1
|
Lai S, Wei W, Yang S, Wu Y, Shi M, Meng S, Tao X, Chen S, Chen R, Su J, Yuan Z, Ye L, Liang H, Xie Z, Jiang J. Machine learning-driven in-hospital mortality prediction in HIV/AIDS patients with Cytomegalovirus infection: a single-centred retrospective study. J Med Microbiol 2024; 73. [PMID: 39606806 DOI: 10.1099/jmm.0.001935] [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: 11/29/2024] Open
Abstract
Introduction. Cytomegalovirus (CMV) is a widely disseminated betaherpesvirus that typically induces latant infections. In immunocompromised populations, especially transplant and HIV-infected patients, CMV infection increases in-hospital mortality.Gap statement. Although machine learning models have been widely used in clinical diagnosis and prognosis prediction, reports on machine learning model predictions for the in-hospital mortality of HIV/AIDS patients with CMV infection have not been reported.Aim. Analyze the general gemographic and clinical characteristics of HIV/AIDS patients with CMV infection and identify the factors affecting the prognosis of this population, which will help to reduce their in-hospital mortality.Methods. Hospitalized HIV/AIDS patients with CMV infection were recruited from the Fourth People's Hospital of Nanning, Guangxi, from 2012 to 2019. After dividing them into survival and death groups based on their in-hospital survival status, their general and clinical profiles were described. Following 1 : 3 propensity score matching to equalize baseline characteristics, three machine-learning models (Random Forest, Support Vector Machine and eXtreme Gradient Boosting) were deployed to forecast factors influencing prognosis. The SHapley Additive exPlanations tool explained the models.Results. A total of 1102 HIV/AIDS patients with CMV infection were analysed. There was no statistical difference in the general condition of the study subjects (P>0.05). Prevalent complications/coinfections included pneumonia (63.6%), tuberculosis (47.2%) and oral fungal infections (44.6%). There were significant differences between the groups in pneumonia, cryptococcosis and hypoproteinaemia (P<0.05). The differences in laboratory indicators between patients were also statistically significant (P<0.05). The three machine learning models demonstrated good performance, identifying primary predictors of mortality. Pneumonia, urea, indirect bilirubin and platelet distribution width exhibited positive associations with death, with higher levels correlating with an increased mortality risk. Conversely, CD4 T-cell count, CD8 T-cell count and platelet displayed negative correlations with mortality.Conclusions. HIV/AIDS patients with CMV infection exhibit distinctive clinical features impacting survival outcomes. Machine learning models accurately identify key influencing factors and predict mortality risk in this population, which appears to be essential to reducing in-hospital mortality.
Collapse
Affiliation(s)
- Shiyi Lai
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Wudi Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Shixiong Yang
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Yuting Wu
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Minjuan Shi
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Sirun Meng
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Xing Tao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Shanshan Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Rongfeng Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zongxiang Yuan
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zhiman Xie
- The Fourth People's Hospital of Nanning, Nanning, Guangxi, PR China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, PR China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, PR China
| |
Collapse
|
2
|
Nunes M, Vlok M, Proal A, Kell DB, Pretorius E. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc Diabetol 2024; 23:254. [PMID: 39014464 PMCID: PMC11253362 DOI: 10.1186/s12933-024-02315-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/16/2024] [Indexed: 07/18/2024] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic condition that is characterized by unresolved fatigue, post-exertion symptom exacerbation (PESE), cognitive dysfunction, orthostatic intolerance, and other symptoms. ME/CFS lacks established clinical biomarkers and requires further elucidation of disease mechanisms. A growing number of studies demonstrate signs of hematological and cardiovascular pathology in ME/CFS cohorts, including hyperactivated platelets, endothelial dysfunction, vascular dysregulation, and anomalous clotting processes. To build on these findings, and to identify potential biomarkers that can be related to pathophysiology, we measured differences in protein expression in platelet-poor plasma (PPP) samples from 15 ME/CFS study participants and 10 controls not previously infected with SARS-CoV-2, using DIA LC-MS/MS. We identified 24 proteins that are significantly increased in the ME/CFS group compared to the controls, and 21 proteins that are significantly downregulated. Proteins related to clotting processes - thrombospondin-1 (important in platelet activation), platelet factor 4, and protein S - were differentially expressed in the ME/CFS group, suggestive of a dysregulated coagulation system and abnormal endothelial function. Complement machinery was also significantly downregulated, including C9 which forms part of the membrane attack complex. Additionally, we identified a significant upregulation of lactotransferrin, protein S100-A9, and an immunoglobulin variant. The findings from this experiment further implicate the coagulation and immune system in ME/CFS, and bring to attention the pathology of or imposed on the endothelium. This study highlights potential systems and proteins that require further research with regards to their contribution to the pathogenesis of ME/CFS, symptom manifestation, and biomarker potential, and also gives insight into the hematological and cardiovascular risk for ME/CFS individuals affected by diabetes mellitus.
Collapse
Affiliation(s)
- Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa
| | - Mare Vlok
- Central Analytical Facility: Mass Spectrometry, Stellenbosch University, Tygerberg Campus, Room 6054, Clinical Building, Francie Van Zijl Drive Tygerberg, Cape Town, 7505, South Africa
| | - Amy Proal
- PolyBio Research Foundation, 7900 SE 28th ST, Suite 412, Mercer Island, DC, 98040, USA
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet 200, 2800, Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| |
Collapse
|
3
|
Sui Y, Meyer TJ, Fennessey CM, Keele BF, Dadkhah K, Ma C, LaBranche CC, Breed MW, Kramer JA, Li J, Howe SE, Ferrari G, Williams LD, Cam M, Kelly MC, Shen X, Tomaras GD, Montefiori D, Greten TF, Miller CJ, Berzofsky JA. Innate protection against intrarectal SIV acquisition by a live SHIV vaccine. JCI Insight 2024; 9:e175800. [PMID: 38912579 PMCID: PMC11383375 DOI: 10.1172/jci.insight.175800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/08/2024] [Indexed: 06/25/2024] Open
Abstract
Identifying immune correlates of protection is a major challenge in AIDS vaccine development. Anti-Envelope antibodies have been considered critical for protection against SIV/HIV (SHIV) acquisition. Here, we evaluated the efficacy of an SHIV vaccine against SIVmac251 challenge, where the role of antibody was excluded, as there was no cross-reactivity between SIV and SHIV envelope antibodies. After 8 low-dose intrarectal challenges with SIVmac251, 12 SHIV-vaccinated animals demonstrated efficacy, compared with 6 naive controls, suggesting protection was achieved in the absence of anti-envelope antibodies. Interestingly, CD8+ T cells (and some NK cells) were not essential for preventing viral acquisition, as none of the CD8-depleted macaques were infected by SIVmac251 challenges. Initial investigation of protective innate immunity revealed that protected animals had elevated pathways related to platelet aggregation/activation and reduced pathways related to interferon and responses to virus. Moreover, higher expression of platelet factor 4 on circulating platelet-leukocyte aggregates was associated with reduced viral acquisition. Our data highlighted the importance of innate immunity, identified mechanisms, and may provide opportunities for novel HIV vaccines or therapeutic strategy development.
Collapse
Affiliation(s)
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | | | | - Kimia Dadkhah
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Chi Ma
- Thoracic and GI Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Celia C. LaBranche
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew W. Breed
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Bethesda, Maryland, USA
| | - Josh A. Kramer
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Bethesda, Maryland, USA
| | | | | | | | - LaTonya D. Williams
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Michael C. Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - David Montefiori
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tim F. Greten
- Thoracic and GI Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Christopher J. Miller
- Center for Comparative Medicine, University of California, Davis, Davis, California, USA
| | | |
Collapse
|
4
|
Liu Z, Li L, Zhang H, Pang X, Qiu Z, Xiang Q, Cui Y. Platelet factor 4(PF4) and its multiple roles in diseases. Blood Rev 2024; 64:101155. [PMID: 38008700 DOI: 10.1016/j.blre.2023.101155] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/24/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
Platelet factor 4 (PF4) combines with heparin to form an antigen that could produce IgG antibodies and participate in heparin-induced thrombocytopenia (HIT). PF4 has attracted wide attention due to its role in novel coronavirus vaccine-19 (COVID-9)-induced immune thrombotic thrombocytopenia (VITT) and cognitive impairments. The electrostatic interaction between PF4 and negatively charged molecules is vital in the progression of VITT, which is similar to HIT. Emerging evidence suggests its multiple roles in hematopoietic and angiogenic inhibition, platelet coagulation interference, host inflammatory response promotion, vascular inhibition, and antitumor properties. The emerging pharmacological effects of PF4 may help deepen the exploration of its mechanism, thus accelerating the development of targeted therapies. However, due to its pleiotropic properties, the development of drugs targeting PF4 is at an early stage and faces many challenges. Herein, we discussed the characteristics and biological functions of PF4, summarized PF4-mediated signaling pathways, and discussed its multiple roles in diseases to inform novel approaches for successful clinical translational research.
Collapse
Affiliation(s)
- Zhiyan Liu
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Clinical Pharmacology, Peking University First Hospital, China.
| | - Longtu Li
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Hanxu Zhang
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaocong Pang
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Clinical Pharmacology, Peking University First Hospital, China
| | - Zhiwei Qiu
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Clinical Pharmacology, Peking University First Hospital, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Clinical Pharmacology, Peking University First Hospital, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China; Institute of Clinical Pharmacology, Peking University First Hospital, China.
| |
Collapse
|
5
|
Awamura T, Nakasone ES, Gangcuangco LM, Subia NT, Bali AJ, Chow DC, Shikuma CM, Park J. Platelet and HIV Interactions and Their Contribution to Non-AIDS Comorbidities. Biomolecules 2023; 13:1608. [PMID: 38002289 PMCID: PMC10669125 DOI: 10.3390/biom13111608] [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: 10/05/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Platelets are anucleate cytoplasmic cell fragments that circulate in the blood, where they are involved in regulating hemostasis. Beyond their normal physiologic role, platelets have emerged as versatile effectors of immune response. During an infection, cell surface receptors enable platelets to recognize viruses, resulting in their activation. Activated platelets release biologically active molecules that further trigger host immune responses to protect the body against infection. Their impact on the immune response is also associated with the recruitment of circulating leukocytes to the site of infection. They can also aggregate with leukocytes, including lymphocytes, monocytes, and neutrophils, to immobilize pathogens and prevent viral dissemination. Despite their host protective role, platelets have also been shown to be associated with various pathophysiological processes. In this review, we will summarize platelet and HIV interactions during infection. We will also highlight and discuss platelet and platelet-derived mediators, how they interact with immune cells, and the multifaceted responsibilities of platelets in HIV infection. Furthermore, we will give an overview of non-AIDS comorbidities linked to platelet dysfunction and the impact of antiretroviral therapy on platelet function.
Collapse
Affiliation(s)
- Thomas Awamura
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Elizabeth S. Nakasone
- University of Hawai‘i Cancer Center, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
| | - Louie Mar Gangcuangco
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Natalie T. Subia
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Aeron-Justin Bali
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Dominic C. Chow
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Cecilia M. Shikuma
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| |
Collapse
|
6
|
Carestia A, Godin LC, Jenne CN. Step up to the platelet: Role of platelets in inflammation and infection. Thromb Res 2023; 231:182-194. [PMID: 36307228 DOI: 10.1016/j.thromres.2022.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022]
Abstract
Platelets are anucleated cells derived from megakaryocytes that are primarily responsible for hemostasis. However, in recent years, these cytoplasts have become increasingly recognized as immune cells, able to detect, interact with, and kill pathogens. As platelets are involved in both immunity and coagulation, they have a central role in immunothrombosis, a physiological process in which immune cells induce the formation of microthrombi to both prevent the spread of pathogens, and to help facilitate clearance. In this review, we will highlight the role of platelets as key players in the inflammatory and innate immune response against bacterial and viral infection, including direct and indirect interactions with pathogens and other immune cells.
Collapse
Affiliation(s)
- Agostina Carestia
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Laura C Godin
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
7
|
Xue M, Gao Z, Yan M, Bao Y. Profiling risk factors for separation of infection complications in patients with gastrointestinal and nodal diffuse large B-cell lymphoma. BMC Infect Dis 2023; 23:711. [PMID: 37864133 PMCID: PMC10589955 DOI: 10.1186/s12879-023-08671-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023] Open
Abstract
OBJECTIVE To identify risk factors for infection complications in patients with gastrointestinal diffuse large B-cell lymphoma (GI-DLBCL) and nodal DLBCL (N-DLBCL) during treatment, respectively. METHODS Total 51 GI-DLBCL patients and 80 N-DLBCL patients were included after retrieving clinical data from a single medical center in the past ten years. Logistic regression analysis was utilized to analyze patients' data, including baseline demographics, treatments and laboratory values, to determine independent risk factors of infection in these patients. RESULTS Total 28 of 51 patients (54.9%) in the GI-DLBCL group and 52 of 80 patients (65%) in the N-DLBCL group were observed infection events during treatment. A multivariate logistic regression model revealed that Ann-arbor stage IV (P = 0.034; odds ratio [OR]: 10.635; 95% confidence interval [CI]: 1.152-142.712), extra-nodal lesions ≥ 2 (P = 0.041; OR: 23.116; 95%CI: 1.144-466.949) and high serum lactate dehydrogenase (LDH) at the time of diagnosis (LDH > 252U/L; P = 0.033; OR: 6.058; 95%CI: 1.159-31.659) were independent risk factors for the development of infection in patients with GI-DLBCL after systemic treatment. In the N-DLBCL group, high serum C-reactive protein (CRP) (P = 0.027; OR: 1.104; 95%CI: 1.011-1.204) and a low platelet count (P = 0.041; OR: 0.991; 95%CI: 0.982-1.000) at routine blood tests just before infection occurred were identified as significant risk factors related to infection events during treatment. CONCLUSIONS Discordant independent risk factors induced infection may be present during the treatment in patients with GI-DLBCL and N-DLBCL. Close monitoring these risk factors is likely an effective strategy to prevent microbial infections in these patients.
Collapse
Affiliation(s)
- Min Xue
- Graduate School, Bengbu Medical College, 2600 Donghai Road, Bengbu, 233000, Anhui, China
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China
| | - Zhenzhen Gao
- The Department of Oncology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China
| | - Miaolong Yan
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China
| | - Yi Bao
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China.
- The Department of Oncology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China.
| |
Collapse
|
8
|
Zhao J, Xu X, Gao Y, Yu Y, Li C. Crosstalk between Platelets and SARS-CoV-2: Implications in Thrombo-Inflammatory Complications in COVID-19. Int J Mol Sci 2023; 24:14133. [PMID: 37762435 PMCID: PMC10531760 DOI: 10.3390/ijms241814133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The SARS-CoV-2 virus, causing the devastating COVID-19 pandemic, has been reported to affect platelets and cause increased thrombotic events, hinting at the possible bidirectional interactions between platelets and the virus. In this review, we discuss the potential mechanisms underlying the increased thrombotic events as well as altered platelet count and activity in COVID-19. Inspired by existing knowledge on platelet-pathogen interactions, we propose several potential antiviral strategies that platelets might undertake to combat SARS-CoV-2, including their abilities to internalize the virus, release bioactive molecules to interfere with viral infection, and modulate the functions of immune cells. Moreover, we discuss current and potential platelet-targeted therapeutic strategies in controlling COVID-19, including antiplatelet drugs, anticoagulants, and inflammation-targeting treatments. These strategies have shown promise in clinical settings to alleviate the severity of thrombo-inflammatory complications and reduce the mortality rate among COVID-19 patients. In conclusion, an in-depth understanding of platelet-SARS-CoV-2 interactions may uncover novel mechanisms underlying severe COVID-19 complications and could provide new therapeutic avenues for managing this disease.
Collapse
Affiliation(s)
| | | | | | - Yijing Yu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (J.Z.); (X.X.); (Y.G.)
| | - Conglei Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (J.Z.); (X.X.); (Y.G.)
| |
Collapse
|
9
|
Lakerveld AJ, van Erp EA, van Kasteren PB. Binding of respiratory syncytial virus particles to platelets does not result in their degranulation in vitro. Access Microbiol 2023; 5:acmi000481.v3. [PMID: 37601435 PMCID: PMC10436017 DOI: 10.1099/acmi.0.000481.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe respiratory infection in infants and the elderly. The mechanisms behind severe RSV disease are incompletely understood, but a dysregulated immune response probably plays an important role. Platelets are increasingly being recognized as immune cells and are involved in the pathology of several viruses. The release of chemokines from platelets upon activation may attract, for example, neutrophils to the site of infection, which is a hallmark of RSV pathology. In addition, since RSV infections are sometimes associated with cardiovascular events and platelets express several known RSV receptors, we investigated the effect of RSV exposure on platelet degranulation. Washed human platelets were incubated with sucrose-purified RSV particles. P-selectin and CD63 surface expression and CCL5 secretion were measured to assess platelet degranulation. We found that platelets bind and internalize RSV particles, but this does not result in degranulation. Our results suggest that platelets do not play a direct role in RSV pathology by releasing chemokines to attract inflammatory cells.
Collapse
Affiliation(s)
- Anke J. Lakerveld
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Elisabeth A. van Erp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Puck B. van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
10
|
Castell N, Guerrero-Martin SM, Rubin LH, Shirk EN, Brockhurst JK, Lyons CE, Najarro KM, Queen SE, Carlson BW, Adams RJ, Morrell CN, Gama L, Graham DR, Zink C, Mankowski JL, Clements JE, Metcalf Pate KA. Effect of Single Housing on Innate Immune Activation in Immunodeficiency Virus-Infected Pigtail Macaques ( Macaca nemestrina ) as a Model of Psychosocial Stress in Acute HIV Infection. Psychosom Med 2022; 84:966-975. [PMID: 36162063 PMCID: PMC9553260 DOI: 10.1097/psy.0000000000001132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 07/27/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Simian immunodeficiency virus (SIV) infection of macaques recapitulates many aspects of HIV pathogenesis and is similarly affected by both genetic and environmental factors. Psychosocial stress is associated with immune system dysregulation and worse clinical outcomes in people with HIV. This study assessed the impact of single housing, as a model of psychosocial stress, on innate immune responses of pigtailed macaques ( Macaca nemestrina ) during acute SIV infection. METHODS A retrospective analysis of acute SIV infection of 2- to si6-year-old male pigtailed macaques was performed to compare the innate immune responses of socially ( n = 41) and singly ( n = 35) housed animals. Measures included absolute monocyte count and subsets, and in a subset ( n ≤ 18) platelet counts and activation data. RESULTS SIV infection resulted in the expected innate immune parameter changes with a modulating effect from housing condition. Monocyte number increased after infection for both groups, driven by classical monocytes (CD14 + CD16 - ), with a greater increase in socially housed animals (227%, p < .001, by day 14 compared with preinoculation time points). Platelet numbers recovered more quickly in the socially housed animals. Platelet activation (P-selectin) increased by 65% ( p = .004) and major histocompatibility complex class I surface expression by 40% ( p = .009) from preinoculation only in socially housed animals, whereas no change in these measures occurred in singly housed animals. CONCLUSIONS Chronic psychosocial stress produced by single housing may play an immunomodulatory role in the innate immune response to acute retroviral infection. Dysregulated innate immunity could be one of the pathways by which psychosocial stress contributes to immune suppression and increased disease severity in people with HIV.
Collapse
|
11
|
Jan MW, Chiu CY, Chen JJ, Chang TH, Tsai KJ. Human Platelet Lysate Induces Antiviral Responses against Parechovirus A3. Viruses 2022; 14:v14071499. [PMID: 35891479 PMCID: PMC9316291 DOI: 10.3390/v14071499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/27/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Human platelet lysate (hPL) contains abundant growth factors for inducing human cell proliferation and may be a suitable alternative to fetal bovine serum (FBS) as a culture medium supplement. However, the application of hPL in virological research remains blank. Parechovirus type-A3 (PeV-A3) belongs to Picornaviridae, which causes meningoencephalitis in infants and young children. To understand the suitability of hPL-cultured cells for PeV-A3 infection, the infection of PeV-A3 in both FBS- and hPL-cultured glioblastoma (GBM) cells were compared. Results showed reduced PeV-A3 infection in hPL-cultured cells compared with FBS-maintained cells. Mechanistic analysis revealed hPL stimulating type I interferon (IFN) antiviral pathway, through which phospho-signal transducer and activator of transcription 1 (STAT1), STAT2, interferon regulatory factor 3 (IRF3) were activated and antiviral genes, such as IFN-α, IFN-β, and Myxovirus resistance protein 1 (MxA), were also detected. In addition, an enhanced PeV-A3 replication was detected in the hPL-cultured GBM cells treated with STAT-1 inhibitor (fludarabine) and STAT1 shRNA. These results in vitro suggested an unexpected effect of hPL-activated type I IFN pathway response to restrict virus replication and that hPL may be a potential antiviral bioreagent.
Collapse
Affiliation(s)
- Ming-Wei Jan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Chih-Yun Chiu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan;
| | - Jih-Jung Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Tsung-Hsien Chang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan;
- Correspondence: (T.-H.C.); (K.-J.T.)
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Correspondence: (T.-H.C.); (K.-J.T.)
| |
Collapse
|
12
|
Schrottmaier WC, Schmuckenschlager A, Pirabe A, Assinger A. Platelets in Viral Infections - Brave Soldiers or Trojan Horses. Front Immunol 2022; 13:856713. [PMID: 35419008 PMCID: PMC9001014 DOI: 10.3389/fimmu.2022.856713] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Viral infections are often associated with platelet activation and haemostatic complications. In line, low platelet counts represent a hallmark for poor prognosis in many infectious diseases. The underlying cause of platelet dysfunction in viral infections is multifaceted and complex. While some viruses directly interact with platelets and/or megakaryocytes to modulate their function, also immune and inflammatory responses directly and indirectly favour platelet activation. Platelet activation results in increased platelet consumption and degradation, which contributes to thrombocytopenia in these patients. The role of platelets is often bi-phasic. Initial platelet hyper-activation is followed by a state of platelet exhaustion and/or hypo-responsiveness, which together with low platelet counts promotes bleeding events. Thereby infectious diseases not only increase the thrombotic but also the bleeding risk or both, which represents a most dreaded clinical complication. Treatment options in these patients are limited and new therapeutic strategies are urgently needed to prevent adverse outcome. This review summarizes the current literature on platelet-virus interactions and their impact on viral pathologies and discusses potential intervention strategies. As pandemics and concomitant haemostatic dysregulations will remain a recurrent threat, understanding the role of platelets in viral infections represents a timely and pivotal challenge.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anna Schmuckenschlager
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
14
|
Ostermeier B, Soriano-Sarabia N, Maggirwar SB. Platelet-Released Factors: Their Role in Viral Disease and Applications for Extracellular Vesicle (EV) Therapy. Int J Mol Sci 2022; 23:2321. [PMID: 35216433 PMCID: PMC8876984 DOI: 10.3390/ijms23042321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Platelets, which are small anuclear cell fragments, play important roles in thrombosis and hemostasis, but also actively release factors that can both suppress and induce viral infections. Platelet-released factors include sCD40L, microvesicles (MVs), and alpha granules that have the capacity to exert either pro-inflammatory or anti-inflammatory effects depending on the virus. These factors are prime targets for use in extracellular vesicle (EV)-based therapy due to their ability to reduce viral infections and exert anti-inflammatory effects. While there are some studies regarding platelet microvesicle-based (PMV-based) therapy, there is still much to learn about PMVs before such therapy can be used. This review provides the background necessary to understand the roles of platelet-released factors, how these factors might be useful in PMV-based therapy, and a critical discussion of current knowledge of platelets and their role in viral diseases.
Collapse
Affiliation(s)
| | | | - Sanjay B. Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, 2300 I Street NW, Washington, DC 20037, USA; (B.O.); (N.S.-S.)
| |
Collapse
|
15
|
Zacharopoulou P, Marchi E, Ogbe A, Robinson N, Brown H, Jones M, Parolini L, Pace M, Grayson N, Kaleebu P, Rees H, Fidler S, Goulder P, Klenerman P, Frater J. Expression of type I interferon-associated genes at antiretroviral therapy interruption predicts HIV virological rebound. Sci Rep 2022; 12:462. [PMID: 35013427 PMCID: PMC8748440 DOI: 10.1038/s41598-021-04212-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Although certain individuals with HIV infection can stop antiretroviral therapy (ART) without viral load rebound, the mechanisms under-pinning 'post-treatment control' remain unclear. Using RNA-Seq we explored CD4 T cell gene expression to identify evidence of a mechanism that might underpin virological rebound and lead to discovery of associated biomarkers. Fourteen female participants who received 12 months of ART starting from primary HIV infection were sampled at the time of stopping therapy. Two analysis methods (Differential Gene Expression with Gene Set Enrichment Analysis, and Weighted Gene Co-expression Network Analysis) were employed to interrogate CD4+ T cell gene expression data and study pathways enriched in post-treatment controllers versus early rebounders. Using independent analysis tools, expression of genes associated with type I interferon responses were associated with a delayed time to viral rebound following treatment interruption (TI). Expression of four genes identified by Cox-Lasso (ISG15, XAF1, TRIM25 and USP18) was converted to a Risk Score, which associated with rebound (p < 0.01). These data link transcriptomic signatures associated with innate immunity with control following stopping ART. The results from this small sample need to be confirmed in larger trials, but could help define strategies for new therapies and identify new biomarkers for remission.
Collapse
Affiliation(s)
- P Zacharopoulou
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - E Marchi
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - A Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - N Robinson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - H Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M Jones
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - L Parolini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - N Grayson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - P Kaleebu
- Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda
| | - H Rees
- Wits Reproductive Health and HIV Institute of the University of the Witwatersrand in Johannesburg, Johannesburg, South Africa
| | - S Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, UK
- Imperial College NIHR Biomedical Research Centre, London, UK
| | - P Goulder
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - P Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- National Institute of Health Research Biomedical Research Centre, Oxford, UK
| | - J Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- National Institute of Health Research Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
16
|
Dai XP, Wu FY, Cui C, Liao XJ, Jiao YM, Zhang C, Song JW, Fan X, Zhang JY, He Q, Wang FS. Increased Platelet-CD4+ T Cell Aggregates Are Correlated With HIV-1 Permissiveness and CD4+ T Cell Loss. Front Immunol 2021; 12:799124. [PMID: 34987521 PMCID: PMC8720770 DOI: 10.3389/fimmu.2021.799124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic HIV-1 infection is associated with persistent inflammation, which contributes to disease progression. Platelet-T cell aggregates play a critical role in maintaining inflammation. However, the phenotypic characteristics and clinical significance of platelet-CD4+ T cell aggregates remain unclear in different HIV-infected populations. In this study, we quantified and characterized platelet-CD4+ T cell aggregates in the peripheral blood of treatment-naïve HIV-1-infected individuals (TNs), immunological responders to antiretroviral therapy (IRs), immunological non-responders to antiretroviral therapy (INRs), and healthy controls (HCs). Flow cytometry analysis and immunofluorescence microscopy showed increased platelet-CD4+ T cell aggregate formation in TNs compared to HCs during HIV-1 infection. However, the frequencies of platelet-CD4+ T cell aggregates decreased in IRs compared to TNs, but not in INRs, which have shown severe immunological dysfunction. Platelet-CD4+ T cell aggregate frequencies were positively correlated with HIV-1 viral load but negatively correlated with CD4+ T cell counts and CD4/CD8 ratios. Furthermore, we observed a higher expression of CD45RO, HIV co-receptors, HIV activation/exhaustion markers in platelet-CD4+ T cell aggregates, which was associated with HIV-1 permissiveness. High levels of caspase-1 and caspase-3, and low levels of Bcl-2 in platelet-CD4+ T cell aggregates imply the potential role in CD4+ T cell loss during HIV-1 infection. Furthermore, platelet-CD4+ T cell aggregates contained more HIV-1 gag viral protein and HIV-1 DNA than their platelet-free CD4+ T cell counterparts. The platelet-CD4+ T cell aggregate levels were positively correlated with plasma sCD163 and sCD14 levels. Our findings demonstrate that platelet-CD4+ T cell aggregate formation has typical characteristics of HIV-1 permissiveness and is related to immune activation during HIV-1 infection.
Collapse
Affiliation(s)
- Xiao-Peng Dai
- Medical School of Chinese People’s Liberation Army of China (PLA), Beijing, China
- Noncommissioned Officer School, Army Medical University, Shijiazhuang, China
| | - Feng-Ying Wu
- Medical School of Chinese People’s Liberation Army of China (PLA), Beijing, China
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Cheng Cui
- Noncommissioned Officer School, Army Medical University, Shijiazhuang, China
| | - Xue-Jiao Liao
- The Third People’s Hospital of Shenzhen, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yan-Mei Jiao
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Wen Song
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ji-Yuan Zhang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- *Correspondence: Fu-Sheng Wang, ; Ji-Yuan Zhang, ; Qing He,
| | - Qing He
- The Third People’s Hospital of Shenzhen, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Fu-Sheng Wang, ; Ji-Yuan Zhang, ; Qing He,
| | - Fu-Sheng Wang
- Medical School of Chinese People’s Liberation Army of China (PLA), Beijing, China
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese People’s Liberation Army of China (PLA) General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- *Correspondence: Fu-Sheng Wang, ; Ji-Yuan Zhang, ; Qing He,
| |
Collapse
|
17
|
Chebbo M, Duez C, Alessi MC, Chanez P, Gras D. Platelets: a potential role in chronic respiratory diseases? Eur Respir Rev 2021; 30:30/161/210062. [PMID: 34526315 PMCID: PMC9488457 DOI: 10.1183/16000617.0062-2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/05/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate cells known for their role in haemostasis and thrombosis. In recent years, an increasing number of observations have suggested that platelets are also immune cells and key modulators of immunity. They express different receptors and molecules that allow them to respond to pathogens, and to interact with other immune cells. Platelets were linked to the pathogenesis of some inflammatory disorders including respiratory diseases such as asthma and idiopathic pulmonary fibrosis. Here, we discuss the involvement of platelets in different immune responses, and we focus on their potential role in various chronic lung diseases. In addition to their essential role in haemostasis and thrombosis, platelets are strong modulators of different immune responses, and could be involved in the physiopathology of several chronic airway diseaseshttps://bit.ly/3cB6Xnj
Collapse
Affiliation(s)
| | | | - Marie C Alessi
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, CHU de la Timone, Laboratoire d'hématologie, Marseille, France
| | - Pascal Chanez
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, Hôpital NORD, Clinique des Bronches, Allergie et Sommeil, Marseille, France
| | - Delphine Gras
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France
| |
Collapse
|
18
|
Recent Advances in the Discovery and Function of Antimicrobial Molecules in Platelets. Int J Mol Sci 2021; 22:ijms221910230. [PMID: 34638568 PMCID: PMC8508203 DOI: 10.3390/ijms221910230] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/14/2022] Open
Abstract
The conventional function described for platelets is maintaining vascular integrity. Nevertheless, increasing evidence reveals that platelets can additionally play a crucial role in responding against microorganisms. Activated platelets release molecules with antimicrobial activity. This ability was first demonstrated in rabbit serum after coagulation and later in rabbit platelets stimulated with thrombin. Currently, multiple discoveries have allowed the identification and characterization of PMPs (platelet microbicidal proteins) and opened the way to identify kinocidins and CHDPs (cationic host defense peptides) in human platelets. These molecules are endowed with microbicidal activity through different mechanisms that broaden the platelet participation in normal and pathologic conditions. Therefore, this review aims to integrate the currently described platelet molecules with antimicrobial properties by summarizing the pathways towards their identification, characterization, and functional evaluation that have promoted new avenues for studying platelets based on kinocidins and CHDPs secretion.
Collapse
|
19
|
Hottz ED, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Bozza FA, Bozza PT. Platelet-leukocyte interactions in the pathogenesis of viral infections. Platelets 2021; 33:200-207. [PMID: 34260328 DOI: 10.1080/09537104.2021.1952179] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Evolving evidence demonstrates that platelets have major roles in viral syndromes through previously unrecognized viral sensing and effector functions. Activated platelets and increased platelet-leukocyte aggregates are observed in clinical and experimental viral infections. The mechanisms and outcomes of platelet-leukocyte interactions depend on the interacting leukocyte as well as on the pathogen and pathological conditions. In this review, we discuss the mechanisms involved in platelet interactions with leukocytes and its functions during viral infections. We focus on the contributions of human platelet-leukocyte interactions to pathophysiological and protective responses during viral infections of major global health relevance, including acquired immunodeficiency syndrome (AIDS), dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS), influenza pneumonia, and COVID-19.
Collapse
Affiliation(s)
- Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil.,Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Fernando A Bozza
- Laboratory of Clinical Research in Intensive Care Medicine, National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro , Brazil.,Intensive Care Medicine, D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Promoting platelets is a therapeutic option to combat severe viral infection of the lung. Blood Adv 2021; 4:1640-1642. [PMID: 32315397 PMCID: PMC7189284 DOI: 10.1182/bloodadvances.2020001669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
|
21
|
Platelets function as an acute viral reservoir during HIV-1 infection by harboring virus and T-cell complex formation. Blood Adv 2021; 4:4512-4521. [PMID: 32946568 DOI: 10.1182/bloodadvances.2020002420] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/14/2020] [Indexed: 12/22/2022] Open
Abstract
Platelets were recently found to harbor infectious HIV virions in infected individuals who are on antiretroviral treatment with poor CD4+ T-cell recovery. In this study, we screened platelets from recently infected individuals, before and after antiretroviral therapy, for the presence of virus and examined platelet activation, as well as CD4+ T-cell recovery. This was followed by in vitro studies assessing platelet-CD4+ T-cell complex formation as a contributing factor to viral transmission. HIV+ platelets were detected in 10 of 10 acutely infected individuals with no prior history of antiretroviral therapy. The percentage of HIV+ platelets dropped significantly after 3 months of antiretroviral therapy in all of the study participants. These individuals also demonstrated significant recovery of CD4+ T cells. Interestingly, the percentage of HIV+ platelets correlated positively with viral load but not with CD4+ T-cell count. Furthermore, we found that platelet activation with soluble CD40L or thrombin receptor activator peptide 6 (TRAP6) increased platelet-virus interactions in vitro. TRAP6-mediated interactions were reduced by platelet antagonists, aspirin, and R406. We demonstrated that platelets transmit the virus to CD4+ T cells, and this transinfection was abolished by inhibiting platelet-T-cell complex formation via exposure to an anti-CD62P antibody. Additionally, treatment with TRAP6 significantly increased the transinfection, which was also inhibited by aspirin and R206. These results reveal that platelets have the potential to promote HIV viral spread during the acute stage of infection, by harboring infectious virus transmitting infection to susceptible CD4+ T cells through complex formation.
Collapse
|
22
|
Allaoui A, Khawaja AA, Badad O, Naciri M, Lordkipanidzé M, Guessous F, Zaid Y. Platelet Function in Viral Immunity and SARS-CoV-2 Infection. Semin Thromb Hemost 2021; 47:419-426. [PMID: 33851385 DOI: 10.1055/s-0041-1726033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Platelets, as nonnucleated blood components, are classically recognized for their pivotal role in hemostasis. In recent years, however, accumulating evidence points to a nonhemostatic role for platelets, as active participants in the inflammatory and immune responses to microbial organisms in infectious diseases. This stems from the ability of activated platelets to secrete a plethora of immunomodulatory cytokines and chemokines, as well as directly interplaying with viral receptors. While much attention has been given to the role of the cytokine storm in the severity of the coronavirus disease 2019 (COVID-19), less is known about the contribution of platelets to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we give a brief overview on the platelet contribution to antiviral immunity and response during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Afaf Allaoui
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Akif A Khawaja
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Oussama Badad
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco.,Department of Plant, Southern Illinois University, Carbondale, Illinois
| | - Mariam Naciri
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montréal, Quebec, Canada.,Faculty of pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Fadila Guessous
- Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia.,Department of Biological Sciences, Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco.,Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco
| |
Collapse
|
23
|
Jeyaraman M, Muthu S, Khanna M, Jain R, Anudeep TC, Muthukanagaraj P, Siddesh SE, Gulati A, Satish AS, Jeyaraman N, Khanna V. Platelet lysate for COVID-19 pneumonia-a newer adjunctive therapeutic avenue. Stem Cell Investig 2021; 8:11. [PMID: 34268440 PMCID: PMC8256133 DOI: 10.21037/sci-2020-042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/27/2021] [Indexed: 02/05/2023]
Abstract
The linchpin for COVID-19 pathogenesis is the severe inflammatory process in the respiratory tract wherein the accumulation of excessive cytokines paves the way for a series of systemic hemodynamic alterations and mortality. The mortality rate is higher in individuals with co-morbidities and advancing age. The absence of a specific therapy is responsible for this uncontrolled spread and the significant mortality. This renders potential insight for considering biologics as a plausible option to repair and regenerate the affected lung tissue and pulverize the causative organism. The plausible role of megakaryocytes against invading microbes was not clearly understood. Platelet lysate is an acellular product consisting of regenerative molecules released from a cluster of platelets. It attenuates the changes caused by immune reactions in allogenic utility with the introduction of growth factors, cytokines, and proteins at supraphysiologic levels and thereby serves as a regenerative immunomodulatory agent to combat COVID-19. This platelet lysate can be used in nebulized form for such acute respiratory distress conditions in COVID-19 elderly patients. Platelet lysate may emerge as a pivotal player provided investigations pace up in this context. Here, we discuss how the platelet lysate can plausibly perquisite to relegate COVID-19. Undertaking prospective randomized controlled trials to prove its efficacy is the need of the hour in this pandemic scenario.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Sathish Muthu
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Orthopedics, Government Medical College & Hospital, Dindigul, Tamil Nadu, India
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Prasad Institute of Medical Science and Hospital, Lucknow, Uttar Pradesh, India
| | - Rashmi Jain
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Talagavadi Channaiah Anudeep
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Plastic Surgery, Topiwala National Medical College and BYL Nair Ch. Hospital, Mumbai, Maharashtra, India
| | - Purushothaman Muthukanagaraj
- Department of Internal Medicine & Psychiatry, SUNY-Upstate Binghamton Clinical Campus, Binghamton, New York, USA
| | | | - Arun Gulati
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Kalpana Chawla Government Medical College & Hospital, Karnal, Haryana, India
| | | | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Kasturba Medical College, MAHE Unievrsity, Manipal, Karnataka, India
| | - Venus Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Pathology, Prasad Institute of Medical Science and Hospital, Lucknow, Uttar Pradesh, India
| |
Collapse
|
24
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
25
|
Jeyaraman M, Ranjan R, Kumar R, Arora A, Chaudhary D, Ajay SS, Jain R. Cellular Therapy: Shafts of Light Emerging for COVID-19. Stem Cell Investig 2020; 7:11. [PMID: 32695804 PMCID: PMC7367471 DOI: 10.21037/sci-2020-022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
The COVID-19 pandemic has presented with debilitating respiratory consequences especially more pronounced in high risk individuals. Individuals with underlying systemic diseases are more prone and vulnerable to suffer severe consequences of SARS-CoV-2 infectivity. The pathophysiological changes identified cytokine storm mechanism for out setting the series of adverse clinical conditions. Thereby, associating it with high mortality rates. This warrants urgent consideration of divergent modalities such as the cellular therapy. Cellular therapy (CT) is a new medical paradigm wherein cellular material is administered to patients for therapeutic purposes. In this regard, mesenchymal stem cells (MSCs) have yielded the most promising results among stromal vascular fraction (SVF); placental cells; natural killer (NK) cell and platelet lysate respectively. Following the administration of the CT as per preferred route, these play pivotal role in modifying the microenvironment of the lung tissue with their distinct sets of mechanism. Evidences have shown how their immunomodulatory action repairs and prevents lung injury which in turn improvise the compliance of lungs. In this review article we have discussed these emerging novel approaches and their target step serving as a ray of hope to combat severe form of COVID-19. Currently these aren't approved for preventing or treating COVID-19 cases, however clinical trials are afoot to dispense the utmost understanding in terms of efficacy and safety concerns.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rakesh Kumar
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Arunabh Arora
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Dushyant Chaudhary
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | | | - Rashmi Jain
- School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
26
|
Page MJ, Pretorius E. A Champion of Host Defense: A Generic Large-Scale Cause for Platelet Dysfunction and Depletion in Infection. Semin Thromb Hemost 2020; 46:302-319. [PMID: 32279287 PMCID: PMC7339151 DOI: 10.1055/s-0040-1708827] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thrombocytopenia is commonly associated with sepsis and infections, which in turn are characterized by a profound immune reaction to the invading pathogen. Platelets are one of the cellular entities that exert considerable immune, antibacterial, and antiviral actions, and are therefore active participants in the host response. Platelets are sensitive to surrounding inflammatory stimuli and contribute to the immune response by multiple mechanisms, including endowing the endothelium with a proinflammatory phenotype, enhancing and amplifying leukocyte recruitment and inflammation, promoting the effector functions of immune cells, and ensuring an optimal adaptive immune response. During infection, pathogens and their products influence the platelet response and can even be toxic. However, platelets are able to sense and engage bacteria and viruses to assist in their removal and destruction. Platelets greatly contribute to host defense by multiple mechanisms, including forming immune complexes and aggregates, shedding their granular content, and internalizing pathogens and subsequently being marked for removal. These processes, and the nature of platelet function in general, cause the platelet to be irreversibly consumed in the execution of its duty. An exaggerated systemic inflammatory response to infection can drive platelet dysfunction, where platelets are inappropriately activated and face immunological destruction. While thrombocytopenia may arise by condition-specific mechanisms that cause an imbalance between platelet production and removal, this review evaluates a generic large-scale mechanism for platelet depletion as a repercussion of its involvement at the nexus of responses to infection.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
27
|
Campbell RA, Schwertz H, Hottz ED, Rowley JW, Manne BK, Washington AV, Hunter-Mellado R, Tolley ND, Christensen M, Eustes AS, Montenont E, Bhatlekar S, Ventrone CH, Kirkpatrick BD, Pierce KK, Whitehead SS, Diehl SA, Bray PF, Zimmerman GA, Kosaka Y, Bozza PT, Bozza FA, Weyrich AS, Rondina MT. Human megakaryocytes possess intrinsic antiviral immunity through regulated induction of IFITM3. Blood 2019; 133:2013-2026. [PMID: 30723081 PMCID: PMC6509546 DOI: 10.1182/blood-2018-09-873984] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023] Open
Abstract
Evolving evidence indicates that platelets and megakaryocytes (MKs) have unexpected activities in inflammation and infection; whether viral infections upregulate biologically active, antiviral immune genes in platelets and MKs is unknown, however. We examined antiviral immune genes in these cells in dengue and influenza infections, viruses that are global public health threats. Using complementary biochemical, pharmacological, and genetic approaches, we examined the regulation and function of interferon-induced transmembrane protein 3 (IFITM3), an antiviral immune effector gene not previously studied in human platelets and MKs. IFITM3 was markedly upregulated in platelets isolated from patients during clinical influenza and dengue virus (DENV) infections. Lower IFITM3 expression in platelets correlated with increased illness severity and mortality in patients. Administering a live, attenuated DENV vaccine to healthy subjects significantly increased platelet IFITM3 expression. Infecting human MKs with DENV selectively increased type I interferons and IFITM3. Overexpression of IFITM3 in MKs was sufficient to prevent DENV infection. In naturally occurring, genetic loss-of-function studies, MKs from healthy subjects harboring a homozygous mutation in IFITM3 (rs12252-C, a common single-nucleotide polymorphism in areas of the world where DENV is endemic) were significantly more susceptible to DENV infection. DENV-induced MK secretion of interferons prevented infection of bystander MKs and hematopoietic stem cells. Thus, viral infections upregulate IFITM3 in human platelets and MKs, and IFITM3 expression is associated with adverse clinical outcomes. These observations establish, for the first time, that human MKs possess antiviral functions, preventing DENV infection of MKs and hematopoietic stem cells after local immune signaling.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
| | - Hansjorg Schwertz
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
- Rocky Mountain Center for Occupational and Environmental Health, University of Utah, Salt Lake City, UT
| | - Eugenio D Hottz
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Instituto Nacional de Infectologia Evandro Chagas and
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jesse W Rowley
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
| | | | - A Valance Washington
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
- Department of Internal Medicine, Universidad Central del Caribe, Bayamón, Puerto Rico
| | - Robert Hunter-Mellado
- Department of Biology, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
- Department of Internal Medicine, Universidad Central del Caribe, Bayamón, Puerto Rico
| | - Neal D Tolley
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | | | - Alicia S Eustes
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Emilie Montenont
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Seema Bhatlekar
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Cassandra H Ventrone
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, VT
| | - Beth D Kirkpatrick
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, VT
| | - Kristen K Pierce
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, VT
| | - Stephen S Whitehead
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sean A Diehl
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, VT
| | - Paul F Bray
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
| | - Guy A Zimmerman
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Patricia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- Instituto Nacional de Infectologia Evandro Chagas and
- Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil; and
| | - Andrew S Weyrich
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine and
- Department of Internal Medicine, George E. Wahlen Veterans Affairs Medical Center and Geriatric Research, Education, and Clinical Center, Salt Lake City, UT
| |
Collapse
|
28
|
Lv L, Li Y, Fan X, Xie Z, Liang H, Shen T. HCV coinfection aggravated the decrease of platelet counts, but not mean platelet volume in chronic HIV-infected patients. Sci Rep 2018; 8:17497. [PMID: 30504858 PMCID: PMC6269489 DOI: 10.1038/s41598-018-35705-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/09/2018] [Indexed: 01/06/2023] Open
Abstract
Either HIV or HCV monoinfection could result in an abnormal status of platelets. As two key indicators reflecting activation and function of platelets, the changes of platelet counts and mean platelet volume (MPV) in HIV/HCV-coinfected patients have not been clearly identified. In the present study, a total of 318 former plasma donors were investigated in 2006, and 66% (201 individuals) of primary recruiters were followed up in 2014. By horizontal comparison in 2006, the decrease of platelet counts in HIV/HCV coinfection was greater than that in HIV or HCV monoinfection. MPV scores were lower in HIV monoinfection compared with healthy controls, while no difference was found in HIV/HCV coinfection. Platelet counts were shown to be negatively correlated with MPV scores in total recruited population (r = 0.432, P < 0.001). Interestingly, by comparison of data from two time points of 2006 and 2014, significant decrease of platelets (P = 0.004) and increase of MPV (P = 0.004) were found only in HCV monoinfected patients, which may associate with slow progression of hepatic fibrosis induced by chronic HCV infection. Nonetheless, no significant changes of platelet counts and MPV were found from 2006 to 2014 in coinfected patients. In conclusion, HCV coinfection aggravated the decrease of platelet counts, but not MPV score in chronic HIV infection. MPV showed poor applicability in reflecting the status of platelets in HIV/HCV-coinfected patients.
Collapse
Affiliation(s)
- Linting Lv
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yuantao Li
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xueying Fan
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhe Xie
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Hua Liang
- State Key laboratory of Infectious Disease Prevention and Control (SKLID), National Center for AIDS/STD Control and Prevention, China CDC, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, 102206, China.
| | - Tao Shen
- Department of Microbiology and Center of Infectious Disease, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
29
|
Adenosine signaling and adenosine deaminase regulation of immune responses: impact on the immunopathogenesis of HIV infection. Purinergic Signal 2018; 14:309-320. [PMID: 30097807 DOI: 10.1007/s11302-018-9619-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Infection by human immunodeficiency virus (HIV) causes the acquired immune deficiency syndrome (AIDS), which has devastating effects on the host immune system. HIV entry into host cells and subsequent viral replication induce a proinflammatory response, hyperactivating immune cells and leading them to death, disfunction, and exhaustion. Adenosine is an immunomodulatory molecule that suppresses immune cell function to protect tissue integrity. The anti-inflammatory properties of adenosine modulate the chronic inflammation and immune activation caused by HIV. Lack of adenosine contributes to pathogenic events in HIV infection. However, immunosuppression by adenosine has its shortcomings, such as impairing the immune response, hindering the elimination of the virus and control of viral replication. By attempting to control inflammation, adenosine feeds a pathogenic cycle affecting immune cells. Deamination of adenosine by ADA (adenosine deaminase) counteracts the negative effects of adenosine in immune cells, boosting the immune response. This review comprises the connection between adenosinergic system and HIV immunopathogenesis, exploring defects in immune cell function and the role of ADA in protecting these cells against damage.
Collapse
|
30
|
Melo CFOR, Delafiori J, Dabaja MZ, de Oliveira DN, Guerreiro TM, Colombo TE, Nogueira ML, Proenca-Modena JL, Catharino RR. The role of lipids in the inception, maintenance and complications of dengue virus infection. Sci Rep 2018; 8:11826. [PMID: 30087415 PMCID: PMC6081433 DOI: 10.1038/s41598-018-30385-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
Dengue fever is a viral condition that has become a recurrent issue for public health in tropical countries, common endemic areas. Although viral structure and composition have been widely studied, the infection phenotype in terms of small molecules remains poorly established. This contribution providing a comprehensive overview of the metabolic implications of the virus-host interaction using a lipidomic-based approach through direct-infusion high-resolution mass spectrometry. Our results provide further evidence that lipids are part of both the immune response upon Dengue virus infection and viral infection maintenance mechanism in the organism. Furthermore, the species described herein provide evidence that such lipids may be part of the mechanism that leads to blood-related complications such as hemorrhagic fever, the severe form of the disease.
Collapse
Affiliation(s)
| | - Jeany Delafiori
- INNOVARE Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Mohamad Ziad Dabaja
- INNOVARE Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Diogo Noin de Oliveira
- INNOVARE Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Tatiane Melina Guerreiro
- INNOVARE Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Tatiana Elias Colombo
- School of Medicine from São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | | | - Jose Luiz Proenca-Modena
- Laboratory of Study of Emerging Viruses (LEVE), Department of Genetic, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Rodrigo Ramos Catharino
- INNOVARE Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil.
| |
Collapse
|
31
|
Seyoum M, Enawgaw B, Melku M. Human blood platelets and viruses: defense mechanism and role in the removal of viral pathogens. Thromb J 2018; 16:16. [PMID: 30026673 PMCID: PMC6048695 DOI: 10.1186/s12959-018-0170-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/18/2018] [Indexed: 12/19/2022] Open
Abstract
Platelets are small non-nucleated cell fragments and the second most abundant cell that play crucial role in managing vascular integrity and regulating hemostasis. Recent finding shows, beyond its hemostatic function platelets also play a main role in fighting against pathogen including viruses. With their receptors, platelet interacts with viral pathogen and this interaction between platelets and viral pathogens result in activation of platelets. Activated platelet releases different molecules that have antiviral activity including kinocidins and other platelet microbicidal peptides. In addition, activated platelet has antiviral role by different mechanism including; phagocytosis of viral pathogen, produce reactive oxygen species and interact with and activate other immune cells. In other side, antiplatelet treatments are one of defending mechanism of viral pathogen. This narrative review summarizes what is known regarding the role of human platelets in fighting viral pathogen.
Collapse
Affiliation(s)
- Masresha Seyoum
- University of Gondar hospital, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Bamlaku Enawgaw
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Melku
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
32
|
Hottz ED, Bozza FA, Bozza PT. Platelets in Immune Response to Virus and Immunopathology of Viral Infections. Front Med (Lausanne) 2018; 5:121. [PMID: 29761104 PMCID: PMC5936789 DOI: 10.3389/fmed.2018.00121] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/12/2018] [Indexed: 01/04/2023] Open
Abstract
Platelets are essential effector cells in hemostasis. Aside from their role in coagulation, platelets are now recognized as major inflammatory cells with key roles in the innate and adaptive arms of the immune system. Activated platelets have key thromboinflammatory functions linking coagulation to immune responses in various infections, including in response to virus. Recent studies have revealed that platelets exhibit several pattern recognition receptors (PRR) including those from the toll-like receptor, NOD-like receptor, and C-type lectin receptor family and are first-line sentinels in detecting and responding to pathogens in the vasculature. Here, we review the main mechanisms of platelets interaction with viruses, including their ability to sustain viral infection and replication, their expression of specialized PRR, and activation of thromboinflammatory responses against viruses. Finally, we discuss the role of platelet-derived mediators and platelet interaction with vascular and immune cells in protective and pathophysiologic responses to dengue, influenza, and human immunodeficiency virus 1 infections.
Collapse
Affiliation(s)
- Eugenio D Hottz
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Departamento de Bioquimica, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Fernando A Bozza
- Laboratório de Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Xu J, Yi J, Zhang H, Feng F, Gu S, Weng L, Zhang J, Chen Y, An N, Liu Z, An Q, Yin W, Hu X. Platelets directly regulate DNA damage and division of Staphylococcus aureus. FASEB J 2018; 32:3707-3716. [PMID: 29430991 DOI: 10.1096/fj.201701190r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Platelets (PLTs) are classically used in the clinical setting to maintain hemostasis. Recent evidence supports important roles for PLTs in host inflammatory and immune responses, and PLT-rich plasma has been demonstrated to inhibit the growth of bacteria in vitro and in vivo; however, few studies have examined whether PLTs can inhibit bacterial growth directly, and related mechanisms have not been elucidated further. Accordingly, in this study, we evaluated the effects of PLTs on bacterial growth. We washed and purified PLTs from peripheral blood, then confirmed that PLTs significantly inhibited the growth of Staphylococcus aureus when cocultured in vitro. Moreover, PLTs damaged DNA and blocked cell division in S. aureus. During coculture, PLT-derived TGF-β1 was dramatically down-regulated compared with that in PLT culture alone, and the addition of TGF-β1 to the coculture system promoted the inhibition of PLTs on S. aureus. Analysis of a murine S. aureus infection model demonstrated that the depletion of PLTs exacerbated the severity of infection, whereas the transfusion of PLTs alleviated this infection. Our observations demonstrate that PLTs could directly inhibit the growth of S. aureus by damaging DNA and blockage cell division, and that PLT-derived TGF-β1 may play an important role in this machinery.-Xu, J., Yi, J., Zhang, H., Feng, F., Gu, S., Weng, L., Zhang, J., Chen, Y., An, N., Liu, Z., An, Q., Yin, W., Hu, X. Platelets directly regulate DNA damage and division of Staphylococcus aureus.
Collapse
Affiliation(s)
- Jinmei Xu
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jing Yi
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Huijie Zhang
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Fan Feng
- Department of Digestive Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shunli Gu
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lihong Weng
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Jing Zhang
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yaozhen Chen
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ning An
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zheng Liu
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Qunxing An
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wen Yin
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xingbin Hu
- Department of Transfusion Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Tobolowsky FA, Wada N, Martinez-Maza O, Magpantay L, Koletar SL, Palella FJ, Brown TT, Lake JE. Brief report: Circulating markers of fibrosis are associated with immune reconstitution status in HIV-infected men. PLoS One 2018; 13:e0191606. [PMID: 29381717 PMCID: PMC5790272 DOI: 10.1371/journal.pone.0191606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/08/2018] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Lymphoid tissue fibrosis may contribute to incomplete immune reconstitution on antiretroviral therapy (ART) via local CD4+ T lymphocyte (CD4) depletion. Hyaluronic acid (HA) increases with fibrotic burden. CXCL4 concentrations increase in response to pro-fibrotic stimuli, but lower CXCL4 concentrations in HIV-infected individuals may reflect successful immune evasion by HIV. We investigated relationships between circulating HA and CXCL4 concentrations and immune reconstitution on ART in HIV-infected Multicenter AIDS Cohort Study participants. METHODS HIV-infected men on ART for >1 year with cryopreserved plasma samples and suppressed post-ART HIV-1 RNA were included. Men with post-ART CD4 <200 cells/mm3 were defined as immunologic non-responders (n = 25). Age-/race-matched men with post-ART CD4 >500 cells/mm3 served as controls (n = 49). HA and CXCL4 concentrations were measured via ELISA. RESULTS Median pre-ART CD4 was 297 cells/mm3 for non-responders vs 386 cells/mm3 for controls. Median post-ART CD4 was 141 cells/mm3 for non-responders and 815 cells/mm3 for controls. HIV infection duration was 23 years, with median time on ART 13 years for non-responders vs 11 years for controls. Pre-ART HA and CXCL4 concentrations did not vary by eventual immune reconstitution status. Post-ART HA concentrations tended to be higher (85 vs 36 ng/mL, p = 0.07) and CXCL4 concentrations were lower (563 vs 1459 ng/mL, p = 0.01) among non-responders. Among men with paired pre-/post-ART samples, non-responders had greater HA increases and CXCL4 decreases than controls (HA: 50 vs 12 ng/mL, p = 0.04; CXCL4: -1258 vs -405 ng/mL, p = 0.01). CONCLUSIONS Higher circulating concentrations of HA and lower concentrations of CXCL4 are associated with failure of immune reconstitution on ART.
Collapse
Affiliation(s)
- F. A. Tobolowsky
- Department of Internal Medicine, Division of Infectious Diseases, University of Colorado, Denver, Colorado, United States of America
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| | - N. Wada
- Department of Epidemiology, Division of General Epidemiology and Methodology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - O. Martinez-Maza
- Department of Obstetrics and Gynecology, Epidemiology, Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - L. Magpantay
- Department of Obstetrics and Gynecology, Epidemiology, Microbiology, Immunology & Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - S. L. Koletar
- Department of Internal Medicine, Division of Infectious Diseases, Ohio State University, Columbus, Ohio, United States of America
| | - F. J. Palella
- Department of Internal Medicine, Division of Infectious Diseases, Northwestern University, Chicago, Illinois, United States of America
| | - T. T. Brown
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - J. E. Lake
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Department of Internal Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
35
|
Mietsch M, Sauermann U, Mätz-Rensing K, Klippert A, Daskalaki M, Stolte-Leeb N, Stahl-Hennig C. Revisiting a quarter of a century of simian immunodeficiency virus (SIV)-associated cardiovascular diseases at the German Primate Center. Primate Biol 2017; 4:107-115. [PMID: 32110698 PMCID: PMC7041533 DOI: 10.5194/pb-4-107-2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Human immunodeficiency virus (HIV) comorbidities have become
clinically more important due to antiretroviral therapy. Although therapy
increases life expectancy, it does not completely suppress immune activation
and its associated complications. The simian immunodeficiency virus
(SIV)-infected rhesus macaque (Macaca mulatta) represents a valuable
model for the investigation of SIV-associated diseases. Although
cardiovascular (CV) changes are common in HIV-infected patients, there are
only a few reports on the incidence of CV findings in SIV-infected animals.
In addition, potential associations between pathohistological findings and
hematological parameters are still unclear. We therefore conducted a retrospective analysis of 195 SIV-infected rhesus
macaques that were euthanized with AIDS-related symptoms at the German
Primate Center, Goettingen, over a 25-year period. Pathological findings
were correlated with hematological data. The main findings included myocarditis (12.8 %), endocarditis
(9.7 %),
and arteriopathy (10.3 %) in various organs. Thrombocytopenia occurred
more frequently in macaques with endocarditis or arteriopathy than in
macaques without CV disease (80 % in animals with endocarditis, 60 %
in animals with arteriopathy, p<0.0001 and p=0.0016, respectively). Further investigations of the interaction between coagulation markers,
proinflammatory cytokines, and biomarkers associated with endothelial
dysfunction (e.g., D-dimers) and histological data (vascular wall structure)
may unravel the mechanisms underlying HIV/SIV-associated CV comorbidities.
Collapse
Affiliation(s)
- Matthias Mietsch
- Unit of Infection Models, German Primate Center, 37077 Goettingen, Germany.,These authors contributed equally to this work
| | - Ulrike Sauermann
- Unit of Infection Models, German Primate Center, 37077 Goettingen, Germany.,These authors contributed equally to this work
| | | | - Antonina Klippert
- Unit of Infection Models, German Primate Center, 37077 Goettingen, Germany
| | - Maria Daskalaki
- Unit of Infection Models, German Primate Center, 37077 Goettingen, Germany
| | - Nicole Stolte-Leeb
- Unit of Infection Models, German Primate Center, 37077 Goettingen, Germany
| | | |
Collapse
|
36
|
Abstract
Virus–platelet interplay is complex. Diverse virus types have been shown to associate with numerous distinct platelet receptors. This association can benefit the virus or the host, and thus the platelet is somewhat of a renegade. Evidence is accumulating to suggest that viruses are capable of entering platelets. For at least one type of RNA virus (dengue virus), the platelet has the necessary post-translational and packaging machinery required for production of replicative viral progeny. As a facilitator of immunity, the platelet also participates in eradicating the virus by direct and indirect mechanisms involving presentation of the pathogen to the innate and adaptive immune systems, thus enhancing inflammation by release of cytokines and other agonists. Virus-induced thrombocytopenia is caused by tangential imbalance of thrombopoeisis, autoimmunity, and loss of platelet function and integrity.
Collapse
|
37
|
Abstract
Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Bhanu K Manne
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA
| | | | - Matthew T Rondina
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA.,c Department of Internal Medicine , Salt Lake City , Utah , USA.,d The GRECC, George E. Wahlen Salt Lake City VAMC , Salt Lake City , Utah , USA
| |
Collapse
|
38
|
Parker ZF, Rux AH, Riblett AM, Lee FH, Rauova L, Cines DB, Poncz M, Sachais BS, Doms RW. Platelet Factor 4 Inhibits and Enhances HIV-1 Infection in a Concentration-Dependent Manner by Modulating Viral Attachment. AIDS Res Hum Retroviruses 2016; 32:705-17. [PMID: 26847431 DOI: 10.1089/aid.2015.0344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Platelet factor 4 (PF4) has been recently shown to inhibit infection by a broad range of human immunodeficiency virus type 1 (HIV-1) isolates in vitro. We found that the inhibitory effects of PF4 are limited to a defined concentration range where PF4 exists largely in a monomeric state. Under these conditions, PF4 bound the HIV-1 envelope protein and inhibited HIV-1 attachment to the cell surface. However, as concentrations increased to the point where PF4 exists largely in tetrameric or higher-order forms, viral infection in vitro was enhanced. Enhancement could be inhibited by mutations in PF4 that shift the oligomeric equilibrium toward the monomeric state, or by using soluble glycosaminoglycans (GAGs) to which tetrameric PF4 avidly binds. We conclude that at physiologically relevant concentrations, oligomeric PF4 enhances infection by HIV-1 by interacting with the viral envelope protein as well as cell surface GAGs, enhancing virus attachment to the cell surface. This effect was not specific to HIV-1, as enhancement was seen with some but not all other viruses tested. The biphasic effects of PF4 on HIV-1 infection suggest that native PF4 will not be a useful antiviral agent and that PF4 could contribute to the hematologic abnormalities commonly seen in HIV-infected individuals by enhancing virus infection in the bone marrow.
Collapse
Affiliation(s)
- Zahra F. Parker
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Ann H. Rux
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Amber M. Riblett
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Fang-Hua Lee
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Lubica Rauova
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Douglas B. Cines
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Mortimer Poncz
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Bruce S. Sachais
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- New York Blood Center, New York, New York
| | - Robert W. Doms
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Departments of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Effect of antiretroviral therapy in thromboregulation through the hydrolysis of adenine nucleotides in platelets of HIV patients. Biomed Pharmacother 2016; 79:321-8. [PMID: 27044844 DOI: 10.1016/j.biopha.2016.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 07/08/2015] [Accepted: 02/23/2016] [Indexed: 01/16/2023] Open
Abstract
The human immunodeficiency virus (HIV) infection results in biochemical and vascular dysfunctions. The highly active antiretroviral therapy (HAART) markedly reduces mortality and opportunistic diseases associated with acquired immunodeficiency syndrome (AIDS). This increased survival time predisposes the development of cardiovascular diseases. Platelets present purinergic system ectoenzymes such as E-NTPDase, E-5'-nucleotidase and E-ADA on its surface. In view of this, the aim of this study was to evaluate the activity of these ectoenzymes in platelets as well as the platelet aggregation and lipid profile of patients with HIV infection and also patients receiving HAART. The results showed an increase in the E-NTPDase activity for ATP hydrolysis in the HIV group compared with the control group and the HIV/HAART group. When assessing the activity E-NTPDase hydrolysis to ADP, the results revealed an increase in activity in the HIV group when compared to the control group, and a decrease in activity when in the HIV/HAART group when compared to the control and HIV groups. The activity of E-5'-nucleotidase revealed an increase in AMP hydrolysis in the HIV group, as the results from control and HIV/HAART groups showed no statistical difference. Regarding the E-ADA activity, the HIV and HIV/HAART groups revealed a decreased deamination of adenosine when compared with the control group. Furthermore, we observed an increased platelet aggregation of HIV/HAART group compared with the control group. Thus, our results suggest that antiretroviral treatment against HIV has a significant effect on the activity of purinergic system ectoenzymes demonstrating that thromboregulation is involved in the process.
Collapse
|
40
|
Ahamed J, Terry H, Choi ME, Laurence J. Transforming growth factor-β1-mediated cardiac fibrosis: potential role in HIV and HIV/antiretroviral therapy-linked cardiovascular disease. AIDS 2016; 30:535-42. [PMID: 26605511 PMCID: PMC4738098 DOI: 10.1097/qad.0000000000000982] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
HIV infection elevates the incidence of cardiovascular disease (CVD) independent of traditional risk factors. Autopsy series document cardiac inflammation and endomyocardial fibrosis in the HIV+ treatment naïve, and gadolinium enhancement magnetic resonance imaging has identified prominent myocardial fibrosis in the majority of HIV+ individuals despite use of suppressive antiretroviral therapies (ART). The extent of such disease may correlate with specific ART regimens. For example, HIV-infected patients receiving ritonavir (RTV)-boosted protease inhibitors have the highest prevalence of CVD, and RTV-exposed rodents exhibit cardiac dysfunction coupled with cardiac and vascular fibrosis, independent of RTV-mediated lipid alterations. We recently showed that platelet transforming growth factor (TGF)-β1 is a key contributor to cardiac fibrosis in murine models. We hypothesize that in the HIV+/ART naïve, cardiac fibrosis is a consequence of proinflammatory cytokine and/or ART-linked platelet activation with release of TGF-β1. Resultant TGF-β1/Smad signaling would promote collagen synthesis and organ fibrosis. We document these changes in a pilot immunohistochemical evaluation of cardiac tissue from two ART-naive pediatric AIDS patients. In terms of ART, we showed that RTV inhibits immunoproteasome degradation of TRAF6, a nuclear adapter signaling molecule critical to the regulation of proinflammatory cytokine signaling pathways involved in osteoclast differentiation and accelerated osteoporosis. We now present a model illustrating how RTV could similarly amplify TGF-β1 signaling in the promotion of cardiac fibrosis and accelerated CVD. Supportive clinical data correlate RTV use with elevation of NT-proBNP, a biomarker for CVD. We discuss potential interventions involving intrinsic modulators of inflammation and collagen degradation, including carbon monoxide-based therapeutics.
Collapse
Affiliation(s)
- Jasimuddin Ahamed
- aDivision of Hematology and Medical OncologybDivision of Nephrology and Hypertension, Weill Cornell Medical College, New YorkcCardiovascular Biology Research Program, Oklahoma Medical Research Foundation and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | | | | |
Collapse
|
41
|
Kiebala M, Singh MV, Piepenbrink MS, Qiu X, Kobie JJ, Maggirwar SB. Platelet Activation in Human Immunodeficiency Virus Type-1 Patients Is Not Altered with Cocaine Abuse. PLoS One 2015; 10:e0130061. [PMID: 26076359 PMCID: PMC4467977 DOI: 10.1371/journal.pone.0130061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 12/30/2022] Open
Abstract
Recent work has indicated that platelets, which are anucleate blood cells, significantly contribute to inflammatory disorders. Importantly, platelets also likely contribute to various inflammatory secondary disorders that are increasingly associated with Human Immunodeficiency Virus Type-1 (HIV) infection including neurological impairments and cardiovascular complications. Indeed, HIV infection is often associated with increased levels of platelet activators. Additionally, cocaine, a drug commonly abused by HIV-infected individuals, leads to increased platelet activation in humans. Considering that orchestrated signaling mechanisms are essential for platelet activation, and that nuclear factor-kappa B (NF-κB) inhibitors can alter platelet function, the role of NF-κB signaling in platelet activation during HIV infection warrants further investigation. Here we tested the hypothesis that inhibitory kappa B kinase complex (IKK) activation would be central for platelet activation induced by HIV and cocaine. Whole blood from HIV-positive and HIV-negative individuals, with or without cocaine abuse was used to assess platelet activation via flow cytometry whereas IKK activation was analyzed by performing immunoblotting and in vitro kinase assays. We demonstrate that increased platelet activation in HIV patients, as measured by CD62P expression, is not altered with reported cocaine use. Furthermore, cocaine and HIV do not activate platelets in whole blood when treated ex vivo. Finally, HIV-induced platelet activation does not involve the NF-κB signaling intermediate, IKKβ. Platelet activation in HIV patients is not altered with cocaine abuse. These results support the notion that non-IKK targeting approaches will be better suited for the treatment of HIV-associated inflammatory disorders.
Collapse
Affiliation(s)
- Michelle Kiebala
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| | - Meera V. Singh
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Michael S. Piepenbrink
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - James J. Kobie
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Sanjay B. Maggirwar
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
42
|
Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 2015; 26:311-27. [DOI: 10.1016/j.cytogfr.2014.11.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
|
43
|
Chabert A, Hamzeh-Cognasse H, Pozzetto B, Cognasse F, Schattner M, Gomez RM, Garraud O. Human platelets and their capacity of binding viruses: meaning and challenges? BMC Immunol 2015; 16:26. [PMID: 25913718 PMCID: PMC4411926 DOI: 10.1186/s12865-015-0092-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/03/2015] [Indexed: 01/16/2023] Open
Abstract
Blood platelets are first aimed at ensuring primary hemostasis. Beyond this role, they have been acknowledged as having functions in the maintenance of the vascular arborescence and, more recently, as being also innate immune cells, devoted notably to the detection of danger signals, of which infectious ones. Platelets express pathogen recognition receptors that can sense bacterial and viral moieties. Besides, several molecules that bind epithelial or sub-endothelial molecules and, so forth, are involved in hemostasis, happen to be able to ligate viral determinants, making platelets capable of either binding viruses or even to be infected by some of them. Further, as platelets express both Fc-receptors for Ig and complement receptors, they also bind occasionally virus-Ig or virus-Ig-complement immune complexes. Interplays of viruses with platelets are very complex and viral infections often interfere with platelet number and functions. Through a few instances of viral infections, the present review aims at presenting some of the most important interactions from pathophysiological and clinical points of view, which are observed between human viruses and platelets.
Collapse
Affiliation(s)
- Adrien Chabert
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France.
| | | | - Bruno Pozzetto
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,Service des Agents infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France.
| | - Fabrice Cognasse
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,EFS Auvergne-Loire, 42023, Saint-Etienne, France.
| | - Mirta Schattner
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, ANM-CONICET, Buenos Aires, Argentina.
| | - Ricardo M Gomez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, UNLP-CONICET, La Plata, Argentina.
| | - Olivier Garraud
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,Institut National de la Transfusion Sanguine, 75015, Paris, France. .,INTS, 6 rue Alexandre-Cabanel, 75015, Paris, France.
| |
Collapse
|
44
|
Abstract
While the interactions between Gram-positive bacteria and platelets have been well characterized, there is a paucity of data on the interaction between other pathogens and platelets. However, thrombocytopenia is a common feature with many infections especially viral hemorrhagic fever. The little available data on these interactions indicate a similarity with bacteria-platelet interactions with receptors such as FcγRIIa and Toll-Like Receptors (TLR) playing key roles with many pathogens. This review summarizes the known interactions between platelets and pathogens such as viruses, fungi and parasites.
Collapse
Affiliation(s)
- Ana Lopez Alonso
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland , Dublin , Ireland
| | | |
Collapse
|
45
|
El-Menshawy N, Eissa M, Farag R, Aboalyazed A. CD235a (Glycophorin-A) Is the Most Predictive Value Among Different Circulating Cellular Microparticles in Thrombocytopenic Human Immunodeficiency Virus Type 1. J Clin Lab Anal 2015; 30:235-43. [PMID: 25716234 DOI: 10.1002/jcla.21842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 01/09/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study was conducted to assess different cellular microparticles (MPs) in thrombocytopenic human immunodeficiency virus type 1 and their significance as disease activity markers. METHODS Thirty-five thrombocytopenic human immunodeficiency diseases and 25 healthy controls with matched age and sex were selected. Viral load was quantitated by COBAS real-time polymerase reaction (PCR) assessment of absolute T-cell subsets CD4, CD8 as a disease progress marker. Platelet MPs, platelet-derived monocyte MPs (CD42a, CD61), erythrocyte MP (CD235a), monocytic MP (CD14), and platelet activity MPs (CD62P, PAC-1) were assessed by multicolor flow cytometry FACSCalibur, while platelet functions were assessed by platelet function analyzer (PFA-100). CD42a, CD61, and platelet activity index represented by PAC-1 and CD62. RESULTS P-selectin in HIV-infected patient samples were significantly greater (P < 0.001) than among controls. There was a negative correlation between the proportion of PAC-1 and CD62 P-selectin-positive MPs and levels of CD4(+) T-cell counts (r = -0.403, P = 0.016; r = -0.438, P = 0.008), respectively. There was a negative correlation between collagen-ADP and levels of CD4(+) T-cell counts (r = -0.368, P = 0.03). There was a significant high expression level of CD14 monocyte MPs in patients than controls (P < 0.0001), overexpression of CD235a (P < 0.0001), and no correlation between CD14 and CD4, whereas there was a significant negative correlation with CD235a (r = -0.394, P = 0.019). A linear regression analysis of CD4 as a disease progression marker with other variable indicators in HIV patients showed that CD235a could be the most sensitive predictor similar to CD4. CONCLUSION Different cellular MPs and platelets activated in HIV patients could have a role in thrombotic events in these patients.
Collapse
Affiliation(s)
- Nadia El-Menshawy
- Clinical Pathology Department, Hematology Unit, Mansoura Medical School, Mansoura University, Egypt
| | - Mohammed Eissa
- Faculty of Medicine, Clinical Pathology Department, Zagazig University, Egypt.,College of Medicine, Pathology Department, King Khaled University, Abha, KSA
| | - Raghada Farag
- Tropical Medical Department, Mansoura Medical School, Mansoura University, Egypt
| | - Ahmed Aboalyazed
- Community Medicine Department, Mansoura Medical School, Mansoura University, Egypt
| |
Collapse
|
46
|
Ali RA, Wuescher LM, Worth RG. Platelets: essential components of the immune system. CURRENT TRENDS IN IMMUNOLOGY 2015; 16:65-78. [PMID: 27818580 PMCID: PMC5096834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Platelets are anucleate cell fragments known for their central role in coagulation and vascular integrity. However, it is becoming increasingly clear that platelets contribute to diverse immunological processes extending beyond the traditional view of platelets as fragmentary mediators of hemostasis and thrombosis. There is recent evidence that platelets participate in: 1) intervention against microbial threats; 2) recruitment and promotion of innate effector cell functions; 3) modulating antigen presentation; and 4) enhancement of adaptive immune responses. In this way, platelets should be viewed as the underappreciated orchestrator of the immune system. This review will discuss recent and historical evidence regarding how platelets influence both innate and adaptive immune responses.
Collapse
|
47
|
Assinger A. Platelets and infection - an emerging role of platelets in viral infection. Front Immunol 2014; 5:649. [PMID: 25566260 PMCID: PMC4270245 DOI: 10.3389/fimmu.2014.00649] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/05/2014] [Indexed: 12/23/2022] Open
Abstract
Platelets are anucleate blood cells that play a crucial role in the maintenance of hemostasis. While platelet activation and elevated platelet counts (thrombocytosis) are associated with increased risk of thrombotic complications, low platelet counts (thrombocytopenia) and several platelet function disorders increase the risk of bleeding. Over the last years, more and more evidence has emerged that platelets and their activation state can also modulate innate and adaptive immune responses and low platelet counts have been identified as a surrogate marker for poor prognosis in septic patients. Viral infections often coincide with platelet activation. Host inflammatory responses result in the release of platelet activating mediators and a pro-oxidative and pro-coagulant environment, which favors platelet activation. However, viruses can also directly interact with platelets and megakaryocytes and modulate their function. Furthermore, platelets can be activated by viral antigen-antibody complexes and in response to some viruses B-lymphocytes also generate anti-platelet antibodies. All these processes contributing to platelet activation result in increased platelet consumption and removal and often lead to thrombocytopenia, which is frequently observed during viral infection. However, virus-induced platelet activation does not only modulate platelet count but also shape immune responses. Platelets and their released products have been reported to directly and indirectly suppress infection and to support virus persistence in response to certain viruses, making platelets a double-edged sword during viral infections. This review aims to summarize the current knowledge on platelet interaction with different types of viruses, the viral impact on platelet activation, and platelet-mediated modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Alice Assinger
- Department of Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria ; Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
48
|
Van Breedam W, Pöhlmann S, Favoreel HW, de Groot RJ, Nauwynck HJ. Bitter-sweet symphony: glycan-lectin interactions in virus biology. FEMS Microbiol Rev 2014; 38:598-632. [PMID: 24188132 PMCID: PMC7190080 DOI: 10.1111/1574-6976.12052] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 09/27/2013] [Accepted: 10/14/2013] [Indexed: 01/01/2023] Open
Abstract
Glycans are carbohydrate modifications typically found on proteins or lipids, and can act as ligands for glycan-binding proteins called lectins. Glycans and lectins play crucial roles in the function of cells and organs, and in the immune system of animals and humans. Viral pathogens use glycans and lectins that are encoded by their own or the host genome for their replication and spread. Recent advances in glycobiological research indicate that glycans and lectins mediate key interactions at the virus-host interface, controlling viral spread and/or activation of the immune system. This review reflects on glycan-lectin interactions in the context of viral infection and antiviral immunity. A short introduction illustrates the nature of glycans and lectins, and conveys the basic principles of their interactions. Subsequently, examples are discussed highlighting specific glycan-lectin interactions and how they affect the progress of viral infections, either benefiting the host or the virus. Moreover, glycan and lectin variability and their potential biological consequences are discussed. Finally, the review outlines how recent advances in the glycan-lectin field might be transformed into promising new approaches to antiviral therapy.
Collapse
Affiliation(s)
- Wander Van Breedam
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
| | - Herman W. Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Raoul J. de Groot
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Hans J. Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
49
|
|
50
|
Nkambule BB, Davison G, Ipp H. The value of flow cytometry in the measurement of platelet activation and aggregation in human immunodeficiency virus infection. Platelets 2014; 26:250-7. [PMID: 24831969 DOI: 10.3109/09537104.2014.909021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human immunodeficiency deficiency virus (HIV) infection is associated with chronic inflammation and an increased risk of thrombotic events. Activated platelets (PLTs) play an important role in both thrombosis and inflammation, and HIV has been shown to induce PLT activation by both direct and indirect mechanisms. P-selectin (CD62P) is a well-described marker of PLT activation, and PLT glycoprotein (GP) IV (CD36) has been identified as a marker of PLT aggregation. Data on PLT function in the context of HIV infection remain inconclusive. Laboratory techniques, such as flow cytometry, enable the assessment of PLTs in their physiological state and environment, with minimal artifactual in vitro activation and aggregation. In this study, we describe a novel flow cytometry PLT assay, which enabled the measurement of PLT function in HIV infection. Forty-one antiretroviral-naïve HIV-positive individuals and 41 HIV-negative controls were recruited from a clinic in the Western Cape. Platelet function was evaluated by assessing the response of platelets to adenosine diphosphate (ADP) at two concentrations (0.04 mM, 0.2 mM). The percentage expression and mean fluorescence intensity (MFI) of CD62P and CD36 was used to evaluate platelet function. These were then correlated with platelet (PLT) count; CD4 count; % CD38/8; viral load and D-dimers. The % CD62P levels were higher in HIV-positive patients (HIV % CD62P 11.33[5.96-29.36] vs. control 2.48[1.56-6.04]; p < 0.0001). In addition, the HIV group showed higher CD62P MFI levels (HIV CD62P MFI 3.25 ± 7.23 vs. control 2.35 ± 1.31, p = 0.0292). Baseline levels of %CD36 expression were significantly higher in HIV-positive patients (%CD36 12.41[6.31-21.83] vs. control 6.04[1.34-13.15]; p = 0.0091). However, the baseline CD36MFI showed no significant difference between the two groups (HIV CD36 MFI 3.09 ± 0.64 vs. control 2.44 ± 0.11, p = 0.4591). The HIV group showed higher levels of % CD36 expression post stimulation with 0.04 mM ADP 43.32 ± 27.41 vs. control 27.47 ± 12.95; p < 0.0214) and no significant difference at 0.2 mM ADP (HIV % CD36 39.06 ± 17.91 vs. control 44.61 ± 18.76; p = 0.3277). Furthermore, the HIV group showed a single phase response to ADP as compared to the control group, which showed a normal biphasic response. We concluded that PLT flow cytometry is valuable in the assessment of levels of PLT activation, and further, that the addition of an endogenous agonist, such as ADP, enabled the measurement of PLT function in HIV infection. We were able to show that, although PLTs are significantly activated in HIV compared to uninfected controls, they retain their functional capacity.
Collapse
Affiliation(s)
- Bongani B Nkambule
- Divisions of Haematology, Department of Pathology, Stellenbosch University and NHLS , Tygerberg , South Africa and
| | | | | |
Collapse
|