262-020-00982-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Ganciclovir (GCV) and valganciclovir (VGCV) are the first-line agents used to prevent and treat cytomegalovirus (CMV) infection in allogeneic haematopoietic stem cell transplant (alloHCT) patients. OBJECTIVE The aim of this work was to describe available data for the clinical pharmacokinetics, pharmacodynamics and toxicodynamics of GCV and VGCV and the potential of a therapeutic drug monitoring strategy to improve outcomes in the alloHCT population. METHODS We systematically reviewed the pharmacokinetics (dose-exposure), pharmacodynamics (exposure-efficacy) and toxicodynamics (exposure-toxicity) of GCV and VGCV in alloHCT patients with CMV infection. Studies including alloHCT patients treated for CMV infection reporting the pharmacokinetics, pharmacodynamics and toxicodynamics of GCV or VGCV were searched for using the PUBMED and EMBASE databases from 1946 to 2019. Only studies involving participants > 12 years of age and available in the English language were included. RESULTS A total of 179 patients were included in the 14 studies that met the inclusion criteria, of which 6 examined GCV pharmacokinetics only, while 8 also examined GCV pharmacodynamics and toxicodynamics. Reported pharmacokinetic parameters showed considerable interpatient variability and were different from other populations, such as solid organ transplant and human immunodeficiency virus-infected patients. Only one study found a correlation between neutropenia and elevated peak and trough GCV concentrations, with no other significant pharmacodynamic and toxicodynamic relationships identified. While therapeutic drug monitoring of GCV is performed in some institutions, no association between GCV therapeutic drug monitoring and clinical outcomes was identified. CONCLUSION Further studies of the pharmacokinetics, pharmacodynamics and toxicodynamics of GCV/VGCV in alloHCT patients are required to identify a more robust therapeutic range and to subsequently quantify the potential value of therapeutic drug monitoring of GCV/VGCV in the alloHCT population.
Collapse
Affiliation(s)
- Philip Roland Selby
- School of Medicine, University of Adelaide, Adelaide, SA, Australia. .,Pharmacy Department, Royal Adelaide Hospital, Port Road, Adelaide, SA, 5000, Australia.
| | - Sepehr Shakib
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sandra L Peake
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Department of Intensive Care Medicine, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Morgyn S Warner
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Infectious Diseases Unit, The Queen Elizabeth Hospital, Adelaide, SA, Australia.,SA Pathology, Adelaide, SA, Australia
| | - David Yeung
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,SA Pathology, Adelaide, SA, Australia.,Haematology Unit, Royal Adelaide Hospital, Adelaide, SA, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Uwe Hahn
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,SA Pathology, Adelaide, SA, Australia.,Haematology Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Jason A Roberts
- Faculty of Medicine and Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, St Lucia, QLD, Australia.,Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| |
Collapse
|
32
|
Gliga S, Fiedler M, Dornieden T, Achterfeld A, Paul A, Horn PA, Herzer K, Lindemann M. Comparison of Three Cellular Assays to Predict the Course of CMV Infection in Liver Transplant Recipients. Vaccines (Basel) 2021; 9:vaccines9020088. [PMID: 33504093 PMCID: PMC7911226 DOI: 10.3390/vaccines9020088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
To estimate protection from cytomegalovirus (CMV) replication after solid organ transplantation, CMV serology has been considered insufficient and thus CMV immunity is increasingly assessed by cellular in vitro methods. We compared two commercially available IFN-γ ELISpot assays (T-Track CMV and T-SPOT.CMV) and an IFN-γ ELISA (QuantiFERON-CMV). Currently, there is no study comparing these three assays. The assays were performed in 56 liver transplant recipients at the end of antiviral prophylaxis and one month thereafter. In CMV high- or intermediate-risk patients the two ELISpot assays showed significant correlation (p < 0.0001, r > 0.6) but the correlation of the ELISpot assays with QuantiFERON-CMV was weaker. Results of both ELISpot assays were similarly predictive of protection from CMV-DNAemia ≥500 copies/mL [CMV pp65 T-SPOT.CMV at the end of prophylaxis: area under curve (AUC) = 0.744, cut-off 142 spot forming units (SFU), sensitivity set to 100%, specificity 46%; CMV IE-1 T-Track CMV at month 1: AUC = 0.762, cut-off 3.5 SFU, sensitivity set to 100%, specificity 59%]. The QuantiFERON-CMV assay was inferior, reaching a specificity of 23% when setting the sensitivity to 100%. In conclusion, both CMV-specific ELISpot assays appear suitable to assess protection from CMV infection/reactivation in liver transplant recipients.
Collapse
Affiliation(s)
- Smaranda Gliga
- Institute for Transfusion Medicine, University Hospital Essen, Virchowstraße 179, 45147 Essen, Germany; (S.G.); (T.D.); (P.A.H.)
- Institute for Virology, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany;
| | - Melanie Fiedler
- Institute for Virology, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany;
| | - Theresa Dornieden
- Institute for Transfusion Medicine, University Hospital Essen, Virchowstraße 179, 45147 Essen, Germany; (S.G.); (T.D.); (P.A.H.)
| | - Anne Achterfeld
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany; (A.A.); (K.H.)
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany;
| | - Andreas Paul
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany;
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, Virchowstraße 179, 45147 Essen, Germany; (S.G.); (T.D.); (P.A.H.)
| | - Kerstin Herzer
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany; (A.A.); (K.H.)
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Essen-Duisburg, 45147 Essen, Germany;
- Knappschaftsklinik Bad Neuenahr, 53474 Bad Neuenahr-Ahrweiler, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, Virchowstraße 179, 45147 Essen, Germany; (S.G.); (T.D.); (P.A.H.)
- Correspondence: ; Tel.: +49-201-723-4217
| |
Collapse
|
33
|
Wu CS, Chyuan IT, Chiu YL, Chen WL, Shen CY, Hsu PN. Preserved specific anti-viral T-cell response but associated with decreased lupus activity in SLE patients with cytomegalovirus infection. Rheumatology (Oxford) 2021; 59:3340-3349. [PMID: 32306043 DOI: 10.1093/rheumatology/keaa143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/26/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES SLE is an autoimmune disease characterized by aberrant autoantibody production and immune dysfunctions. Whether the anti-CMV immunity is impaired in SLE patients is poorly understood. We investigated the specific anti-viral T-cell response in SLE patients with CMV infection and its possible impacts on clinical manifestations in lupus. METHODS CD28 null T-cell percentages were measured by flow cytometry in 89 SLE patients and 58 healthy controls. A specific anti-CMV CD8 T-cell response was assessed ex vivo by the production of intracellular cytokines in response to CMV phosphoprotein 65 (pp65) by flow cytometry. Clinical manifestations and immune parameters were analysed in SLE patients according to their CMV serostatus. RESULTS CD28 null T cells were significantly expanded in SLE patients. When the anti-CMV pp65 CD8 polyfunctional T cell response was analysed, as defined by production of at least three of four functional cytokines or effectors (intracellular IFN-γ, IL-2, TNF-α and surface CD107a), the results demonstrated that it was not impaired in SLE patients. In contrast, when comparing clinical manifestations, there were lower anti-ds-DNA levels and decreased SLEDAI in SLE patients with CMV infection. Furthermore, the expansion of CD4+CD28 null T cells was negatively associated with anti-ds-DNA levels and SLEDAI in these lupus patients. CONCLUSION In SLE patients with CMV infection, the specific anti-CMV CD8 T-cell response is preserved but is associated with decreased disease activity and lower anti-DNA levels among these patients, suggesting CMV infection may mitigate lupus activity.
Collapse
Affiliation(s)
- Chien-Sheng Wu
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City
| | - I-Tsu Chyuan
- Department of Internal Medicine.,Department of Medical Research, Cathay General Hospital, Taipei.,School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City
| | - Yen-Ling Chiu
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City.,Graduate Program in Biomedical Informatics, Yuan Ze University, Taoyuan City
| | - Wen-Lin Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City
| | | | - Ping-Ning Hsu
- Department of Internal Medicine.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Aghbash PS, Hemmat N, Nahand JS, Shamekh A, Memar MY, Babaei A, Baghi HB. The role of Th17 cells in viral infections. Int Immunopharmacol 2021; 91:107331. [PMID: 33418239 DOI: 10.1016/j.intimp.2020.107331] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
The present review provides an overview of recent advances regarding the function of Th17 cells and their produced cytokines in the progression of viral diseases. Viral infections alone do not lead to virus-induced malignancies, as both genetic and host safety factors are also involved in the occurrence of malignancies. Acquired immune responses, through the differentiation of Th17 cells, form the novel components of the Th17 cell pathway when reacting with viral infections all the way from the beginning to its final stages. As a result, instead of inducing the right immune responses, these events lead to the suppression of the immune system. In fact, the responses from Th17 cells during persistent viral infections causes chronic inflammation through the production of IL-17 and other cytokines which provide a favorable environment for tumor growth and its development. Additionally, during the past decade, these cells have been understood to be involved in tumor progression and metastasis. However, further research is required to understand Th17 cells' immune mechanisms in the vast variety of viral diseases. This review aims to determine the roles and effects of the immune system, especially Th17 cells, in the progression of viral diseases; which can be highly beneficial for the diagnosis and treatment of these infections.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran; Drug Applied Research Centre, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, ZIP Code 14155 Tehran, Iran; Student Research Committee, Iran University of Medical Sciences, ZIP Code 14155 Tehran, Iran
| | - Ali Shamekh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran
| | - Abouzar Babaei
- Department of Virology, Faculty of Medicine, Tarbiat Modares University, ZIP Code 14155 Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, ZIP Code 15731 Tabriz, Iran.
| |
Collapse
|
35
|
A Comprehensive Evaluation of Risk Factors for Pneumocystis Jirovecii Pneumonia in Adult Solid Organ Transplant Recipients: a Systematic Review and Meta-Analysis. Transplantation 2020; 105:2291-2306. [PMID: 33323766 DOI: 10.1097/tp.0000000000003576] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND There is no consensus guidance on when to reinitiate Pneumocystis jirovecii pneumonia (PJP) prophylaxis in solid organ transplant (SOT) recipients at increased risk. The 2019 American Society of Transplantation Infectious Diseases Community of Practice (AST IDCOP) guidelines suggested to continue or reinstitute PJP prophylaxis in those receiving intensified immunosuppression for graft rejection, CMV infection, higher dose of corticosteroids, or prolonged neutropenia. METHODS A literature search was conducted evaluating all literature from existence through April 22, 2020 using MEDLINE and EMBASE. (PROSPERO: CRD42019134204) RESULTS:: A total of 30 studies with 413 276 SOT recipients were included. The following factors were associated with PJP development: acute rejection (pooled odds ratio (pOR) = 2.35 (1.69, 3.26), study heterogeneity index (I)= 23.4%), cytomegalovirus (CMV)-related illnesses (pOR = 3.14 (2.30, 4.29), I=48%), absolute lymphocyte count < 500 cells/mm (pOR = 6.29[3.56, 11.13], I 0%), BK-related diseases (pOR = 2.59[1.22, 5.49], I 0%), HLA mismatch ≥ 3 (pOR = 1.83 [1.06, 3.17], I= 0%), rituximab use (pOR =3.03 (1.82, 5.04); I =0%) and polyclonal antibodies use for rejection (pOR = 3.92 [1.87, 8.19], I= 0%). On the other hand, sex, CMV mismatch, interleukin-2 inhibitors, corticosteroids for rejection, and plasmapheresis were not associated with developing PJP. CONCLUSION PJP prophylaxis should be considered in SOT recipients with lymphopenia, BK-related infections and rituximab exposure in addition to the previously mentioned risk factors in the AST IDCOP guidelines.
Collapse
|
36
|
Hatayama Y, Hashimoto Y, Motokura T. Frequent co-reactivation of Epstein-Barr virus in patients with cytomegalovirus viremia under immunosuppressive therapy and/or chemotherapy. J Int Med Res 2020; 48:300060520972880. [PMID: 33249964 PMCID: PMC7708703 DOI: 10.1177/0300060520972880] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objective Co-reactivation of cytomegalovirus (CMV) and Epstein–Barr virus (EBV) occurs in iatrogenically immunosuppressed patients, but the clinical relevance of this is unknown. We aimed to determine the frequency of EBV reactivation in patients with CMV viremia and to explore its clinical significance. Methods Serum or plasma CMV and EBV DNA was detected by quantitative real-time PCR in 82 patients who received immunosuppressive therapy and/or chemotherapy and underwent CMV antigenemia tests. Results CMV DNA was positive in 55 patients, with EBV reactivation being found in 29 of these (52.7%). EBV co-reactivation was significantly associated with aging (>64 years vs. ≤64 years, odds ratio 4.07, 95% confidence interval 1.06–15.6). When older patients were divided into two groups according to age, EBV co-reactivation occurred more frequently in early-old patients (aged 65–74 years) than in late-old patients (aged ≥75 years) (100.0% vs. 53.3%, respectively). Steroid pulse treatment was administered significantly more often in the early-old group than in those aged ≤64 years and ≥75 years (72.7% vs 27.6% vs 14.3%, respectively). Conclusions Co-reactivation of EBV in patients with CMV viremia highlighted early-old patients and may reflect treatment intensity as well as immunosenescence.
Collapse
Affiliation(s)
- Yuki Hatayama
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Tottori, Japan.,Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yuki Hashimoto
- Division of Clinical Laboratory, Tottori University Hospital, Yonago, Tottori, Japan
| | - Toru Motokura
- Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
37
|
Dubey S, Rodrigues C, Nikam C, Samant R. Cytomegalovirus in Indian systemic lupus erythematosus patients: troublemaker or onlooker? Pan Afr Med J 2020; 37:38. [PMID: 33209165 PMCID: PMC7648478 DOI: 10.11604/pamj.2020.37.38.18836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/22/2020] [Indexed: 11/11/2022] Open
Abstract
Introduction cytomegalovirus (CMV) infection has been reported to be associated with onset/exacerbation of systemic lupus erythematosus (SLE). In an attempt to verify this, we studied CMV infection in SLE patients. Methods forty-two SLE patients were studied at 3-time points; disease onset/flare, at peak of immunosuppression (at 6 weeks) and at low doses of immunosuppression (at 6 months). We studied healthy blood donors as controls, only once. Clinical assessment and SLE Disease Activity Index scoring were done at each visit. RT-PCR and ELISA were performed to detect CMV viral-load and anti-CMV antibodies (Ab) respectively. Results nine of 106 patients had detectable viral-load (145-50,000 copies/ml). Of these nine, three patients had significant viral-load, 6 patients had low viral-loads of doubtful clinical significance. None of the patients developed CMV disease. Six of 42 cases were positive for IgM Abs. All controls were negative for CMV DNA as well as CMV IgM Abs. All samples from patients and controls were positive for CMV IgG Ab indicating widespread prevalence. Conclusion significantly, a higher seroprevalence of CMV IgM Abs against CMV observed in SLE patients when compared to controls, indicating possible reactivation due to immune modulation.
Collapse
Affiliation(s)
- Shalini Dubey
- Research Laboratories, P.D. Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Camilla Rodrigues
- Research Laboratories, P.D. Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Chaitali Nikam
- Research Laboratories, P.D. Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Rohini Samant
- Rheumatology Department, P. D. Hinduja Hospital and Medical Research Centre, Mumbai, India
| |
Collapse
|
38
|
Yoshihara R, Komai T, Shoda H, Fujio K. Elevation of cytomegalovirus antigenemia predicts serious infection and death in patients receiving immunosuppressive therapies for autoimmune diseases. Int J Rheum Dis 2020; 23:1534-1540. [PMID: 33051990 DOI: 10.1111/1756-185x.13990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/04/2020] [Accepted: 09/13/2020] [Indexed: 11/27/2022]
Abstract
AIM We examined the relationship between cytomegalovirus (CMV) reactivation and serious infections. METHOD We conducted a single-center retrospective chart review study with 43 autoimmune disease patients experiencing CMV reactivation. We investigated the risk factors for serious infections among the patients using logistic regression analysis. RESULTS We identified that the maximum count of CMV antigenemia during the course of infection (CMV Ag MAX) was significantly associated with serious infection by multivariate analysis (adjusted odds ratio: 1.509; 95% confidence interval: 1.071-2.125). The receiver operating characteristic curve of CMV Ag MAX count showed a predictive value for serious infections (76.9% in sensitivity and 93.3% in specificity) and death (83.3% in sensitivity and 91.9% in specificity), and the cut-off count of serious infections and death was 6 and 10 per 105 white blood cell count, respectively. CONCLUSION We suggest that the counts of CMV Ag MAX can reflect the extent of compromise in the immune system, and can be a predictive marker for serious infections and death.
Collapse
Affiliation(s)
- Risa Yoshihara
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
39
|
Quan H, Kim J, Na YR, Kim JH, Kim BJ, Kim BJ, Hong JJ, Hwang ES, Seok SH. Human Cytomegalovirus-Induced Interleukin-10 Production Promotes the Proliferation of Mycobacterium massiliense in Macrophages. Front Immunol 2020; 11:518605. [PMID: 33013921 PMCID: PMC7511582 DOI: 10.3389/fimmu.2020.518605] [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] [Received: 12/09/2019] [Accepted: 08/18/2020] [Indexed: 01/24/2023] Open
Abstract
Human cytomegalovirus (HCMV) exploits the interleukin-10 (IL-10) pathway as a part of its infection cycle through the manipulation of the host IL-10 signaling cascade. Based on its immunomodulatory nature, HCMV attenuates the host immune response and facilitates the progression of co-infection with other pathogens in an immune-competent host. To investigate the impact of HCMV infection on the burden of non-tuberculous mycobacteria (NTM), whose prevalence is growing rapidly worldwide, macrophages were infected with HCMV and further challenged with Mycobacterium massiliense in vitro. The results showed that HCMV infection significantly increased host IL-10 synthesis and promoted the proliferation of M. massiliense in an IL-10-dependent manner. Transcriptomic analysis revealed that HCMV infection dampened the regulatory pathways of interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-1 (IL-1), consequently abrogating the immune responses to M. massiliense coinfection in macrophages. These findings provide a mechanistic basis of how HCMV infection may facilitate the development of pathogenic NTM co-infection by upregulating IL-10 expression.
Collapse
Affiliation(s)
- Hailian Quan
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
| | - Jiyeon Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea.,Global Center for Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Yi Rang Na
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea.,Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Jung Heon Kim
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea.,Global Center for Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Byoung-Jun Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Eung Soo Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea.,Global Center for Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, South Korea.,Global Center for Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
40
|
Impact of Natural Occurring ERAP1 Single Nucleotide Polymorphisms within miRNA-Binding Sites on HCMV Infection. Int J Mol Sci 2020; 21:ijms21165861. [PMID: 32824160 PMCID: PMC7461596 DOI: 10.3390/ijms21165861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus that causes serious problems in people with a compromised immune system, whereas it coexists asymptomatically within the host with a healthy immune system. Like other viruses, HCMV has adopted multiples strategies to manipulate the host’s immune responses. Among them, expression of viral microRNAs (miRNAs) is one of the most intriguing. HCMV miR-UL112-5p and miR-US4-1 have been found to contribute to immune evasion by targeting the endoplasmic reticulum aminopeptidase 1 (ERAP1), a highly polymorphic key component of antigen processing. The current incomplete picture on the interplay between viral miRNAs and host immunity implies the need to better characterize the host genetic determinants. Naturally occurring single nucleotide polymorphisms (SNPs) within the miRNA binding sites of target genes may affect miRNA–target interactions. In this review, we focus on the relevance of 3′ untranslated region (3′UTR) ERAP1 SNPs within miRNA binding sites in modulating miRNA–mRNA interactions and the possible consequent individual susceptibility to HCMV infection. Moreover, we performed an in silico analysis using different bioinformatic algorithms to predict ERAP1 variants with a putative powerful biological function. This evidence provides a basis to deepen the knowledge on how 3′UTR ERAP1 variants may alter the mechanism of action of HCMV miRNAs, in order to develop targeted antiviral therapies.
Collapse
|
41
|
Cytomegalovirus and Inflammatory Bowel Diseases (IBD) with a Special Focus on the Link with Ulcerative Colitis (UC). Microorganisms 2020; 8:microorganisms8071078. [PMID: 32698383 PMCID: PMC7409252 DOI: 10.3390/microorganisms8071078] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/16/2022] Open
Abstract
Cytomegalovirus (CMV) infects approximately 40% of adults in France and persists lifelong as a latent agent in different organs, including gut. A close relationship is observed between inflammation that favors viral expression and viral replication that exacerbates inflammation. In this context, CMV colitis may impact the prognosis of patients suffering from inflammatory bowel diseases (IBDs), and notably those with ulcerative colitis (UC). In UC, the mucosal inflammation and T helper cell (TH) 2 cytokines, together with immunomodulatory drugs used for controlling flare-ups, favor viral reactivation within the gut, which, in turn, increases mucosal inflammation, impairs corticoid and immunosuppressor efficacy (the probability of steroid resistance is multiplied by more than 20 in the case of CMV colitis), and enhances the risk for colectomy. This review emphasizes the virological tools that are recommended for exploring CMV colitis during inflammatory bowel diseases (IBD) and underlines the interest of using ganciclovir for treating flare-ups associated to CMV colitis in UC patients.
Collapse
|
42
|
Strongyloides stercoralis, Human T-cell Lymphotropic Virus Type-1 and Cytomegalovirus Coinfection in an Allogeneic Hematopoietic Stem-Cell Transplant Recipient. Transplant Direct 2020; 6:e573. [PMID: 32766428 PMCID: PMC7339139 DOI: 10.1097/txd.0000000000001021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 11/26/2022] Open
|
43
|
Poole E, Neves TC, Oliveira MT, Sinclair J, da Silva MCC. Human Cytomegalovirus Interleukin 10 Homologs: Facing the Immune System. Front Cell Infect Microbiol 2020; 10:245. [PMID: 32582563 PMCID: PMC7296156 DOI: 10.3389/fcimb.2020.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Human Cytomegalovirus (HCMV) can cause a variety of health disorders that can lead to death in immunocompromised individuals and neonates. The HCMV lifecycle comprises both a lytic (productive) and a latent (non-productive) phase. HCMV lytic infection occurs in a wide range of terminally differentiated cell types. HCMV latency has been less well-studied, but one characterized site of latency is in precursor cells of the myeloid lineage. All known viral genes are expressed during a lytic infection and a subset of these are also transcribed during latency. The UL111A gene which encodes the viral IL-10, a homolog of the human IL-10, is one of these genes. During infection, different transcript isoforms of UL111A are generated by alternative splicing. The most studied of the UL111A isoforms are cmvIL-10 (also termed the "A" transcript) and LAcmvIL-10 (also termed the "B" transcript), the latter being a well-characterized latency associated transcript. Both isoforms can downregulate MHC class II, however they differ in a number of other immunomodulatory properties, such as the ability to bind the IL10 receptor and induce signaling through STAT3. There are also a number of other isoforms which have been identified which are expressed by differential splicing during lytic infection termed C, D, E, F, and G, although these have been less extensively studied. HCMV uses the viral IL-10 proteins to manipulate the immune system during lytic and latent phases of infection. In this review, we will discuss the literature on the viral IL-10 transcripts identified to date, their encoded proteins and the structures of these proteins as well as the functional properties of all the different isoforms of viral IL-10.
Collapse
Affiliation(s)
- Emma Poole
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tainan Cerqueira Neves
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Martha Trindade Oliveira
- Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - John Sinclair
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
44
|
Barbu MG, Condrat CE, Thompson DC, Bugnar OL, Cretoiu D, Toader OD, Suciu N, Voinea SC. MicroRNA Involvement in Signaling Pathways During Viral Infection. Front Cell Dev Biol 2020; 8:143. [PMID: 32211411 PMCID: PMC7075948 DOI: 10.3389/fcell.2020.00143] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
The study of miRNAs started in 1993, when Lee et al. observed their involvement in the downregulation of a crucial protein known as LIN-14 in the nematode Caenorhabditis elegans. Since then, great progress has been made regarding research on microRNAs, which are now known to be involved in the regulation of various physiological and pathological processes in both animals and humans. One such example is represented by their interaction with various signaling pathways during viral infections. It has been observed that these pathogens can induce the up-/downregulation of various host miRNAs in order to elude the host's immune system. In contrast, some miRNAs studied could have an antiviral effect, enabling the defense mechanisms to fight the infection or, at the very least, they could induce the pathogen to enter a latent state. At the same time, some viruses encode their own miRNAs, which could further modulate the host's signaling pathways, thus favoring the survival and replication of the virus. The goal of this extensive literature review was to present how miRNAs are involved in the regulation of various signaling pathways in some of the most important and well-studied human viral infections. Further on, knowing which miRNAs are involved in various viral infections and what role they play could aid in the development of antiviral therapeutic agents for certain diseases that do not have a definitive cure in the present. The clinical applications of miRNAs are extremely important, as miRNAs targeted inhibition may have substantial therapeutic impact. Inhibition of miRNAs can be achieved through many different methods, but chemically modified antisense oligonucleotides have shown the most prominent effects. Though scientists are far from completely understanding all the molecular mechanisms behind the complex cross-talks between miRNA pathways and viral infections, the general knowledge is increasing on the different roles played by miRNAs during viral infections.
Collapse
Affiliation(s)
- Madalina Gabriela Barbu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Carmen Elena Condrat
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Dana Claudia Thompson
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Oana Larisa Bugnar
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
| | - Dragos Cretoiu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Oana Daniela Toader
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, Bucharest, Romania
| | - Nicolae Suciu
- Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, Bucharest, Romania
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, Bucharest, Romania
| | - Silviu Cristian Voinea
- Department of Surgical Oncology, Institute of Oncology Prof. Dr. Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
45
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
46
|
Park SY, Jung JH, Kwon H, Shin S, Kim YH, Chong YP, Lee SO, Choi SH, Kim YS, Woo JH, Kim SH, Han DJ. Epidemiology and risk factors associated with Pneumocystis jirovecii pneumonia in kidney transplant recipients after 6-month trimethoprim-sulfamethoxazole prophylaxis: A case-control study. Transpl Infect Dis 2020; 22:e13245. [PMID: 31943590 DOI: 10.1111/tid.13245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/15/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pneumocystis jirovecii pneumonia (PCP) is an important cause of morbidity and mortality in kidney transplant recipients (KTRs), and prophylaxis with trimethoprim-sulfamethoxazole (TMP-SMX) is recommended. The aim of this study was to investigate incidence and risk factors for PCP in KTRs after 6-month TMP-SMX prophylaxis. METHODS We conducted a case-control study of patients with PCP who received 6-month PCP prophylaxis with TMP-SMX after kidney transplantation (KT). In cases of rejection, PCP prophylaxis was provided for six additional months after anti-rejection therapy. Cytomegalovirus (CMV) infection was not considered an indication for PCP prophylaxis due to concerns of nephrotoxicity associated with TMP-SMX. RESULTS Among 3941 kidney or pancreas-kidney transplant recipients, 67 (1.7%) developed PCP after discontinuing TMP-SMX. A total of 47 patients with KT PCP and 94 controls were included. Duration of PCP prophylaxis was similar between cases and controls (median 6 months, P = .53). In multivariate analysis, rejection (OR 3.9; 95% CI 1.4-11.1) and CMV infection (OR 2.4; 95% CI 1.0-5.8) were independently associated with PCP development after TMP-SMX. Rejection or CMV infection was observed in 70% of patients with PCP. Time to PCP development after rejection (median [IQR] 6 [5-19] months) was slightly shorter than after CMV infection (median [IQR] 9 [5-12] months; P = .18). CONCLUSION Post-prophylaxis PCP occurred in <2% of KTRs, and about two-thirds of these experienced rejection or CMV infection. These data suggest that at least 6 to 9-month additional chemoprophylaxis may be needed to prevent PCP in KTRs with transplant rejection or CMV infection.
Collapse
Affiliation(s)
- Se Yoon Park
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Division of Infectious Diseases, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Joo Hee Jung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyunwook Kwon
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Shin
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Hoon Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong-Phil Chong
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Oh Lee
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yang Soo Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun Hee Woo
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Duck Jong Han
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Meester I, Rivera-Silva GF, González-Salazar F. Immune System Sex Differences May Bridge the Gap Between Sex and Gender in Fibromyalgia. Front Neurosci 2020; 13:1414. [PMID: 32009888 PMCID: PMC6978848 DOI: 10.3389/fnins.2019.01414] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
The fibromyalgia syndrome (FMS) is characterized by chronic widespread pain, sleep disturbances, fatigue, and cognitive alterations. A limited efficacy of targeted treatment and a high FMS prevalence (2–5% of the adult population) sums up to high morbidity. Although, altered nociception has been explained with the central sensitization hypothesis, which may occur after neuropathy, its molecular mechanism is not understood. The marked female predominance among FMS patients is often attributed to a psychosocial predisposition of the female gender, but here we will focus on sex differences in neurobiological processes, specifically those of the immune system, as various immunological biomarkers are altered in FMS. The activation of innate immune sensors is compatible with a neuropathy or virus-induced autoimmune diseases. Considering sex differences in the immune system and the clustering of FMS with autoimmune diseases, we hypothesize that the female predominance in FMS is due to a neuropathy-induced autoimmune pathophysiology. We invite the scientific community to verify the autoimmune hypothesis for FMS.
Collapse
Affiliation(s)
- Irene Meester
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico
| | - Gerardo Francisco Rivera-Silva
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico
| | - Francisco González-Salazar
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico.,Laboratory of Cellular Physiology, Northeast Center of Research, Mexican Institute of Social Security, Monterrey, Mexico
| |
Collapse
|
48
|
Frascaroli G, Rossini G, Maltoni V, Bartoletti M, Ortolani P, Gredmark-Russ S, Gelsomino F, Moroni A, Silenzi S, Castellani G, Sambri V, Mastroianni A, Brune W, Varani S. Genetic and Functional Characterization of Toll-Like Receptor Responses in Immunocompetent Patients With CMV Mononucleosis. Front Cell Infect Microbiol 2020; 10:386. [PMID: 32850485 PMCID: PMC7426556 DOI: 10.3389/fcimb.2020.00386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Human cytomegalovirus (CMV) modulates both innate and adaptive immune responses. However, limited data are available on the role of receptors of innate immunity, such as toll-like receptors (TLRs) in contributing to antiviral responses and inflammation. Objectives: The aim of this translational study was to characterize TLR responses in immunocompetent patients with primary and symptomatic CMV infection. Study Design: The study population consisted of 40 patients suffering from CMV mononucleosis and 124 blood donors included as controls. We evaluated the association between TLR2, 3, 4, 7 and 9 gene single nucleotide polymorphism (SNP) and susceptibility to symptomatic CMV infection in immunocompetent adults. Additionally, functional TLR-mediated cytokine responses in supernatants of short-term cultures of whole blood from patients with CMV mononucleosis and blood donors were evaluated. Results: TLR2 and TLR7/8 responses were altered in CMV infected patients as compared to healthy donors and were associated with the release of higher levels of the pro-inflammatory cytokines IL-6 and TNF-α, but not of the anti-inflammatory mediator IL-10. The analysis on the TLR SNPs indicated no difference between patients with CMV infection and the control group. Conclusions: No variation in the TLR2,3,4,7 and 9 genes was associated to the development of symptomatic CMV infection in immunocompetent adults. Nevertheless, TLR-mediated responses in CMV-infected patients appeared to be skewed toward a pro-inflammatory profile, which may contribute to the development of inflammatory symptoms during the CMV mononucleotic syndrome.
Collapse
Affiliation(s)
- Giada Frascaroli
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- *Correspondence: Giada Frascaroli
| | - Giada Rossini
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Virginia Maltoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Michele Bartoletti
- Infectious Diseases Unit, Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | | | - Sara Gredmark-Russ
- Center for Infectious Medicine, ANA Futura, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Francesco Gelsomino
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Alessandra Moroni
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Silvia Silenzi
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy and Galvani Center for Biocomplexity, University of Bologna, Bologna, Italy
| | - Vittorio Sambri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Unit of Microbiology, The Romagna Hub Laboratory, Pievesestina, Italy
| | - Antonio Mastroianni
- Unit of Infectious and Tropical Diseases, St. Annunziata Hospital, Cosenza, Italy
- Unit of Infectious Diseases, G.B. Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Wolfram Brune
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stefania Varani
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
49
|
Yap XZ, McCall MBB, Sauerwein RW. Fast and fierce versus slow and smooth: Heterogeneity in immune responses to Plasmodium in the controlled human malaria infection model. Immunol Rev 2020; 293:253-269. [PMID: 31605396 PMCID: PMC6973142 DOI: 10.1111/imr.12811] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Controlled human malaria infection (CHMI) is an established model in clinical malaria research. Upon exposure to Plasmodium falciparum parasites, malaria-naive volunteers differ in dynamics and composition of their immune profiles and subsequent capacity to generate protective immunity. CHMI volunteers are either inflammatory responders who have prominent cellular IFN-γ production primarily driven by adaptive T cells, or tempered responders who skew toward antibody-mediated humoral immunity. When exposed to consecutive CHMIs under antimalarial chemoprophylaxis, individuals who can control parasitemia after a single immunization (fast responders) are more likely to be protected against a subsequent challenge infection. Fast responders tend to be inflammatory responders who can rapidly induce long-lived IFN-γ+ T cell responses. Slow responders or even non-responders can also be protected, but via a more diverse range of responses that take a longer time to reach full protective efficacy, in part due to their tempered phenotype. The latter group can be identified at baseline before CHMI by higher expression of inhibitory ligands CTLA-4 and TIM-3 on CD4+ T cells. Delineating heterogeneity in human immune responses to P. falciparum will facilitate rational design and strategy towards effective malaria vaccines.
Collapse
Affiliation(s)
- Xi Zen Yap
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Matthew B. B. McCall
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Robert W. Sauerwein
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
50
|
Fani M, Zandi M, Rezayi M, Khodadad N, Langari H, Amiri I. The Role of microRNAs in the Viral Infections. Curr Pharm Des 2019; 24:4659-4667. [PMID: 30636585 DOI: 10.2174/1381612825666190110161034] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs with 19 to 24 nucleotides which are evolutionally conserved. MicroRNAs play a regulatory role in many cellular functions such as immune mechanisms, apoptosis, and tumorigenesis. The main function of miRNAs is the post-transcriptional regulation of gene expression via mRNA degradation or inhibition of translation. In fact, many of them act as an oncogene or tumor suppressor. These molecular structures participate in many physiological and pathological processes of the cell. The virus can also produce them for developing its pathogenic processes. It was initially thought that viruses without nuclear replication cycle such as Poxviridae and RNA viruses can not code miRNA, but recently, it has been proven that RNA viruses can also produce miRNA. The aim of this articles is to describe viral miRNAs biogenesis and their effects on cellular and viral genes.
Collapse
Affiliation(s)
- Mona Fani
- Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Majid Rezayi
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nastaran Khodadad
- Virology Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Langari
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iraj Amiri
- Computational Optics Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|