1
|
Deng M, Biao R, Jiang M, Fu J, Zhao H, Du J. Over-activation and dysfunction of platelet-NK cell aggregates in HIV-infected individuals. J Transl Med 2025; 23:584. [PMID: 40420100 PMCID: PMC12105288 DOI: 10.1186/s12967-025-06591-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) infection is associated with over-activation, which contributes to disease progression. Platelet-leukocyte aggregates play a critical role in HIV-1 infection. However, research on the characteristics of platelet-natural killer (NK) cell aggregates in HIV-infected individuals still has certain limitations. METHODS Platelet-NK cell aggregates in the peripheral blood of participants were detected by flow cytometry and confirmed by imaging flow cytometry. Platelet activation was evaluated by CD62P expression. The expression of various activating and inhibitory receptors, markers of apoptosis, lipid droplets, interferon-gamma (IFN-γ), Granzyme B, and Perforin in platelet-NK cell aggregates were assessed. The signaling lymphocyte activating molecule (SLAM) family receptors in both platelets and NK cells and the levels of phosphorylation signals in NK cells were respectively measured through flow cytometry. RESULTS In this study, we observed an increase in platelet-NK cell aggregates that were negatively correlated with CD4 count, a prognostic marker for HIV-1 disease progression. Furthermore, platelet activation was inversely associated with both HIV-1 disease progression and the platelet-NK cell aggregates. However, antiretroviral therapy (ART) couldn't restore the levels of these aggregates or platelet activation. Compared to platelet-free NK cells, platelet-NK cell aggregates exhibited over-activation (CD69) and exhaustion phenotypes (CD39, LAG-3, PD-1), increased levels of apoptosis (Annexin V and CD95) and lipid droplets (Bodipy 493/503 and LipidTOX). Furthermore, NK cells' cytokine secretion (IFN-γ) and cytotoxic function (Granzyme B and Perforin) within the aggregates were declined. Screening results of SLAM receptors in NK cells and platelets suggested that platelets may transmit signals to NK cells via SLAMF5. Moreover, elevated levels of p-Fyn, p-PLC-γ2, p-SHP-1, and p-SHP-2 denoted disturbances in the downstream signals of the SLAM family within platelet-NK cell aggregates. CONCLUSION Our study indicates that platelet-NK cell aggregates exhibit characteristics of over-activation and dysfunction during HIV-1 infection. Hyperactivated platelets and the formation of platelet-NK cell aggregates contribute to the HIV-1 disease progression and the inflammation of the immune system. These findings may implicate potential targets of overactivated platelets for HIV-1 disease progression.
Collapse
Affiliation(s)
- Meiju Deng
- Clinical Center for HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Ruojia Biao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Meiqing Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jiantao Fu
- Clinical Center for HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Hongxin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Juan Du
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- Beijing Institute of Infectious Diseases, Beijing, 100015, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
2
|
George Pryzdial EL, Perrier JR, Rashid MU, West HE, Sutherland MR. Viral coagulation: pushing the envelope. J Thromb Haemost 2024; 22:3366-3382. [PMID: 39260743 DOI: 10.1016/j.jtha.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Many virus types affect the blood clotting system with correlations to pathology that range widely from thrombosis to hemorrhage linking to inflammation. Here we overview the intricate crosstalk induced by infection between proteins on the virus encoded by either the host or virus genomes, coagulation proteins, platelets, leukocytes, and endothelial cells. For blood-borne viruses with an outer covering acquired from the host cell, the envelope, a key player may be the cell-derived trigger of coagulation on the virus surface, tissue factor (TF). TF is a multifunctional transmembrane cofactor that accelerates factor (F)VIIa-dependent activation of FX to FXa, leading to clot formation. However, the nascent TF/FVIIa/FXa complex also facilitates G protein-coupled modulation of cells via protease-activated receptor 2. As a viral envelope constituent, TF can bypass the physiological modes of regulation, thereby initiating the activation of neighboring platelets, leukocytes, and endothelial cells. A thromboinflammatory environment is predicted due to feedback amplification in response to cellular release of cytokines, procoagulant proteins, neutrophil extracellular traps, and stimulus-induced accessibility of adhesive receptors, resulting in cellular aggregates. The pathobiological effects of thromboinflammation ultimately contribute to innate and adaptive immunity for viral clearance. In contrast, the preceding stages of viral infection may be enhanced via the TF-protease axis.
Collapse
Affiliation(s)
- Edward Louis George Pryzdial
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.
| | - John Ruggles Perrier
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mahamud-Ur Rashid
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Henry Euan West
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Michael Ross Sutherland
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
Manganaro JE, Emanuel K, Lamberty BG, George JW, Stauch KL. Pink1/Parkin deficiency alters circulating lymphocyte populations and increases platelet-T cell aggregates in rats. Sci Rep 2024; 14:23861. [PMID: 39394439 PMCID: PMC11470019 DOI: 10.1038/s41598-024-74775-w] [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: 05/16/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Parkinson's disease (PD) is the most common progressive neurodegenerative movement disorder and results from the selective loss of dopaminergic neurons in the substantia nigra pars compacta. Pink1 and Parkin are proteins that function together in mitochondrial quality control, and when they carry loss-of-function mutations lead to familial forms of PD. While much research has focused on central nervous system alterations in PD, peripheral contributions to PD pathogenesis are increasingly appreciated. We report Pink1/Parkin regulate glycolytic and mitochondrial oxidative metabolism in peripheral blood mononuclear cells (PBMCs) from rats. Pink1/Parkin deficiency induces changes in the circulating lymphocyte populations, namely increased CD4 + T cells and decreased CD8 + T cells and B cells. Loss of Pink1/Parkin leads to elevated platelet counts in the blood and increased platelet-T cell aggregation. Platelet-lymphocyte aggregates are associated with increased thrombosis risk suggesting targeting the Pink1/Parkin pathway in the periphery might have therapeutic potential.
Collapse
Affiliation(s)
- Jane E Manganaro
- College of Medicine, Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Katy Emanuel
- College of Medicine, Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benjamin G Lamberty
- College of Medicine, Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph W George
- College of Medicine, Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelly L Stauch
- College of Medicine, Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Peshkova AD, Saliakhutdinova SM, Sounbuli K, Selivanova YA, Andrianova IA, Khabirova AI, Litvinov RI, Weisel JW. The differential formation and composition of leukocyte-platelet aggregates induced by various cellular stimulants. Thromb Res 2024; 241:109092. [PMID: 39024901 PMCID: PMC11411814 DOI: 10.1016/j.thromres.2024.109092] [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: 05/01/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.
Collapse
Affiliation(s)
- Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | | | - Khetam Sounbuli
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Yuliya A Selivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Rustem I Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John W Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Wu F, Li Y, Jiang N, Jiang X, Liu X, Dai X, Wang F. Increased platelet-CD8+ T-cell aggregates displaying high activation, exhaustion, and tendency to death correlate with disease progression in people with HIV-1. J Leukoc Biol 2024; 116:166-176. [PMID: 38450750 DOI: 10.1093/jleuko/qiae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Platelets engage in HIV-1 infection by interacting with immune cells, which has been realized broadly. However, the potential interaction between platelets and CD8+ T cells remains unidentified. Here, treatment-naive individuals with HIV-1, complete immunological responders to antiretroviral therapy, and healthy controls were enrolled. First, we found that treatment-naive individuals with HIV-1 had low platelet numbers and high CD8+ T-cell counts when compared with complete immunological responders to antiretroviral therapy and healthy controls, leading to a low platelet/CD8+ T-cell ratio in peripheral blood, which could effectively differentiate the status of HIV-1 infection. Moreover, cytokines that may have been derived from platelets were higher in the plasma of people with HIV-1 despite viral suppression. Furthermore, we demonstrated that platelet-CD8+ T-cell aggregates were elevated in treatment-naive individuals with HIV-1, which positively correlated with HIV-1 viral load but negatively correlated with CD4+ T-cell count and CD4/CD8 ratio. Finally, we revealed that platelet-CD8+ T-cell aggregates correlate with enhanced activation/exhaustion and pyroptosis/apoptosis compared with free CD8+ T cells. Moreover, platelet-induced caspase 1 activation of CD8+ T cells correlated with IL-1β and IL-18 plasma levels. In brief, we reveal the importance of platelets in HIV-1 infection, which might secrete more cytokines and mediate CD8+ T-cell phenotypic characteristics by forming platelet-CD8+ T-cell aggregates, which are related to poor prognosis.
Collapse
Affiliation(s)
- Fengying Wu
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Yuanchun Li
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Nan Jiang
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Xu Jiang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Xiaoqing Liu
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Xiaopeng Dai
- Noncommissioned Officer School, Army Medical University, No.450 Zhongshan West Road, Qiaoxi District, Shijiazhuang, Hebei 050081, China
| | - Fusheng Wang
- Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, No.100 West Fourth Ring Middle Road, Fengtai District, Beijing 100039, China
| |
Collapse
|
6
|
Manganaro JE, Emanuel K, Lamberty BG, George JW, Stauch KL. Pink1/Parkin deficiency alters circulating lymphocyte populations and increases platelet-T cell aggregates in rats. RESEARCH SQUARE 2024:rs.3.rs-4431604. [PMID: 38854001 PMCID: PMC11160909 DOI: 10.21203/rs.3.rs-4431604/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Parkinson's disease (PD) is the most common progressive neurodegenerative movement disorder and results from the selective loss of dopaminergic neurons in the substantia nigra pars compacta. Pink1 and Parkin are proteins that function together in mitochondrial quality control, and when they carry loss-of-function mutations lead to familial forms of PD. While much research has focused on central nervous system alterations in PD, peripheral contributions to PD pathogenesis are increasingly appreciated. We report Pink1/Parkin regulate glycolytic and mitochondrial oxidative metabolism in peripheral blood mononuclear cells (PBMCs) from rats. Pink1/Parkin deficiency induces changes in the circulating lymphocyte populations, namely increased CD4 + T cells and decreased CD8 + T cells and B cells. Loss of Pink1/Parkin leads to elevated platelet counts in the blood and increased platelet-T cell aggregation. Platelet-lymphocyte aggregates are associated with increased thrombosis risk, and venous thrombosis is a cause of sudden death in PD, suggesting targeting the Pink1/Parkin pathway in the periphery has therapeutic potential.
Collapse
|
7
|
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
|
8
|
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
|
9
|
Shirasaki K, Minai K, Kawai M, Tanaka TD, Ogawa K, Inoue Y, Morimoto S, Nagoshi T, Ogawa T, Komukai K, Yoshimura M. Unique crosstalk between platelet and leukocyte counts during treatment for acute coronary syndrome: A retrospective observational study. Medicine (Baltimore) 2022; 101:e32439. [PMID: 36595999 PMCID: PMC9803419 DOI: 10.1097/md.0000000000032439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In the pathophysiology of acute coronary syndrome (ACS), platelet (PLT) and neutrophil (Neu) crosstalk may be important for activating coagulation and inflammation. It has been speculated that PLTs and Neu may affect each other's cell counts; however, few studies have investigated this hypothesis. In this study, we measured changes in blood cell counts in 245 patients with ACS during treatment and investigated the mutual effects of each blood cell type. Path diagrams were drawn using structural equation modeling, and temporal changes in the count of each blood cell type and the relevance of these changes were analyzed. Throughout the treatment period, the numbers of all blood cell types (red blood cells [RBCs], leukocytes, and PLTs) were associated with each other before and after treatment. A detailed examination of the different cell types revealed that the PLT count at admission had a significant positive effect on the leukocyte (especially Neu) count after treatment. Conversely, the leukocyte (especially Neu) count at admission had a significant positive effect on the PLT count after treatment. During ACS, PLTs and leukocytes, especially Neu, stimulate each other to increase their numbers. The formation of a PLT-leukocyte complex may increase coagulation activity and inflammation, which can lead to a further increase in the counts of both blood cell types.
Collapse
Affiliation(s)
- Keisuke Shirasaki
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Chiba, Japan
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kosuke Minai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
- * Correspondence: Kosuke Minai, Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-shimbashi, Minato-ku, Tokyo, Japan (e-mail: )
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshikazu D. Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasunori Inoue
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takayuki Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kimiaki Komukai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Chiba, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Shen CL, Wu YF. Flow cytometry for evaluating platelet immunophenotyping and function in patients with thrombocytopenia. Tzu Chi Med J 2022; 34:381-387. [PMID: 36578648 PMCID: PMC9791859 DOI: 10.4103/tcmj.tcmj_117_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/10/2022] [Accepted: 06/08/2022] [Indexed: 01/19/2023] Open
Abstract
Platelets play an essential role in primary hemostasis through bleeding and thromboembolism. Thus, the diagnosis or evaluation of impaired hereditary, acquired, and drug-related platelet dysfunction has become imperative. The assessment of the platelet function is too complex for routine platelet function study. The major methods involved in platelet function study include platelet function analyzer testing, thromboelastography, thromboelastometry, light transmission aggregometry, and flow cytometry. The current review article focuses on the methods with flow cytometry for immunophenotyping of platelet and evaluating platelet function for platelet disorders, especially in patients with thrombocytopenia. According to the consensus published by the International Society on Thrombosis and Haemostasis, for inherited and acquired platelet disorders, the two major measures by which flow cytometry determines platelet function are glycoprotein IIb/IIIa/P-selectin (CD62p) expression and percentage of leukocyte-platelet aggregates. Using flow cytometry to determine platelet function has several advantages, including good sensitivity to low platelet counts, small blood volume required, and the nonnecessity of centrifugation. However, flow cytometry has still many limitations and challenges, with standardization for routine laboratory testing also proving difficult. Although flow cytometry is available for multipurpose and sensitive study of platelet functions at the same time, the challenging analysis gradually increases and needs to be addressed before reality.
Collapse
Affiliation(s)
- Chih-Lung Shen
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yi-Feng Wu
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Yi-Feng Wu, Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Section 3, Chung-Yang Road, Hualien, Taiwan. E-mail:
| |
Collapse
|
11
|
Ogweno G. Challenges in Platelet Functions in HIV/AIDS Management. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The interest in platelet functions in HIV/AIDS is due to the high incidence of microvascular thrombosis in these individuals. A lot of laboratory data have been generated regarding platelet functions in this population. The tests demonstrate platelet hyperactivity but decreased aggregation, though results are inconsistent depending on the study design. Antiretroviral treatments currently in use display complex interactions. Many studies on platelet functions in these patients have been for research purposes, but none have found utility in guiding drug treatment of thrombosis.
Collapse
|
12
|
Li P, Zhang C, Wang M, Zhang X, Zhang Y, Tang K, Hu H, Jia X, Zhuang R, Jin B, Ma Y, Zhang Y. Elevation of Myeloperoxidase Correlates with Disease Severity in Patients with Hantaan Virus Infection. Viral Immunol 2022; 35:418-424. [PMID: 35675645 DOI: 10.1089/vim.2022.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hantaan orthohantavirus (HTNV) can cause hemorrhagic fever with renal syndrome (HFRS) characterized by acute kidney injury and hemorrhage. Neutrophils are the most abundant innate immune cell and the body's first line of defense against pathogens. Currently, an increasing number of studies have shown that neutrophils may be a mixed blessing in terms of viral infections. However, the role of neutrophils in HFRS patients with HTNV infection has not been fully declared. In this study, we analyzed plasma levels of both myeloperoxidase (MPO) and MPO-DNA in HFRS patients, together with the clinical parameters. Neutrophil-platelet aggregates (NPAs) during the acute and convalescent phases of HFRS were also assessed. The results showed that plasma MPO-DNA levels had no change in different disease phases or severities of HFRS patients. Whereas plasma MPO significantly increased in the acute phase and critical/severe groups of HFRS patients. Furthermore, plasma MPO was positively correlated with inflammatory clinical parameters, such as white blood cell counts, neutrophil counts, and renal injury-related parameters, such as blood urea nitrogen, blood uric acid, and serum creatinine, as well as negatively correlated with and platelet counts. In addition, NPAs increased both in acute and convalescent phase in HFRS patients compared with normal controls. These results suggested that elevated plasma MPO in HFRS patients correlated with disease severity, together with the increases of NPAs in HFRS patients, which may provide new insights into potential role of neutrophils in the pathogenesis of HFRS.
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China.,Brigade of Cadet, Air Force Medical University, Xi'an, China
| | - Chunmei Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Meng Wang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Xiyue Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Yusi Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Kang Tang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Haifeng Hu
- Center for Infectious Diseases, Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiaozhou Jia
- Department of Infectious Disease, Xi'an Eighth Hospital, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Ying Ma
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| |
Collapse
|
13
|
Caetano DG, Ribeiro-Alves M, Hottz ED, Vilela LM, Cardoso SW, Hoagland B, Grinsztejn B, Veloso VG, Morgado MG, Bozza PT, Guimarães ML, Côrtes FH. Increased biomarkers of cardiovascular risk in HIV-1 viremic controllers and low persistent inflammation in elite controllers and art-suppressed individuals. Sci Rep 2022; 12:6569. [PMID: 35449171 PMCID: PMC9023525 DOI: 10.1038/s41598-022-10330-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/05/2022] [Indexed: 11/15/2022] Open
Abstract
HIV controllers (HICs) are models of HIV functional cure, although some studies have shown persistent inflammation and increased rates of atherosclerosis in HICs. Since immune activation/inflammation contributes to the pathogenesis of cardiovascular diseases (CVD), we evaluated clinical data and inflammation markers in HIV-1 viremic controllers (VC), elite controllers (EC), and control groups (HIV positive individuals with virological suppression by antiretroviral therapy-cART; HIV negative individuals-HIVneg) to assess whether they presented elevated levels of inflammation markers also associated with CVD. We observed the highest frequencies of activated CD8+ T cells in VCs, while EC and cART groups presented similar but slightly altered frequencies of this marker when compared to the HIVneg group. Regarding platelet activation, both HICs groups presented higher expression of P-selectin in platelets when compared to control groups. Monocyte subset analyses revealed lower frequencies of classical monocytes and increased frequencies of non-classical and intermediate monocytes among cART individuals and in EC when compared to HIV negative individuals, but none of the differences were significant. For VC, however, significant decreases in frequencies of classical monocytes and increases in the frequency of intermediate monocytes were observed in comparison to HIV negative individuals. The frequency of monocytes expressing tissue factor was similar among the groups on all subsets. In terms of plasma markers, VC had higher levels of many inflammatory markers, while EC had higher levels of VCAM-1 and ICAM-1 compared to control groups. Our data showed that VCs display increased levels of inflammation markers that have been associated with CVD risk. Meanwhile, ECs show signals of lower but persistent inflammation, comparable to the cART group, indicating the potential benefits of alternative therapies to decrease inflammation in this group.
Collapse
Affiliation(s)
- Diogo Gama Caetano
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute - IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Marcelo Ribeiro-Alves
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Eugênio Damaceno Hottz
- Laboratory of Immunothrombosis, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute - IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Larissa Melo Vilela
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Sandra Wagner Cardoso
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Brenda Hoagland
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Beatriz Grinsztejn
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Valdilea Gonçalves Veloso
- Laboratory of Clinical Research in STD and AIDS, National Institute of Infectology Evandro Chagas - INI, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Mariza Gonçalves Morgado
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute - IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute - IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Fernanda Heloise Côrtes
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute - IOC, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Madzime M, Rossouw TM, Theron AJ, Anderson R, Steel HC. Interactions of HIV and Antiretroviral Therapy With Neutrophils and Platelets. Front Immunol 2021; 12:634386. [PMID: 33777022 PMCID: PMC7994251 DOI: 10.3389/fimmu.2021.634386] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophils are important components of the innate immune system that mediate pathogen defense by multiple processes including phagocytosis, release of proteolytic enzymes, production of reactive oxygen species, and neutrophil extracellular trap formation. Abnormalities of neutrophil count and function have been described in the setting of HIV infection, with the majority of antiretroviral agents (ARVs), excluding zidovudine, having been reported to correct neutropenia. Questions still remain, however, about their impact on neutrophil function, particularly the possibility of persistent neutrophil activation, which could predispose people living with HIV to chronic inflammatory disorders, even in the presence of virally-suppressive treatment. In this context, the effects of protease inhibitors and integrase strand transfer inhibitors, in particular, on neutrophil function remain poorly understood and deserve further study. Besides mediating hemostatic functions, platelets are increasingly recognized as critical role players in the immune response against infection. In the setting of HIV, these cells have been found to harbor the virus, even in the presence of antiretroviral therapy (ART) potentially promoting viral dissemination. While HIV-infected individuals often present with thrombocytopenia, they have also been reported to have increased platelet activation, as measured by an upregulation of expression of CD62P (P-selectin), CD40 ligand, glycoprotein IV, and RANTES. Despite ART-mediated viral suppression, HIV-infected individuals reportedly have sustained platelet activation and dysfunction. This, in turn, contributes to persistent immune activation and an inflammatory vascular environment, seemingly involving neutrophil-platelet-endothelium interactions that increase the risk for development of comorbidities such as cardiovascular disease (CVD) that has become the leading cause of morbidity and mortality in HIV-infected individuals on treatment, clearly underscoring the importance of unraveling the possible etiologic roles of ARVs. In this context, abacavir and ritonavir-boosted lopinavir and darunavir have all been linked to an increased risk of CVD. This narrative review is therefore focused primarily on the role of neutrophils and platelets in HIV transmission and disease, as well as on the effect of HIV and the most common ARVs on the numbers and functions of these cells, including neutrophil-platelet-endothelial interactions.
Collapse
Affiliation(s)
- Morris Madzime
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Theresa M Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annette J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Helen C Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
17
|
Raadsen M, Du Toit J, Langerak T, van Bussel B, van Gorp E, Goeijenbier M. Thrombocytopenia in Virus Infections. J Clin Med 2021; 10:877. [PMID: 33672766 PMCID: PMC7924611 DOI: 10.3390/jcm10040877] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Thrombocytopenia, which signifies a low platelet count usually below 150 × 109/L, is a common finding following or during many viral infections. In clinical medicine, mild thrombocytopenia, combined with lymphopenia in a patient with signs and symptoms of an infectious disease, raises the suspicion of a viral infection. This phenomenon is classically attributed to platelet consumption due to inflammation-induced coagulation, sequestration from the circulation by phagocytosis and hypersplenism, and impaired platelet production due to defective megakaryopoiesis or cytokine-induced myelosuppression. All these mechanisms, while plausible and supported by substantial evidence, regard platelets as passive bystanders during viral infection. However, platelets are increasingly recognized as active players in the (antiviral) immune response and have been shown to interact with cells of the innate and adaptive immune system as well as directly with viruses. These findings can be of interest both for understanding the pathogenesis of viral infectious diseases and predicting outcome. In this review, we will summarize and discuss the literature currently available on various mechanisms within the relationship between thrombocytopenia and virus infections.
Collapse
Affiliation(s)
- Matthijs Raadsen
- Department of Viroscience, Erasmus MC Rotterdam, Doctor molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.R.); (T.L.); (E.v.G.)
| | - Justin Du Toit
- Department of Haematology, Wits University Donald Gordon Medical Centre Johannesburg, Johannesburg 2041, South Africa;
| | - Thomas Langerak
- Department of Viroscience, Erasmus MC Rotterdam, Doctor molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.R.); (T.L.); (E.v.G.)
| | - Bas van Bussel
- Department of Intensive Care Medicine, Maastricht University Medical Center Plus, 6229 HX Maastricht, The Netherlands;
- Care and Public Health Research Institute (CAPHRI), Maastricht University, 6229 GT Maastricht, The Netherlands
| | - Eric van Gorp
- Department of Viroscience, Erasmus MC Rotterdam, Doctor molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.R.); (T.L.); (E.v.G.)
- Department of Internal Medicine, Erasmus MC Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Marco Goeijenbier
- Department of Viroscience, Erasmus MC Rotterdam, Doctor molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.R.); (T.L.); (E.v.G.)
- Department of Internal Medicine, Erasmus MC Rotterdam, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
18
|
Oduguwa E, Dongarwar D, Salihu HM. Trends in Premature Deaths among Women Living with HIV/AIDS and Cervical Cancer. South Med J 2021; 113:651-658. [PMID: 33263137 DOI: 10.14423/smj.0000000000001184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES There is a lack of updated information on premature death and years of potential life lost (YPLL) among human immunodeficiency (HIV)-positive women with cervical cancer. We hypothesize that increased access to preventive resources such as antiretroviral therapy, preexposure prophylaxis, and human papillomavirus vaccines has reduced premature mortality and YPLL in these women in the previous decades. METHODS We used data from the National Inpatient Sample database from 2003 to the third quarter of 2015, and restricted the analysis to HIV-positive women with or without cervical cancer. Joinpoint regression models were run to identify trends in the rates of HIV and cervical cancer. Overall and age-stratified YPLL were calculated for HIV-positive women with cervical cancer. Adjusted survey logistic regression models were built to determine the predictive factors of in-hospital mortality among women living with HIV. RESULTS Among hospitalized women, low-income, non-Hispanic Blacks, and patients aged 40 to 59 years experienced greater frequencies of HIV/cervical cancer comorbidity. The prevalence of HIV hospitalizations increased by an average annual percentage of 0.9% (95% confidence interval 0.3-1.6). YPLL decreased in HIV-positive women living with and without cervical cancer by 4.9% and 4.3%, respectively. The trajectory for YPLL was not uniform across age groups. YPLL decreased substantially in women aged 20 to 29 years with HIV/cervical cancer comorbidity. Cervical cancer remained a significant predictor of mortality among HIV-positive women when adjusted for age, race, and insurance coverage. CONCLUSIONS Within a large, national sample from 2003 to 2015, we found an overall declining trend in YPLL in women living with HIV/cervical cancer comorbidity. In-hospital mortality among HIV-positive women was associated with cervical cancer, age, race, and insurance coverage. We recommend further investigation into the quality of HIV and cervical cancer treatment and prevention services for the sociodemographic groups described.
Collapse
Affiliation(s)
- Emmanuella Oduguwa
- From the Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, Houston, Texas
| | - Deepa Dongarwar
- From the Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, Houston, Texas
| | - Hamisu M Salihu
- From the Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
19
|
Li Q, Wang Y, Xue W, Bian Z, Gao Y, Zeng Y, Tang L, Tang T, Tian Y, Guo W. Immunomodulatory effects of platelets on the severity of hand, foot, and mouth disease infected with enterovirus 71. Pediatr Res 2021; 89:814-822. [PMID: 32516798 PMCID: PMC8049866 DOI: 10.1038/s41390-020-0970-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/26/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Enterovirus 71 (EV71) infection contributes to hand, foot, and mouth disease (HFMD) with severe neurogenic complications, leading to higher morbidity. In addition to their typical roles in coagulation, platelets could serve as essential immune regulatory cells to play a key role in the pathogenesis of this viral infection. METHODS Platelet parameters were measured using an automatic hematology analyzer. T-helper type 1 (Th1) and Th2 cells were analyzed by flow cytometry. The levels of cytokines and key transcription factors were determined. RESULTS The levels of platelet count and plateletcrit were positively associated with the severity of HFMD. Th1 and Th2 cells as well as their corresponding cytokines were increased in the severe group compared to the healthy volunteers. Moreover, the levels of platelets were negatively correlated with the level of interferon-γ (IFN-γ), but positively correlated with the frequency of Th1 cells. Coculture of platelets and naive CD4+ T cells showed that platelets from mild patients promote Th1 cell differentiation and IFN-γ secretion. CONCLUSIONS Our study has shown for the first time that the distinct roles of platelets are responsible for the regulation of pathogenic CD4+ T cell differentiation and function in the pathogenesis of HFMD caused by EV71. IMPACT Our study has shown for the first time that the distinct roles of platelets are responsible for the regulation of pathogenic CD4+ T cell differentiation and function in the pathogenesis of HFMD caused by EV71. For the first time, we have discovered the role of platelets in children's HFMD caused by EV71 infection, which may provide a better treatment for HFMD in the future. This article describes new discoveries in platelet immunity.
Collapse
Affiliation(s)
- Qianwen Li
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yimeng Wang
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenyao Xue
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengying Bian
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Gao
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yu Zeng
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lei Tang
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tiejun Tang
- grid.254147.10000 0000 9776 7793Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ye Tian
- Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Wei Guo
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
20
|
Murray KD, Singh MV, Zhuang Y, Uddin MN, Qiu X, Weber MT, Tivarus ME, Wang HZ, Sahin B, Zhong J, Maggirwar SB, Schifitto G. Pathomechanisms of HIV-Associated Cerebral Small Vessel Disease: A Comprehensive Clinical and Neuroimaging Protocol and Analysis Pipeline. Front Neurol 2020; 11:595463. [PMID: 33384655 PMCID: PMC7769815 DOI: 10.3389/fneur.2020.595463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: We provide an in-depth description of a comprehensive clinical, immunological, and neuroimaging study that includes a full image processing pipeline. This approach, although implemented in HIV infected individuals, can be used in the general population to assess cerebrovascular health. Aims: In this longitudinal study, we seek to determine the effects of neuroinflammation due to HIV-1 infection on the pathomechanisms of cerebral small vessel disease (CSVD). The study focuses on the interaction of activated platelets, pro-inflammatory monocytes and endothelial cells and their impact on the neurovascular unit. The effects on the neurovascular unit are evaluated by a novel combination of imaging biomarkers. Sample Size: We will enroll 110 HIV-infected individuals on stable combination anti-retroviral therapy for at least three months and an equal number of age-matched controls. We anticipate a drop-out rate of 20%. Methods and Design: Subjects are followed for three years and evaluated by flow cytometric analysis of whole blood (to measure platelet activation, platelet monocyte complexes, and markers of monocyte activation), neuropsychological testing, and brain MRI at the baseline, 18- and 36-month time points. MRI imaging follows the recommended clinical small vessel imaging standards and adds several advanced sequences to obtain quantitative assessments of brain tissues including white matter microstructure, tissue susceptibility, and blood perfusion. Discussion: The study provides further understanding of the underlying mechanisms of CSVD in chronic inflammatory disorders such as HIV infection. The longitudinal study design and comprehensive approach allows the investigation of quantitative changes in imaging metrics and their impact on cognitive performance.
Collapse
Affiliation(s)
- Kyle D Murray
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States
| | - Meera V Singh
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Yuchuan Zhuang
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, United States
| | - Md Nasir Uddin
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Xing Qiu
- Department of Biostatistics, University of Rochester, Rochester, NY, United States
| | - Miriam T Weber
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Madalina E Tivarus
- Department of Imaging Sciences, University of Rochester, Rochester, NY, United States.,Department of Neuroscience, University of Rochester, Rochester, NY, United States
| | - Henry Z Wang
- Department of Imaging Sciences, University of Rochester, Rochester, NY, United States
| | - Bogachan Sahin
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Jianhui Zhong
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States.,Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, United States.,Department of Biostatistics, University of Rochester, Rochester, NY, United States
| | - Sanjay B Maggirwar
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, United States
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester, Rochester, NY, United States.,Department of Imaging Sciences, University of Rochester, Rochester, NY, United States
| |
Collapse
|
21
|
Manne BK, Denorme F, Middleton EA, Portier I, Rowley JW, Stubben C, Petrey AC, Tolley ND, Guo L, Cody M, Weyrich AS, Yost CC, Rondina MT, Campbell RA. Platelet gene expression and function in patients with COVID-19. Blood 2020; 136:1317-1329. [PMID: 32573711 PMCID: PMC7483430 DOI: 10.1182/blood.2020007214] [Citation(s) in RCA: 689] [Impact Index Per Article: 137.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need to understand the pathogenesis of coronavirus disease 2019 (COVID-19). In particular, thrombotic complications in patients with COVID-19 are common and contribute to organ failure and mortality. Patients with severe COVID-19 present with hemostatic abnormalities that mimic disseminated intravascular coagulopathy associated with sepsis, with the major difference being increased risk of thrombosis rather than bleeding. However, whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection alters platelet function to contribute to the pathophysiology of COVID-19 remains unknown. In this study, we report altered platelet gene expression and functional responses in patients infected with SARS-CoV-2. RNA sequencing demonstrated distinct changes in the gene-expression profile of circulating platelets of COVID-19 patients. Pathway analysis revealed differential gene-expression changes in pathways associated with protein ubiquitination, antigen presentation, and mitochondrial dysfunction. The receptor for SARS-CoV-2 binding, angiotensin-converting enzyme 2 (ACE2), was not detected by messenger RNA (mRNA) or protein in platelets. Surprisingly, mRNA from the SARS-CoV-2 N1 gene was detected in platelets from 2 of 25 COVID-19 patients, suggesting that platelets may take-up SARS-COV-2 mRNA independent of ACE2. Resting platelets from COVID-19 patients had increased P-selectin expression basally and upon activation. Circulating platelet-neutrophil, -monocyte, and -T-cell aggregates were all significantly elevated in COVID-19 patients compared with healthy donors. Furthermore, platelets from COVID-19 patients aggregated faster and showed increased spreading on both fibrinogen and collagen. The increase in platelet activation and aggregation could partially be attributed to increased MAPK pathway activation and thromboxane generation. These findings demonstrate that SARS-CoV-2 infection is associated with platelet hyperreactivity, which may contribute to COVID-19 pathophysiology.
Collapse
Affiliation(s)
| | | | | | | | - Jesse W Rowley
- Molecular Medicine Program
- Department of Internal Medicine
| | - Chris Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute
| | | | | | - Li Guo
- Molecular Medicine Program
| | - Mark Cody
- Molecular Medicine Program
- Department of Pediatrics, University of Utah, Salt Lake City, UT; and
| | - Andrew S Weyrich
- Molecular Medicine Program
- Department of Internal Medicine
- Department of Pathology, and
| | - Christian C Yost
- Molecular Medicine Program
- Department of Pediatrics, University of Utah, Salt Lake City, UT; and
| | - Matthew T Rondina
- Molecular Medicine Program
- Department of Internal Medicine
- Department of Pathology, and
- Department of Internal Medicine, George E. Wahlen Department of Veterans Affairs (VA) Medical Center, and
- Geriatric Research, Education, and Clinical Center (GRECC), VA Salt Lake City Healthcare System, Salt Lake City, UT
| | | |
Collapse
|
22
|
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: 107] [Impact Index Per Article: 21.4] [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
|
23
|
Thompson J, Onyenaka C, Oduguwa E, Dongarwar D, Gendra S, Coker V, Kutse S, Blanco M, Nwangwu O, King C, Enamorado E, Bakare O, Ajewole VB, Spooner KK, Salemi JL, Aliyu MH, Salihu HM, Olaleye OA. Trends and Racial/Ethnic Disparities in the Rates of Pre-eclampsia by HIV Status in the US. J Racial Ethn Health Disparities 2020; 8:670-677. [PMID: 32754847 DOI: 10.1007/s40615-020-00826-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/06/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Preeclampsia and HIV account for a significant proportion of the global burden of disease and pose severe maternal-fetal risks. There is a dearth of literature regarding racial/ethnic disparities in preeclampsia associated with HIV/AIDS in the US. METHODS We retrospectively analyzed data from the National Inpatient Sample (NIS) database from 2002 to 2015 on a cohort of hospitalized pregnant women with or without preeclampsia and HIV. Joinpoint regression models were used to identify trends in the rates of preeclampsia among pregnant women living with or without HIV, stratified by race/ethnicity over the study period. We also assessed the association between preeclampsia and various socio-demographic factors. RESULTS We analyzed over 60 million pregnancy-related hospitalizations, of which 3665 had diagnoses of preeclampsia and HIV, corresponding to a rate of 0.61 per 10,000. There was an increasing trend in the diagnosis of preeclampsia among hospitalized, pregnant women without HIV across each racial/ethnic category. The highest prevalence of preeclampsia was among non-Hispanic (NH) Blacks, regardless of HIV status. CONCLUSION The increase in rates of pre-eclampsia between 2002 and 2015 was mostly noted among pregnant women without HIV. Regardless of HIV status, NH-Blacks experienced the highest discharge prevalence of preeclampsia.
Collapse
Affiliation(s)
- Jamalena Thompson
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Collins Onyenaka
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Emmanuella Oduguwa
- Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77098, USA.
| | - Deepa Dongarwar
- Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77098, USA
| | - Sumaya Gendra
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Vidella Coker
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Seun Kutse
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Mayra Blanco
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Onyinye Nwangwu
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Charlee King
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Elza Enamorado
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Oluwatoyin Bakare
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Veronica B Ajewole
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Kiara K Spooner
- Department of Family and Community Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jason L Salemi
- College of Public Health, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Muktar H Aliyu
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hamisu M Salihu
- Center of Excellence in Health Equity, Training, and Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77098, USA.,Department of Family and Community Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Omonike A Olaleye
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| |
Collapse
|
24
|
Alharbi A, Thompson JP, Brindle NP, Stover CM. Ex vivo modelling of the formation of inflammatory platelet-leucocyte aggregates and their adhesion on endothelial cells, an early event in sepsis. Clin Exp Med 2019; 19:321-337. [PMID: 30191349 PMCID: PMC6647484 DOI: 10.1007/s10238-018-0526-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/27/2018] [Indexed: 12/18/2022]
Abstract
Septicaemia is an acute inflammatory reaction in the bloodstream to the presence of pathogen-associated molecular patterns. Whole blood stimulation assays capture endotoxin-induced formation of aggregates between platelets and leucocytes using flow cytometry. We wanted to assess extent of spontaneous aggregate formation in whole blood stimulation assays and compare the effects of endotoxin and heat-killed, clinically relevant, bacterial pathogens on aggregate formation and then on adhesion of aggregates to TNFα-stimulated endothelial cells. We found that endotoxin (from Escherichia coli or Salmonella enteritidis) was not a suitable stimulus to provoke platelet-leucocyte aggregates in vitro, as it did not further increase the extent of aggregates formed spontaneously in stasis of hirudin-anticoagulated blood. Specifically, whole blood samples stimulated with or without LPS produced aggregates with a mean surface area of 140.97 and 117.68 μm2, respectively. By contrast, incubation of whole blood with heat-killed Klebsiella pneumoniae or Staphylococcus aureus produced significantly enhanced and complex cellular aggregates (with a mean surface area of 470.61 and 518.39 μm2, respectively) which adhered more frequently to TNFα (and free fatty acid)-stimulated endothelial cells. These were reliably captured by scanning electron microscopy. Adhesion of cellular aggregates could be blocked by incubation of endothelial cells with a commercial P-selectin antibody and an angiopoietin-2 ligand trap. In conclusion, we have developed an in vitro method that models the acute inflammatory reaction in whole blood in the presence of sepsis-relevant bacterial pathogen surfaces.
Collapse
Affiliation(s)
- Azzah Alharbi
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jonathan P Thompson
- Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Nicholas P Brindle
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- Department of Molecular & Cell Biology, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
25
|
Lee BC, Lin KH, Hu CY, Lo SC. Thromboelastography characterized CD36 null subjects as slow clot formation and indicative of hypocoagulability. Thromb Res 2019; 178:79-84. [DOI: 10.1016/j.thromres.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/05/2019] [Indexed: 11/16/2022]
|
26
|
Circulating platelet-neutrophil aggregates as risk factor for deep venous thrombosis. ACTA ACUST UNITED AC 2019; 57:707-715. [DOI: 10.1515/cclm-2018-0909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/28/2018] [Indexed: 01/30/2023]
Abstract
Abstract
Background
Platelet-neutrophil aggregates (PNAs) are fundamental mechanisms linking hemostasis and inflammatory processes. Elevated level of PNAs have been reported in inflammatory diseases and coronary artery diseases. However, studies on the correlation between PNAs formation and deep venous thrombosis (DVT) are not available.
Methods
A total of 92 participants were involved in this study, including 32 cases with DVT and 60 cases without DVT. Blood samples coagulated by K2-EDTA or sodium citrate were prepared for blood cell count and blood smears. PNAs and platelet activation were measured using flow cytometry. The correlation between platelet activation level and PNAs level was analyzed by linear regression. Receiver operating characteristic (ROC) analysis was performed, assessing the prognostic performance of PNAs to predict potential risk of DVT occurrence.
Results
PNAs was found in the blood smears of patients with DVT. Significant increased level of PNAs was identified in DVT group (medium 8.43%, interquartile range [IQR] 4.11%–15.69%), compared with that in control group (5.16%, IQR 2.40–9.60, p<0.01). The DVT group also showed a dramatic elevated level of total platelet activation (medium 16.06%, IQR 6.04–22.05) vs. control group (11.26%, IQR 5.54–19.99, p<0.05). The PNAs level was correlated with total platelet activation (r2=0.58, p<0.0001). A significantly high odds ratio (OR) of DVT occurrence was identified when the level of PNAs was higher than 7.4% (OR 3.60, 95% confidence interval [CI] 1.463–8.838, p<0.01).
Conclusions
An elevated level of PNAs was associated with risk of DVT occurrence, which might be a suitable marker predicting DVT development.
Collapse
|
27
|
Mechanisms of Cardiovascular Disease in the Setting of HIV Infection. Can J Cardiol 2018; 35:238-248. [PMID: 30825947 DOI: 10.1016/j.cjca.2018.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Although the initial reports of increased cardiovascular (CV) disease in the setting of advanced AIDS were reported approximately 30 years ago, advances in antiretroviral therapy and immediate initiation of therapy on diagnosis have transformed what was once a deadly infectious disease into a chronic health condition. Accordingly, the types of CV diseases occurring in HIV have shifted from pericardial effusions and dilated cardiomyopathy to atherosclerosis and heart failure. The underlying pathophysiology of HIV-associated CV disease remains poorly understood, partly because of the rapidly evolving nature of HIV treatment and because clinical endpoints take many years to develop. The gut plays an important role in the early pathogenesis of HIV infection as HIV preferentially infects CD4+ T cells, 80% of which are located in gut mucosa. The loss of these T cells damages gut mucosa resulting in increased gut permeability and microbial translocation, which incites chronic inflammation and immune activation. Antiretroviral therapy does not cure HIV infection and immune abnormalities persist. These abnormalities correlate with mortality and CV events. The effects of antiretroviral therapy on CV risk are complex; treatment reduces inflammation and other markers of CV risk but induces lipid abnormalities, most commonly hypertriglyceridemia. On a molecular level, monocytes/macrophages, platelet reactivity, and immune cell activation, which play a role in the general population, may be heightened in the setting of HIV and contribute to HIV-associated atherosclerosis. Chronic inflammation represents an inviting therapeutic target in HIV, as it does in uninfected persons with atherosclerosis.
Collapse
|
28
|
Camon S, Quiros C, Saubi N, Moreno A, Marcos MA, Eto Y, Rofael S, Monclus E, Brown J, McHugh TD, Mallolas J, Perello R. Full blood count values as a predictor of poor outcome of pneumonia among HIV-infected patients. BMC Infect Dis 2018; 18:189. [PMID: 29673334 PMCID: PMC5909258 DOI: 10.1186/s12879-018-3090-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/10/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND To evaluate the predictive value of analytical markers of full blood count that can be assessed in the emergency department for HIV infected patients, with community-acquired pneumonia (CAP). METHODS Prospective 3-year study including all HIV-infected patients that went to our emergency department with respiratory clinical infection, more than 24-h earlier they were diagnosed with CAP and required admission. We assessed the different values of the first blood count performed on the patient as follows; total white blood cells (WBC), neutrophils, lymphocytes (LYM), basophils, eosinophils (EOS), red blood cells (RBC), hemoglobin, hematocrit, mean corpuscular volume, mean corpuscular hemoglobin concentration, mean corpuscular hemoglobin, red blood cell distribution width (RDW), platelets (PLT), mean platelet volume, and platelet distribution width (PDW). The primary outcome measure was 30-day mortality and the secondary, admission to an intensive care unit (ICU). The predictive power of the variables was determined by statistical calculation. RESULTS One hundred sixty HIV-infected patients with pneumonia were identified. The mean age was 42 (11) years, 99 (62%) were male, 79 (49%) had ART. The main route of HIV transmission was through parenteral administration of drugs. Streptococcus pneumonia was the most frequently identified etiologic agent of CAP The univariate analysis showed that the values of PLT (p < 0.009), EOS (p < 0.033), RDW (p < 0.033) and PDW (p < 0.09) were predictor of mortality, but after the logistic regression analysis, no variable was shown as an independent predictor of mortality. On the other hand, higher RDW (OR = 1.2, 95% CI 1.1-1.4, p = 0.013) and a lower number of LYM (OR 2.2, 95% CI 1.1-2.2; p = 0.035) were revealed as independent predictors of admission to ICU. CONCLUSION Red blood cell distribution and lymphocytes were the most useful predictors of disease severity identifying HIV infected patients with CAP who required ICU admission.
Collapse
Affiliation(s)
- S. Camon
- Servicio de Urgencias, Hospital Clínic, Barcelona, Spain
| | - C. Quiros
- Servicio de Urgencias, Hospital Clínic, Barcelona, Spain
| | - N. Saubi
- Servicio de Enfermedades Infecciosas, Hospital Clínic, Barcelona, Spain
| | - A. Moreno
- Servicio de Enfermedades Infecciosas, Hospital Clínic, Barcelona, Spain
| | - M. A. Marcos
- Servicio de Microbiología, Hospital Clínic, Barcelona, Spain
| | - Y. Eto
- Servicio de Enfermedades Infecciosas, Hospital Clínic, Barcelona, Spain
| | - S. Rofael
- Microbiology department, UCL, Royal Free Hospital, London, UK
| | - E. Monclus
- Servicio de Urgencias, Hospital Clínic, Barcelona, Spain
| | - J. Brown
- Pneumology department, Royal Free Hospital, London, UK
| | - T. D. McHugh
- Pneumology department, Royal Free Hospital, London, UK
| | - J. Mallolas
- Servicio de Enfermedades Infecciosas, Hospital Clínic, Barcelona, Spain
| | - R. Perello
- Servicio de Urgencias, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
29
|
Theron AJ, Anderson R, Rossouw TM, Steel HC. The Role of Transforming Growth Factor Beta-1 in the Progression of HIV/AIDS and Development of Non-AIDS-Defining Fibrotic Disorders. Front Immunol 2017; 8:1461. [PMID: 29163528 PMCID: PMC5673850 DOI: 10.3389/fimmu.2017.01461] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022] Open
Abstract
Even after attainment of sustained viral suppression following implementation of highly active antiretroviral therapy, HIV-infected persons continue to experience persistent, low-grade, systemic inflammation. Among other mechanisms, this appears to result from ongoing microbial translocation from a damaged gastrointestinal tract. This HIV-related chronic inflammatory response is paralleled by counteracting, but only partially effective, biological anti-inflammatory processes. Paradoxically, however, this anti-inflammatory response not only exacerbates immunosuppression but also predisposes for development of non-AIDS-related, non-communicable disorders. With respect to the pathogenesis of both sustained immunosuppression and the increased frequency of non-AIDS-related disorders, the anti-inflammatory/profibrotic cytokine, transforming growth factor-β1 (TGF-β1), which remains persistently elevated in both untreated and virally suppressed HIV-infected persons, may provide a common link. In this context, the current review is focused on two different, albeit related, harmful activities of TGF-β1 in HIV infection. First, on the spectrum of anti-inflammatory/immunosuppressive activities of TGF-β1 and the involvement of this cytokine, derived predominantly from T regulatory cells, in driving disease progression in HIV-infected persons via both non-fibrotic and profibrotic mechanisms. Second, the possible involvement of sustained elevations in circulating and tissue TGF-β1 in the pathogenesis of non-AIDS-defining cardiovascular, hepatic, pulmonary and renal disorders, together with a brief comment on potential TGF-β1-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Annette J. Theron
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Ronald Anderson
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Theresa M. Rossouw
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Helen C. Steel
- Faculty of Health Sciences, Department of Immunology, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
30
|
Platelet mitochondrial dysfunction and the correlation with human diseases. Biochem Soc Trans 2017; 45:1213-1223. [PMID: 29054925 DOI: 10.1042/bst20170291] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/10/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022]
Abstract
The platelet is considered as an accessible and valuable tool to study mitochondrial function, owing to its greater content of fully functional mitochondria compared with other metabolically active organelles. Different lines of studies have demonstrated that mitochondria in platelets have function far more than thrombogenesis regulation, and beyond hemostasis, platelet mitochondrial dysfunction has also been used for studying mitochondrial-related diseases. In this review, the interplay between platelet mitochondrial dysfunction and oxidative stress, mitochondrial DNA lesions, electron transfer chain impairments, mitochondrial apoptosis and mitophagy has been outlined. Meanwhile, considerable efforts have been made towards understanding the role of platelet mitochondrial dysfunction in human diseases, such as diabetes mellitus, sepsis and neurodegenerative disorders. Alongside this, we have also articulated our perspectives on the development of potential biomarkers of platelet mitochondrial dysfunction in mitochondrial-related diseases.
Collapse
|
31
|
Davison GM, Nkambule BB, Mkandla Z, Hon GM, Kengne AP, Erasmus RT, Matsha TE. Platelet, monocyte and neutrophil activation and glucose tolerance in South African Mixed Ancestry individuals. Sci Rep 2017; 7:40329. [PMID: 28091589 PMCID: PMC5238515 DOI: 10.1038/srep40329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 12/14/2022] Open
Abstract
Platelet activation has been described in patients with chronic inflammation, however in type 2 diabetes mellitus it remains controversial. We compared levels of platelet leucocyte aggregates, monocyte and granulocyte activation across glucose tolerance statuses in mixed ancestry South Africans. Individuals (206) were recruited from Bellville-South, Cape Town, and included 66% with normal glucose tolerance, 18.7% pre-diabetes, 8.7% screen-detected diabetes and 6.3% known diabetes. Monocyte and neutrophil activation were measured by calculating the percentage of cells expressing CD142 and CD69 while platelet monocyte aggregates were defined as CD14++ CD42b+ events and platelet neutrophil aggregates as CD16++ CD42b+ events. The percentage of monocytes and neutrophils expressing CD69 and CD142 was significantly higher in known diabetes and prediabetes, but, lowest in screen-detected diabetes (both p ≤ 0.016). The pattern was similar for platelet monocyte and neutrophil aggregates (both p ≤ 0.003). In robust linear regressions adjusted for age and gender, known diabetes was significantly and positively associated with the percentage of monocytes expressing CD69 [beta 11.06 (p = 0.016)] and CD42b (PMAs) [19.51 (0.003)] as well as the percentage of neutrophils expressing CD69 [14.19 (<0.0001)] and CD42b [17.7 (0.001)]. We conclude that monitoring platelet activation in diagnosed diabetic patients may have a role in the management and risk stratification.
Collapse
Affiliation(s)
- Glenda M Davison
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Bongani B Nkambule
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Zibusiso Mkandla
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Gloudina M Hon
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Andre P Kengne
- NonCommunicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Rajiv T Erasmus
- Department of Pathology, Faculty of Medicine and Health Sciences, National Health Laboratory Service (NHLS) and Stellenbosch University, Cape Town, South Africa
| | - Tandi E Matsha
- Department of Biomedical sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| |
Collapse
|
32
|
Huang K, Gao J, Du J, Ma N, Zhu Y, Wu P, Zhang T, Wang W, Li Y, Chen Q, Hutchins AP, Yang Z, Zheng Y, Zhang J, Shan Y, Li X, Liao B, Liu J, Wang J, Liu B, Pan G. Generation and Analysis of GATA2 w/eGFP Human ESCs Reveal ITGB3/CD61 as a Reliable Marker for Defining Hemogenic Endothelial Cells during Hematopoiesis. Stem Cell Reports 2016; 7:854-868. [PMID: 27746115 PMCID: PMC5106517 DOI: 10.1016/j.stemcr.2016.09.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 11/29/2022] Open
Abstract
The transition from hemogenic endothelial cells (HECs) to hematopoietic stem/progenitor cells (HS/PCs), or endothelial to hematopoietic transition (EHT), is a critical step during hematopoiesis. However, little is known about the molecular determinants of HECs due to the challenge in defining HECs. We report here the generation of GATA2w/eGFP reporter in human embryonic stem cells (hESCs) to mark cells expressing GATA2, a critical gene for EHT. We show that during differentiation, functional HECs are almost exclusively GATA2/eGFP+. We then constructed a regulatory network for HEC determination and also identified a panel of positive or negative surface markers for discriminating HECs from non-hemogenic ECs. Among them, ITGB3 (CD61) precisely labeled HECs both in hESC differentiation and embryonic day 10 mouse embryos. These results not only identify a reliable marker for defining HECs, but also establish a robust platform for dissecting hematopoiesis in vitro, which might lead to the generation of HSCs in vitro. Generation of GATA2w/eGFP reporter in human ESCs GATA2/eGFP expression defines HECs and HPCs in hESC differentiation CD61 marks HECs and HPCs in hPSC differentiation CD61 defines HECs in YS and AGM E10 mouse embryo
Collapse
Affiliation(s)
- Ke Huang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jiao Gao
- Translational Medicine Center for Stem Cells, 307-Ivy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Juan Du
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ning Ma
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yanling Zhu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Pengfei Wu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Tian Zhang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenqian Wang
- Department of Hematology, Sun-yat Sen University, Guangzhou 510630, China
| | - Yuhang Li
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Qianyu Chen
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Andrew Paul Hutchins
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhongzhou Yang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yi Zheng
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jian Zhang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yongli Shan
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xuejia Li
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Baojian Liao
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jiajun Liu
- Department of Hematology, Sun-yat Sen University, Guangzhou 510630, China
| | - Jinyong Wang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Bing Liu
- Translational Medicine Center for Stem Cells, 307-Ivy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Guangjin Pan
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|