1
|
Ratnapriya S, Yabaji SM. Vaccination and Platelet Biology: Unraveling the Immuno-Hemostatic Interplay. Vaccines (Basel) 2025; 13:403. [PMID: 40333325 PMCID: PMC12031077 DOI: 10.3390/vaccines13040403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
Platelets, which have been traditionally associated with hemostasis and thrombosis functions, now receive attention for their role in immune responses that may affect vaccine development and effectiveness. Through their interactions with immune cells and modulation of inflammation alongside their role in antigen presentation, platelets become integral components of both innate and adaptive immune systems. New research shows platelets can improve vaccine effectiveness while reducing adverse side effects. During vaccine administration, platelets release cytokines and chemokines, which attract and stimulate immune cells to the injection site. Platelets work together with dendritic cells and T cells to support antigen processing and presentation, which leads to strong immune activation. Platelets' pro-inflammatory mediators strengthen local immune responses to boost protective immunity generation. Significant attention has been given to platelet involvement in vaccine-related thrombotic events, including vaccine-induced immune thrombotic thrombocytopenia (VITT). The rarity and severity of these events demonstrate the need to investigate the complex interplay between vaccine mechanisms and platelet activation. Exploration of the platelet-immune axis can lead to new methods for improving both the effectiveness and safety of vaccines. Researchers are working on creating innovative approaches for treatments that target platelet receptors and thrombosis pathways without interfering with the regular hemostatic functions of platelets. New vaccine development methods and personalized immunization strategies can emerge from targeting platelets with adjuvants and immune modulators.
Collapse
Affiliation(s)
- Sneha Ratnapriya
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shivraj M. Yabaji
- The National Emerging Infectious Diseases Laboratory, Boston University, Boston, MA 02215, USA
| |
Collapse
|
2
|
Ma Y, Meng F, Lin Z, Chen Y, Lan T, Yang Z, Diao R, Zhang X, Chen Q, Zhang C, Tian Y, Li C, Fang W, Liang X, Zhang X. Bioengineering Platelets Presenting PD-L1, Galectin-9 and BTLA to Ameliorate Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501139. [PMID: 40019367 PMCID: PMC12021092 DOI: 10.1002/advs.202501139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Autoimmune destruction of pancreatic β-cells leads to impaired insulin production and onset of type 1 diabetes (T1D). Hence, immunomodulation of pancreas-infiltrated immune cells especially the β-cells autoreactive-T cells is a promising way to hinder and reverse the progress of T1D. Herein, megakaryocytes are primed with interferon-γ (IFN-γ) to produce platelets presenting high levels of immunosuppressive checkpoint ligands including programmed death-ligand 1 (PD-L1), Programmed Death-Ligand 2 (PD-L2), the B and T lymphocyte attenuator (BTLA) and Galectin-9 (Gal-9), termed as IFN-γ platelets. The IFN-γ platelets bound and interacted with T cells through immune checkpoint ligands and receptors, which efficaciously induced T cell exhaustion and apoptosis in vitro. Virtually, NOD diabetes mice received IFN-γ platelets treatments prominently preserved β-cell integrity and insulin production, ultimately hindering the progress to hyperglycemia. Intriguingly, both the amount and activity of the pancreas infiltrate-T cells intensively reduced, whereas the magnitude of regulatory T cells (Tregs) remarkably increased, which is attributed to IFN-γ platelets treatments. Moreover, IFN-γ platelets treatment instigated macrophage polarization toward an anti-inflammatory M2 phenotype that may stimulate pancreatic angiogenesis, and promote β-cell proliferation, consequently ameliorating the new-onset T1D.
Collapse
Grants
- 32371425 National Natural Science Foundation of China
- 32201084 National Natural Science Foundation of China
- JCYJ20240813151128037 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- RCYX20200714114643121 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20200109142610136 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20180507181654186 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- ZDSYS20220606100803007 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- 2022A1515012289 Natural Science Foundation of Guangdong Province
- GDMUB2022037 Doctoral personnel scientific research start-up Fund project of Guangdong Medical University
- 2024ZDZX2069 Key Field Special Programs of Guangdong Provincial Ordinary Colleges and Universities
- GDMULCJC2024114 Special Project for Clinical and Basic Sci & Tech Innovation of Guangdong Medical University
- National Natural Science Foundation of China
- Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Yumeng Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Fanqiang Meng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhongda Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yanjun Chen
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Tianyu Lan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhaoxin Yang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Rui Diao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xiaozhou Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Chi Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yishi Tian
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Chanjuan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Wenli Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| |
Collapse
|
3
|
Li M, Nie Y, Yang Z. The association between systemic immune-inflammation index (SII) and early nosocomial infections after cardiopulmonary bypass surgery in children with congenital heart disease. BMC Cardiovasc Disord 2024; 24:698. [PMID: 39633275 PMCID: PMC11619183 DOI: 10.1186/s12872-024-04378-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: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Infections occurring postoperatively in pediatric patients with congenital heart disease (CHD) following cardiopulmonary bypass (CPB) surgery pose a considerable challenge, affecting the duration of hospitalization, financial costs, and patient outcomes. Studies investigating the association between systemic immune-inflammation index (SII) and early infections after CHD surgery are very rare. This study seeks to delineate the link between SII and the occurrence of early nosocomial infections in pediatric patients undergoing CPB surgery for CHD. METHODS A cross-sectional analysis was performed on 325 pediatric patients who underwent CPB surgery for CHD between July 2020 and June 2023. The primary exposure was the SII value on the first postoperative day. The outcome was the occurrence of nosocomial infections within the first week following CPB surgery. Multivariable logistic regression models and subgroup analyses were employed to evaluate the association between SII and the risk of early nosocomial infections. RESULTS The median age of the study cohort was 4.4 years, with a male preponderance of 51.7%. The median SII value was recorded at 0.6 × 10^12/L. The rate of nosocomial infections within the first week post-CPB surgery was 53.5%. An inverse association was observed between SII and the incidence of early nosocomial infections. After controlling for multiple confounders, an increment of 1 × 10^12/L in SII corresponded to a 25% reduction in the likelihood of nosocomial infections (OR = 0.75; 95%CI: 0.57, 0.99; P = 0.044). Subgroup analyses substantiated the consistency of these findings. CONCLUSIONS The study demonstrated that an elevated SII corresponded to a reduced likelihood of early nosocomial infections after CPB surgery in children with CHD, a finding that merits additional investigation.
Collapse
Affiliation(s)
- Mei Li
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center, Pediatric Clinical Medical Research Center of Guangxi, Nanning, 530022, China
| | - Yijun Nie
- Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Zhiyong Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center, Pediatric Clinical Medical Research Center of Guangxi, Nanning, 530022, China.
| |
Collapse
|
4
|
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
|
5
|
Tokarz-Deptuła B, Baraniecki Ł, Palma J, Stosik M, Deptuła W. Characterization of Platelet Receptors and Their Involvement in Immune Activation of These Cells. Int J Mol Sci 2024; 25:12611. [PMID: 39684330 DOI: 10.3390/ijms252312611] [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: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The article characterises platelets, pointing out the role and contribution of their numerous receptors determining their specific and broad immune activity. Three types of platelet receptors are described, that is, extracellular and intracellular receptors-TLR (toll-like receptors), NLR (NOD-like receptor), and RLR (RIG-I-like receptor); extracellular receptors-selectins and integrins; and their other extracellular receptors-CLR (C-type lectin receptor), CD (cluster of differentiation), TNF (tumour necrosis factor), among others. Outlining the contribution of these numerous platelet receptors to the intravascular immunity, it has been shown that they are formed by their fusion with pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and lifestyle-associated molecular patterns (LAMPs). They are initiating and effector components of signal transduction of these cells, and their expression and quantity determine the specific and broad functions of platelets towards influencing vascular endothelial cells, but mainly PRRs (pattern recognition receptors) of blood immune cells. These facts make platelets the fundamental elements that shape not only intravascular homeostasis, as previously indicated, but they become the determinants of immunity in blood vessels. Describing the reactions of the characterised three groups of platelet receptors with PAMP, DAMP and LAMP molecules, the pathways and participation of platelets in the formation and construction of intravascular immune status, in physiological states, but mainly in pathological states, including bacterial and viral infections, are presented, making these cells essential elements in the health and disease of mammals, including humans.
Collapse
Affiliation(s)
| | - Łukasz Baraniecki
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Doctoral School, University of Szczecin, 70-384 Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, 65-516 Zielona Góra, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| |
Collapse
|
6
|
Pezzanite LM, Chow L, Dow SW, Goodrich LR, Gilbertie JM, Schnabel LV. Antimicrobial Properties of Equine Stromal Cells and Platelets and Future Directions. Vet Clin North Am Equine Pract 2023; 39:565-578. [PMID: 37442729 DOI: 10.1016/j.cveq.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023] Open
Abstract
Increasing antimicrobial resistance in veterinary practice has driven the investigation of novel therapeutic strategies including regenerative and biologic therapies to treat bacterial infection. Integration of biological approaches such as platelet lysate and mesenchymal stromal cell (MSC) therapy may represent adjunctive treatment strategies for bacterial infections that minimize systemic side effects and local tissue toxicity associated with traditional antibiotics and that are not subject to antibiotic resistance. In this review, we will discuss mechanisms by which biological therapies exert antimicrobial effects, as well as potential applications and challenges in clinical implementation in equine practice.
Collapse
Affiliation(s)
- Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Steven W Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Laurie R Goodrich
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jessica M Gilbertie
- Department of Microbiology and Immunology, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA
| | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
7
|
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
|
8
|
Gautam I, Huss CW, Storad ZA, Krebs M, Bassiouni O, Ramesh R, Wuescher LM, Worth RG. Activated Platelets Mediate Monocyte Killing of Klebsiella pneumoniae. Infect Immun 2023; 91:e0055622. [PMID: 36853027 PMCID: PMC10016073 DOI: 10.1128/iai.00556-22] [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: 12/09/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Platelets are known for essential activities in hemostasis and for their important contribution to protection against infectious pathogens. Klebsiella pneumoniae is an opportunistic pathogen widely known to cause nosocomial infections. Recently, hypervirulent strains of K. pneumoniae have been emerging, which can cause severe infections in immunocompetent individuals. Combined with the increase in antibiotic resistance, it is important to understand how K. pneumoniae affects components of the immune system. We studied the interactions of human platelets with several K. pneumoniae strains (the wild type encapsulated strain, and a nonencapsulated mutant). Thrombin-stimulated whole human and mouse blood significantly inhibited bacterial growth compared to unstimulated whole blood. Furthermore, we investigated the effect of K. pneumoniae on platelet activation. Both strains induced significant increase in activation of both unstimulated and thrombin-stimulated human platelets. Additionally, only the nonencapsulated mutant increased aggregation of platelets in response to ADP. K. pneumoniae killing assays were then performed with washed platelets in the presence or absence of thrombin. Surprisingly, washed platelets failed to exhibit any effects on the growth of K. pneumoniae. We further explored the impact of platelets on monocyte-mediated killing of K. pneumoniae. Importantly, we found that activated platelets significantly enhanced monocyte-mediated killing of K. pneumoniae. This effect was likely due to the formation of platelet-monocyte aggregates in blood upon thrombin stimulation. Overall, this study highlights the role of platelets in mediating a protective response against K. pneumoniae and reinforces the importance of platelets in modulating leukocyte behavior.
Collapse
Affiliation(s)
- Iluja Gautam
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Chadwick W. Huss
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Zachary A. Storad
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Michelle Krebs
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Omar Bassiouni
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Rochan Ramesh
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Randall G. Worth
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
9
|
McHenry ML, Simmons J, Hong H, Malone LL, Mayanja-Kizza H, Bush WS, Boom WH, Hawn TR, Williams SM, Stein CM. Tuberculosis severity associates with variants and eQTLs related to vascular biology and infection-induced inflammation. PLoS Genet 2023; 19:e1010387. [PMID: 36972313 PMCID: PMC10079228 DOI: 10.1371/journal.pgen.1010387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 04/06/2023] [Accepted: 02/13/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Tuberculosis (TB) remains a major public health problem globally, even compared to COVID-19. Genome-wide studies have failed to discover genes that explain a large proportion of genetic risk for adult pulmonary TB, and even fewer have examined genetic factors underlying TB severity, an intermediate trait impacting disease experience, quality of life, and risk of mortality. No prior severity analyses used a genome-wide approach. METHODS AND FINDINGS As part of our ongoing household contact study in Kampala, Uganda, we conducted a genome-wide association study (GWAS) of TB severity measured by TBScore, in two independent cohorts of culture-confirmed adult TB cases (n = 149 and n = 179). We identified 3 SNPs (P<1.0 x 10-7) including one on chromosome 5, rs1848553, that was GWAS significant (meta-analysis p = 2.97x10-8). All three SNPs are in introns of RGS7BP and have effect sizes corresponding to clinically meaningful reductions in disease severity. RGS7BP is highly expressed in blood vessels and plays a role in infectious disease pathogenesis. Other genes with suggestive associations defined gene sets involved in platelet homeostasis and transport of organic anions. To explore functional implications of the TB severity-associated variants, we conducted eQTL analyses using expression data from Mtb-stimulated monocyte-derived macrophages. A single variant (rs2976562) associated with monocyte SLA expression (p = 0.03) and subsequent analyses indicated that SLA downregulation following MTB stimulation associated with increased TB severity. Src Like Adaptor (SLAP-1), encoded by SLA, is highly expressed in immune cells and negatively regulates T cell receptor signaling, providing a potential mechanistic link to TB severity. CONCLUSIONS These analyses reveal new insights into the genetics of TB severity with regulation of platelet homeostasis and vascular biology being central to consequences for active TB patients. This analysis also reveals genes that regulate inflammation can lead to differences in severity. Our findings provide an important step in improving TB patient outcomes.
Collapse
Affiliation(s)
- Michael L. McHenry
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Jason Simmons
- TB Research & Training Center, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Hyejeong Hong
- Department of Biobehavioral Health Sciences, University of Pennsylvania School of Nursing, Philadelphia, Pennsylvania, United States of America
| | - LaShaunda L. Malone
- Tuberculosis Research Unit, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Harriet Mayanja-Kizza
- Department of Medicine and Mulago Hospital, School of Medicine, Makerere University, Kampala, Uganda
| | - William S. Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - W. Henry Boom
- Tuberculosis Research Unit, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Thomas R. Hawn
- TB Research & Training Center, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Scott M. Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Cleveland institute for Computational Biology, Cleveland, Ohio, United States of America
| | - Catherine M. Stein
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Tuberculosis Research Unit, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
10
|
Ippolito MM, Kabuya JBB, Hauser M, Kamavu LK, Banda PM, Yanek LR, Malik R, Mulenga M, Bailey JA, Chongwe G, Louis TA, Shapiro TA, Moss WJ. Whole Blood Transfusion for Severe Malarial Anemia in a High Plasmodium falciparum Transmission Setting. Clin Infect Dis 2022; 75:1893-1902. [PMID: 35439307 PMCID: PMC10200327 DOI: 10.1093/cid/ciac304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Severe malaria resulting from Plasmodium falciparum infection is the leading parasitic cause of death in children worldwide, and severe malarial anemia (SMA) is the most common clinical presentation. The evidence in support of current blood transfusion guidelines for patients with SMA is limited. METHODS We conducted a retrospective cohort study of 911 hospitalized children with SMA in a holoendemic region of Zambia to examine the association of whole blood transfusion with in-hospital survival. Data were analyzed in adjusted logistic regression models using multiple imputation for missing data. RESULTS The median age of patients was 24 months (interquartile range, 16-30) and overall case fatality was 16%. Blood transfusion was associated with 35% reduced odds of death in children with SMA (odds ratio, 0.65; 95% confidence interval, .52-.81; P = .0002) corresponding to a number-needed-to-treat (NNT) of 14 patients. Children with SMA complicated by thrombocytopenia were more likely to benefit from transfusion than those without thrombocytopenia (NNT = 5). Longer storage time of whole blood was negatively associated with survival and with the posttransfusion rise in the platelet count but was not associated with the posttransfusion change in hemoglobin concentration. CONCLUSIONS Whole blood given to pediatric patients with SMA was associated with improved survival, mainly among those with thrombocytopenia who received whole blood stored for <4 weeks. These findings point to a potential use for incorporating thrombocytopenia into clinical decision making and management of severe malaria, which can be further assessed in prospective studies, and underline the importance of maintaining reliable blood donation networks in areas of high malaria transmission.
Collapse
Affiliation(s)
- Matthew M Ippolito
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jean-Bertin B Kabuya
- Department of Clinical Sciences, Tropical Diseases Research Centre, Ndola, Zambia
| | - Manuela Hauser
- Faculty of Medicine, University of Basel, Basel, Switzerland
- Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Luc K Kamavu
- Saint Paul’s General Hospital, Nchelenge, Luapula Province, Zambia
| | | | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rubab Malik
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Modest Mulenga
- Directorate of Research and Postgraduate Studies, Lusaka Apex Medical University, Lusaka, Zambia
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Providence, Rhode Island, USA
| | - Gershom Chongwe
- Department of Clinical Sciences, Tropical Diseases Research Centre, Ndola, Zambia
| | - Thomas A Louis
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Theresa A Shapiro
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William J Moss
- The Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | |
Collapse
|
11
|
Indications that the Antimycotic Drug Amphotericin B Enhances the Impact of Platelets on Aspergillus. Antimicrob Agents Chemother 2022; 66:e0068122. [PMID: 36190233 PMCID: PMC9578436 DOI: 10.1128/aac.00681-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets are currently thought to harbor antimicrobial functions and might therefore play a crucial role in infections, e.g., those caused by Aspergillus or mucormycetes. The incidence of invasive fungal infections is increasing, particularly during the coronavirus disease 2019 (COVID-19) pandemic, and such infections continue to be life-threatening in immunocompromised patients. For this reason, the interaction of antimycotics with platelets is a key issue to evaluate modern therapeutic regimens. Amphotericin B (AmB) is widely used for the therapy of invasive fungal infections either as deoxycholate (AmB-D) or as a liposomal formulation (L-AmB). We showed that AmB strongly activates platelets within a few minutes. AmB concentrations commonly measured in the blood of patients were sufficient to stimulate platelets, indicating that this effect is highly relevant in vivo. The stimulating effect was corroborated by a broad spectrum of platelet activation parameters, including degranulation, aggregation, budding of microparticles, morphological changes, and enhanced adherence to fungal hyphae. Comparison between the deoxycholate and the liposomal formulation excluded the possibility that the liposomal part of L-Amb is responsible for these effects, as no difference was visible. The induction of platelet activation and alteration by L-AmB resulted in the activation of other parts of innate immunity, such as stimulation of the complement cascade and interaction with granulocytes. These mechanisms might substantially fuel the antifungal immune reaction in invasive mycoses. On the other hand, thrombosis and excessive inflammatory processes might occur via these mechanisms. Furthermore, the viability of L-AmB-activated platelets was consequently decreased, a process that might contribute to thrombocytopenia in patients.
Collapse
|
12
|
An Artificial Intelligence-guided signature reveals the shared host immune response in MIS-C and Kawasaki disease. Nat Commun 2022; 13:2687. [PMID: 35577777 PMCID: PMC9110726 DOI: 10.1038/s41467-022-30357-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
AbstractMultisystem inflammatory syndrome in children (MIS-C) is an illness that emerged amidst the COVID-19 pandemic but shares many clinical features with the pre-pandemic syndrome of Kawasaki disease (KD). Here we compare the two syndromes using a computational toolbox of two gene signatures that were developed in the context of SARS-CoV-2 infection, i.e., the viral pandemic (ViP) and severe-ViP signatures and a 13-transcript signature previously demonstrated to be diagnostic for KD, and validated our findings in whole blood RNA sequences, serum cytokines, and formalin fixed heart tissues. Results show that KD and MIS-C are on the same continuum of the host immune response as COVID-19. Both the pediatric syndromes converge upon an IL15/IL15RA-centric cytokine storm, suggestive of shared proximal pathways of immunopathogenesis; however, they diverge in other laboratory parameters and cardiac phenotypes. The ViP signatures reveal unique targetable cytokine pathways in MIS-C, place MIS-C farther along in the spectrum in severity compared to KD and pinpoint key clinical (reduced cardiac function) and laboratory (thrombocytopenia and eosinopenia) parameters that can be useful to monitor severity.
Collapse
|
13
|
BTK Inhibitors Impair Platelet-Mediated Antifungal Activity. Cells 2022; 11:cells11061003. [PMID: 35326454 PMCID: PMC8947638 DOI: 10.3390/cells11061003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 01/27/2023] Open
Abstract
In recent years, the introduction of new drugs targeting Bruton’s tyrosine kinase (BTK) has allowed dramatic improvement in the prognosis of patients with chronic lymphocytic leukemia (CLL) and other B-cell neoplasms. Although these small molecules were initially considered less immunosuppressive than chemoimmunotherapy, an increasing number of reports have described the occurrence of unexpected opportunistic fungal infections, in particular invasive aspergillosis (IA). BTK represents a crucial molecule in several signaling pathways depending on different immune receptors. Based on a variety of specific off-target effects on innate immunity, namely on neutrophils, monocytes, pulmonary macrophages, and nurse-like cells, ibrutinib has been proposed as a new host factor for the definition of probable invasive pulmonary mold disease. The role of platelets in the control of fungal growth, through granule-dependent mechanisms, was described in vitro almost two decades ago and is, so far, neglected by experts in the field of clinical management of IA. In the present study, we confirm the antifungal role of platelets, and we show, for the first time, that the exposure to BTK inhibitors impairs several immune functions of platelets in response to Aspergillus fumigatus, i.e., the ability to adhere to conidia, activation (as indicated by reduced expression of P-selectin), and direct killing activity. In conclusion, our experimental data suggest that antiplatelet effects of BTK inhibitors may contribute to an increased risk for IA in CLL patients.
Collapse
|
14
|
Aznab M, Eskandari Roozbahani N, Moazen H. Clinical Characteristics and Risk Factors of COVID-19 in 60 Adult Cancer Patients. Clin Med Insights Oncol 2022; 16:11795549221074168. [PMID: 35110966 PMCID: PMC8801664 DOI: 10.1177/11795549221074168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/22/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND During the pandemic of COVID-19, cancer patients have been considered as one high-risk group in the morbidity and mortality of COVID-19. This study aimed to describe the clinical symptoms and risk factors of COVID-19 in cancer patients. METHOD In a prospective cross-sectional study, during a year, all cancer patients who underwent chemotherapy and/or targeted therapy in our clinic (Kermanshah, Iran) were followed up in terms of getting COVID-19. We analyzed the effect of tumor features and demographic information on clinical manifestations, survival status, therapeutic outcomes, and severity of the disease COVID-19 in 2 categories of cancer (hematologic and solid cancers). RESULTS Most of the patients (68%) were in the solid tumor category, including breast cancer (24.4%), colon cancer (22%), and gastric cancer (9.8%). There was a statistically significant difference between 2 categories of cancer in the clinical manifestations: the stage of cancer and survival status (P < .05). Logistic regression analysis showed that the risk of death in cancer patients with COVID-19 along with symptoms of diarrhea (odds ratio [OR] = 12.8, P = .004), the difficulty of breath (OR = 10.73, P = .034), drop of SO2 (OR = 1.334, P = .003), thrombocytopenia (OR = 1.022, P = .02), anemia (OR = 2.72, P = .011), requiring mechanical ventilation (OR = 9.24, P = .004), pleural infusion (OR = 10.28, P = .02), and intensive care unit (ICU) admission (OR = 7.389, P = .009) increases independent of other variables. The COVID-19 mortality rate in our cancer patients was 23%. CONCLUSIONS Thrombocytopenia, anemia, and diarrhea are symptoms that, along with common symptoms such as lung involvement, difficulty breathing, and the need for a ventilator, increase the risk of death in cancer patients with COVID-19.
Collapse
Affiliation(s)
- Mozaffar Aznab
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Narges Eskandari Roozbahani
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Homa Moazen
- Department of Biostatics and Epidemiology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
15
|
McHenry ML, Wampande EM, Joloba ML, Malone LL, Mayanja-Kizza H, Bush WS, Boom WH, Williams SM, Stein CM. Interaction between M. tuberculosis Lineage and Human Genetic Variants Reveals Novel Pathway Associations with Severity of TB. Pathogens 2021; 10:pathogens10111487. [PMID: 34832643 PMCID: PMC8617877 DOI: 10.3390/pathogens10111487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Tuberculosis (TB) remains a major public health threat globally, especially in sub-Saharan Africa. Both human and Mycobacterium tuberculosis (MTBC) genetic variation affect TB outcomes, but few studies have examined if and how the two genomes interact to affect disease. We hypothesize that long-term coexistence between human genomes and MTBC lineages modulates disease to affect its severity. We examined this hypothesis in our TB household contact study in Kampala, Uganda, in which we identified three MTBC lineages, of which one, L4.6-Uganda, is clearly derived and hence recent. We quantified TB severity using the Bandim TBscore and examined the interaction between MTBC lineage and human single-nucleotide polymorphisms (SNPs) genome-wide, in two independent cohorts of TB cases (n = 149 and n = 127). We found a significant interaction between an SNP in PPIAP2 and the Uganda lineage (combined p = 4 × 10−8). PPIAP2 is a pseudogene that is highly expressed in immune cells. Pathway and eQTL analyses indicated potential roles between coevolving SNPs and cellular replication and metabolism as well as platelet aggregation and coagulation. This finding provides further evidence that host–pathogen interactions affect clinical presentation differently than host and pathogen genetic variation independently, and that human–MTBC coevolution is likely to explain patterns of disease severity.
Collapse
Affiliation(s)
- Michael L. McHenry
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44016, USA; (M.L.M.); (W.S.B.); (S.M.W.)
| | - Eddie M. Wampande
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda; (E.M.W.); (M.L.J.)
| | - Moses L. Joloba
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda; (E.M.W.); (M.L.J.)
| | - LaShaunda L. Malone
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (L.L.M.); (W.H.B.)
| | - Harriet Mayanja-Kizza
- Department of Medicine and Mulago Hospital, School of Medicine, Makerere University, Kampala, Uganda;
| | - William S. Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44016, USA; (M.L.M.); (W.S.B.); (S.M.W.)
| | - W. Henry Boom
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (L.L.M.); (W.H.B.)
| | - Scott M. Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44016, USA; (M.L.M.); (W.S.B.); (S.M.W.)
| | - Catherine M. Stein
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44016, USA; (M.L.M.); (W.S.B.); (S.M.W.)
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda; (E.M.W.); (M.L.J.)
- Correspondence:
| |
Collapse
|
16
|
Padilla S, Nurden AT, Prado R, Nurden P, Anitua E. Healing through the lens of immunothrombosis: Biology-inspired, evolution-tailored, and human-engineered biomimetic therapies. Biomaterials 2021; 279:121205. [PMID: 34710794 DOI: 10.1016/j.biomaterials.2021.121205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
Evolution, from invertebrates to mammals, has yielded and shaped immunoclotting as a defense and repair response against trauma and infection. This mosaic of immediate and local wound-sealing and pathogen-killing mechanisms results in survival, restoration of homeostasis, and tissue repair. In mammals, immunoclotting has been complemented with the neuroendocrine system, platelets, and contact system among other embellishments, adding layers of complexity through interconnecting blood-born proteolytic cascades, blood cells, and the neuroendocrine system. In doing so, immunothrombosis endows humans with survival advantages, but entails vulnerabilities in the current unprecedented and increasingly challenging environment. Immunothrombosis and tissue repair appear to go hand in hand with common mechanisms mediating both processes, a fact that is underlined by recent advances that are deciphering the mechanisms of the repair process and of the biochemical pathways that underpins coagulation, hemostasis and thrombosis. This review is intended to frame both the universal aspects of tissue repair and the therapeutic use of autologous fibrin matrix as a biology-as-a-drug approach in the context of the evolutionary changes in coagulation and hemostasis. In addition, we will try to shed some light on the molecular mechanisms underlying the use of the autologous fibrin matrix as a biology-inspired, evolution-tailored, and human-engineered biomimetic therapy.
Collapse
Affiliation(s)
- Sabino Padilla
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| | - Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| | - Roberto Prado
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Paquita Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| | - Eduardo Anitua
- Eduardo Anitua Foundation for Biomedical Research, Vitoria, Spain; BTI-Biotechnology Institute ImasD, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| |
Collapse
|
17
|
Pudakalakatti S, Audia A, Mukhopadhyay A, Enriquez JS, Bourgeois D, Tayob N, Zacharias NM, Millward SW, Carson D, Farach-Carson MC, Lang FF, Heimberger AB, Bhat KP, Bhattacharya PK. NMR Spectroscopy-Based Metabolomics of Platelets to Analyze Brain Tumors. REPORTS 2021; 4. [PMID: 35937580 PMCID: PMC9352435 DOI: 10.3390/reports4040032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
“Tumor-educated platelets” have recently generated substantial interest for the diagnosis of cancer. We hypothesized that tumor educated platelets from patients with brain tumors will reflect altered metabolism compared to platelets from healthy volunteers. Here, in a pilot study, we have employed nuclear magnetic resonance (NMR) spectroscopy in platelets from brain tumor patients to demonstrate altered metabolism compared to the platelets obtained from healthy volunteers.
Collapse
Affiliation(s)
- Shivanand Pudakalakatti
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alessandra Audia
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Center Center, Houston, TX 77030, USA
| | - Anirudh Mukhopadhyay
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - José S. Enriquez
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
| | | | - Nabihah Tayob
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Niki M. Zacharias
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven W. Millward
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
| | - Daniel Carson
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Mary C. Farach-Carson
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center, Houston, TX 77054, USA
| | - Frederick F. Lang
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amy B. Heimberger
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Krishna P. Bhat
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Center Center, Houston, TX 77030, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
| | - Pratip K. Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77054, USA
- Correspondence:
| |
Collapse
|
18
|
Császár N, Bókkon I. Gut serotonin as a general membrane permeability regulator. Curr Neuropharmacol 2021; 20:269-271. [PMID: 34548000 PMCID: PMC9413787 DOI: 10.2174/1570159x19666210921100542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/15/2021] [Accepted: 08/15/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Császár N
- Psychosomatic Outpatient Clinics, Budapest. Hungary
| | - Bókkon I
- Psychosomatic Outpatient Clinics, Budapest. Hungary
| |
Collapse
|
19
|
Sahoo D, Katkar GD, Shimizu C, Kim J, Khandelwal S, Tremoulet AH, Kanegaye J, Bocchini J, Das S, Burns JC, Ghosh P. An AI-guided signature reveals the nature of the shared proximal pathways of host immune response in MIS-C and Kawasaki disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33880476 DOI: 10.1101/2021.04.11.439347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A significant surge in cases of multisystem inflammatory syndrome in children (MIS-C, also called Pediatric Inflammatory Multisystem Syndrome - PIMS) has been observed amidst the COVID-19 pandemic. MIS-C shares many clinical features with Kawasaki disease (KD), although clinical course and outcomes are divergent. We analyzed whole blood RNA sequences, serum cytokines, and formalin fixed heart tissues from these patients using a computational toolbox of two gene signatures, i.e., the 166-gene viral pandemic (ViP) signature, and its 20-gene severe (s)ViP subset that were developed in the context of SARS-CoV-2 infection and a 13-transcript signature previously demonstrated to be diagnostic for KD. Our analyses revealed that KD and MIS-C are on the same continuum of the host immune response as COVID-19. While both the pediatric syndromes converge upon an IL15/IL15RA -centric cytokine storm, suggestive of shared proximal pathways of immunopathogenesis, they diverge in other laboratory parameters and cardiac phenotypes. The ViP signatures also revealed unique targetable cytokine pathways in MIS-C, place MIS-C farther along in the spectrum in severity compared to KD and pinpoint key clinical (reduced cardiac function) and laboratory (thrombocytopenia and eosinopenia) parameters that can be useful to monitor severity.
Collapse
|
20
|
Moroșan D, Șerban A, Trifan C, Encica S, Pop S, Șerban TC, Rednic S, Damian L. Frenemies within: An Endocarditis Case in Behçet's Disease. J Pers Med 2021; 11:728. [PMID: 34442371 PMCID: PMC8402229 DOI: 10.3390/jpm11080728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/14/2021] [Accepted: 07/25/2021] [Indexed: 01/05/2023] Open
Abstract
A 57-year female patient diagnosed with Behçet's disease, on azathioprine, was noticed to have at a routine examination antinuclear and antiphospholipid antibodies. An overlapping lupus-like syndrome was diagnosed; hydroxychloroquine and aspirin were added. Three years later, the patient presented with dyspnea and sweating, with no fever. A cardiac bruit was noted; a giant vegetation was detected by echocardiography. Laboratory revealed severe thrombocytopenia, antiphospholipid antibodies and low complement. Blood cultures were positive for Abiotrophia defectiva serology and also revealed a chronic Coxiella burnetii infection. Antibiotic therapy, low-dose anticoagulation and control of the underlying disease mildly improved the platelet count, which fully recovered only after cardiac valve replacement. However, the Behçet's disease, initially quiescent, flared after the therapy of infections. We discuss potential links between Behçet's disease and the occurrence of antinuclear and antiphospholipid antibodies and Coxiella endocarditis in this setting. We also highlight the differences between the endocarditis in Behçet's disease, antiphospholipid syndrome, Coxiella burnetii and Abiotrophia defectiva infection, respectively. Intracellular infections may modify the presentation of autoimmune diseases. Confounding clinical features of Coxiella persistent infection and non-bacterial thrombotic endocarditis in Behçet's disease warrant further insight.
Collapse
Affiliation(s)
- Diana Moroșan
- Department of Rheumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babeș St., 400012 Cluj-Napoca, Romania; (D.M.); (S.R.)
| | - Adela Șerban
- Department of Cardiology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babeș St., 400012 Cluj-Napoca, Romania;
- Department of Cardiology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21, Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Cătălin Trifan
- Department of Cardiovascular Surgery, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21, Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Svetlana Encica
- Department of Pathology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21, Calea Moților St., 400001 Cluj-Napoca, Romania
| | - Sorin Pop
- 1st Internal Medicine Department, Emergency Clinical County Hospital Cluj, 3-5 Clinicilor St., 400006 Cluj-Napoca, Romania;
| | - Tudor Costinel Șerban
- Department of Cardiology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21, Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Simona Rednic
- Department of Rheumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babeș St., 400012 Cluj-Napoca, Romania; (D.M.); (S.R.)
- Department of Rheumatology, Centre for Rare Musculoskeletal Autoimmune and Autoinflammatory Diseases, Emergency Clinical County Hospital Cluj, 2-4 Clinicilor St., 400006 Cluj-Napoca, Romania;
| | - Laura Damian
- Department of Rheumatology, Centre for Rare Musculoskeletal Autoimmune and Autoinflammatory Diseases, Emergency Clinical County Hospital Cluj, 2-4 Clinicilor St., 400006 Cluj-Napoca, Romania;
- CMI Reumatologie Dr. Damian, 6-8 P. Maior St., 400002 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Saleh G, Ahmed A, Hassanain O, Emad A, Essameldin S, Ragai M, Saad Y. Nutrition in Cancer Patients Positive for COVID-19; Case Series and a Systematic Review of Literature. Nutr Cancer 2021; 74:450-462. [PMID: 34080508 DOI: 10.1080/01635581.2021.1931363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is making patients vulnerable to diseases by impairing immunity directly or by anticancer therapy. In COVID-19 era, it is mandatory to face cancer with more organized & prompter response. Nutrition plays an important role in prevention & management of cancer patients. The objective of this study is to understand the role of nutrition in cancer patients during Corvid 19 era. We conducted literature searches till May 2020, electronic databases, evidence-based collections, relevant websites and trial registries about SARS-CoV2/COVID-19 and nutrition in cancer patients. Search generated 836 sources; 83/836 sources were relevant. This review summarized role of nutrition in predisposition, prevention and management of COVID-19 in cancer patient and role of vitamins, mineral supplements and microbiota in era of COVID-19. In this review, implementing appropriate nutritional care with vitamins or mineral supplementation & their effect on outcome remain largely unknown. COVID co-infection with cancer whether under chemotherapy or not have worse outcome especially in male adults. Findings may help in creating recommendations on nutritional protocol of management & prevention of complications during ongoing COVID-19 pandemic for all cancer patients.
Collapse
Affiliation(s)
- Gulsen Saleh
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt.,Public Health Department, National Nutrition Institute (NNI), Cairo, Egypt
| | - Aliaa Ahmed
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Tanta, Egypt
| | - Omneya Hassanain
- Epidemiology and Biostatistics Unit, Research Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt
| | - Aya Emad
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt
| | - Samer Essameldin
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt
| | - Marianne Ragai
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt
| | - Yasmin Saad
- Clinical Nutrition Department, 57357 Cancer Children Hospital of Egypt (CCHE), Cairo, Egypt.,Endemic Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
22
|
Pretorius E. Platelets in HIV: A Guardian of Host Defence or Transient Reservoir of the Virus? Front Immunol 2021; 12:649465. [PMID: 33968041 PMCID: PMC8102774 DOI: 10.3389/fimmu.2021.649465] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023] Open
Abstract
The immune and inflammatory responses of platelets to human immunodeficiency virus 1 (HIV-1) and its envelope proteins are of great significance to both the treatment of the infection, and to the comorbidities related to systemic inflammation. Platelets can interact with the HIV-1 virus itself, or with viral membrane proteins, or with dysregulated inflammatory molecules in circulation, ensuing from HIV-1 infection. Platelets can facilitate the inhibition of HIV-1 infection via endogenously-produced inhibitors of HIV-1 replication, or the virus can temporarily hide from the immune system inside platelets, whereby platelets act as HIV-1 reservoirs. Platelets are therefore both guardians of the host defence system, and transient reservoirs of the virus. Such reservoirs may be of particular significance during combination antiretroviral therapy (cART) interruption, as it may drive viral persistence, and result in significant implications for treatment. Both HIV-1 envelope proteins and circulating inflammatory molecules can also initiate platelet complex formation with immune cells and erythrocytes. Complex formation cause platelet hypercoagulation and may lead to an increased thrombotic risk. Ultimately, HIV-1 infection can initiate platelet depletion and thrombocytopenia. Because of their relatively short lifespan, platelets are important signalling entities, and could be targeted more directly during HIV-1 infection and cART.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
23
|
Xu Y, Shao M, Liu N, Tang J, Gu Q, Dong D. Invasive pulmonary aspergillosis is a frequent complication in patients with severe fever with thrombocytopenia syndrome: A retrospective study. Int J Infect Dis 2021; 105:646-652. [PMID: 33640568 DOI: 10.1016/j.ijid.2021.02.088] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Invasive pulmonary aspergillosis (IPA) usually occurs in immunocompromised hosts. It has recently been reported that patients with severe fever with thrombocytopenia syndrome (SFTS) can also develop IPA. The aim of this study was to determine the incidence of IPA in SFTS patients and to investigate the relevant clinical, imaging, and laboratory characteristics. METHODS A retrospective review was conducted of all patients with SFTS admitted to Nanjing Drum Tower Hospital, a tertiary hospital in Nanjing City, China, between January 2016 and December 2019. The patients were divided into two groups according to whether they had IPA: the IPA group and the non-IPA group. Data on clinical manifestations, laboratory findings, imaging characteristics, treatments, and outcomes were collected and analysed. RESULTS A total of 91 SFTS patients were included, of whom 29 (31.9%) developed IPA. In-hospital mortality (22.0%) was higher in the IPA group than in the non-IPA group. Univariate logistic regression showed that diabetes, cough, wheezing, amylase level, vasopressor use, encephalopathy, and intensive care unit transfer were risk factors for the development of IPA. Multivariate logistic regression analysis identified wheezing as an independent predictor of IPA in SFTS patients. CONCLUSIONS SFTS combined with IPA is associated with high morbidity and mortality. It is necessary to strengthen screening for fungal infections after admission in SFTS patients. However, whether early antifungal prophylaxis should be administered needs further investigation.
Collapse
Affiliation(s)
- Ying Xu
- Department of Intensive Care Unit, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Mingran Shao
- Department of Radiology, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ning Liu
- Department of Intensive Care Unit, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jian Tang
- Department of Intensive Care Unit, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Qin Gu
- Department of Intensive Care Unit, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Danjiang Dong
- Department of Intensive Care Unit, the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| |
Collapse
|
24
|
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: 63] [Impact Index Per Article: 15.8] [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
|
25
|
Torres-Juarez F, Trejo-Martínez LA, Layseca-Espinosa E, Leon-Contreras JC, Enciso-Moreno JA, Hernandez-Pando R, Rivas-Santiago B. Platelets immune response against Mycobacterium tuberculosis infection. Microb Pathog 2021; 153:104768. [PMID: 33524564 DOI: 10.1016/j.micpath.2021.104768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/23/2022]
Abstract
Tuberculosis (TB) is the first cause of death by a single infectious agent. Previous reports have highlighted the presence of platelets within Tb granulomas, albeit the immune-associated platelet response to Mycobacterium tuberculosis (Mtb) has not been deeply studied. Our results showed that platelets are recruited into the granuloma in the late stages of tuberculosis. Furthermore, electron-microscopy studies showed that platelets can internalize Mtb and produce host defense peptides (HDPs), such as RNase 7, HBD2 and hPF-4 that bind to the internalized Mtb. Mtb-infected platelets exhibited higher transcription and secretion of IL-1β and TNF-α, whereas IL-10 and IL-6 protein levels decreased. These results suggest that platelets participate in the immune response against Mtb through HDPs and cytokines production.
Collapse
Affiliation(s)
- Flor Torres-Juarez
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico; Laboratory of Immunology, Autonomous University of San Luis Potosí, San Luis Potosi, Mexico
| | - Luis A Trejo-Martínez
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico
| | | | - Juan C Leon-Contreras
- Laboratory of Experimental Pathology, Nacional Institute of Medical Sciences and Nutrition "Salvador Zubiran", CDMX, Mexico
| | - Jose A Enciso-Moreno
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico
| | - Rogelio Hernandez-Pando
- Laboratory of Experimental Pathology, Nacional Institute of Medical Sciences and Nutrition "Salvador Zubiran", CDMX, Mexico
| | - Bruno Rivas-Santiago
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico.
| |
Collapse
|
26
|
Getawa S, Getaneh Z, Melku M. Hematological Abnormalities and Associated Factors Among Undernourished Under-Five Children Attending University of Gondar Specialized Referral Hospital, Northwest Ethiopia. J Blood Med 2020; 11:465-478. [PMID: 33376434 PMCID: PMC7755328 DOI: 10.2147/jbm.s284572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/05/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction The hematopoietic system is one of the systems which can be affected by malnutrition, leading to impaired production of all blood cell lines. Undernourished children with hematological abnormalities like anemia are at higher risk of mortality. Therefore, this study aimed at determining the magnitude and associated factors of hematological abnormalities among undernourished under-five children attending at the University of Gondar Specialized Referral Hospital, Northwest Ethiopia. Methods and Materials An institution-based cross-sectional study was conducted on a total of 251 undernourished under-five children at University of Gondar Specialized Referral Hospital from January to May 2020. A convenient sampling technique was employed to select study participants. Data were collected using a pretested structured questionnaire. Blood samples were collected for complete blood count which were determined by Sysmex KX-21N analyzer. Stool sample was processed via direct wet mount. Thin and thick blood films were examined to assess malaria parasite. The data were entered to EpiData version 4.6.0.0 and analyzed using SPSS version 23 software. Bi-variable and multi-variable binary logistic regression model were fitted to identify factors associated with hematological abnormalities. A p-value <0.05 in the multivariable analysis was considered as statistically significant. Results The overall magnitude of anemia, leukocytosis, thrombocytosis, thrombocytopenia, and leukopenia was 53.4%, 26.7%, 23.9%, 8%, and 2.8%, respectively. Being male, age 6-23 months, high birth order, intestinal parasite infection, edema, not eating vegetables and fruits, and paternal occupation were found to be associated with anemia. Only the age of a child was associated with leukocytosis in undernourished children. Conclusion The current study demonstrated the predominant existence of anemia, leukocytosis, and thrombocytosis among undernourished under-five children. Therefore, early diagnosis, monitoring and setting intervention strategies for anemia especially among children under two years old are required to prevent further complication.
Collapse
Affiliation(s)
- Solomon Getawa
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Zegeye Getaneh
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Melku
- Department of Hematology and Immunohematology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
27
|
Bourdon M, Santulli P, Jeljeli M, Vannuccini S, Marcellin L, Doridot L, Petraglia F, Batteux F, Chapron C. Immunological changes associated with adenomyosis: a systematic review. Hum Reprod Update 2020; 27:108-129. [PMID: 33099635 DOI: 10.1093/humupd/dmaa038] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 07/24/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Adenomyosis is a benign gynecological disorder associated with subfertility, pelvic pain and abnormal uterine bleeding that have significant consequences for the health and quality of life of women. Histologically, it is defined as the presence of ectopic endometrial islets within the myometrium. Its pathogenesis has not yet been elucidated and several pieces of the puzzle are still missing. One process involved in the development of adenomyosis is the increased capacity of some endometrial cells to infiltrate the myometrium. Moreover, the local and systemic immune systems are associated with the onset of the disease and with maintaining it. Numerous observations have highlighted the activation of immune cells and the release of immune soluble factors in adenomyosis. The contribution of immunity occurs in conjunction with hormonal aberrations and activation of the epithelial to mesenchymal transition (EMT) pathway, which promotes migration of endometrial cells. Here, we review current knowledge on the immunological changes in adenomyosis, with the aim of further elucidation of the pathogenesis of this disease. OBJECTIVE AND RATIONALE The objective was to systematically review the literature regarding the role of the immune system in development of adenomyosis in the inner and the outer myometrium, in humans. SEARCH METHODS A systematic review of published human studies was performed in MEDLINE, EMBASE and Cochrane Library databases from 1970 to February 2019 using the combination of Medical Subject Headings (MeSH): Adenomyosis AND ('Immune System' OR 'Gonadal Steroid Hormones'), and free-text terms for the following search terms (and their variants): Adenomyosis AND (immunity OR immune OR macrophage OR 'natural killer cell' OR lymphocyte* OR leucocyte* OR HLA OR inflammation OR 'sex steroid' OR 'epithelial to mesenchymal transition' OR 'EMT'). Studies in which no comparison was made with control patients, without adenomyosis (systemic sample and/or eutopic endometrium), were excluded. OUTCOMES A total of 42 articles were included in our systematic review. Changes in innate and adaptive immune cell numbers were described in the eutopic and/or ectopic endometrium of women with adenomyosis compared to disease-free counterparts. They mostly described an increase in lymphocyte and macrophage cell populations in adenomyosis eutopic endometrium compared to controls. These observations underscore the immune contributions to the disease pathogenesis. Thirty-one cytokines and other markers involved in immune pathways were studied in the included articles. Pro-inflammatory cytokines (interleukin (IL) 6, IL1β, interferon (IFN) α, tumor necrosis factor α, IFNγ) as well as anti-inflammatory or regulatory mediators (IL10, transforming growth factor β…) were found to be elevated in the eutopic endometrium and/or in the ectopic endometrium of the myometrium in women with adenomyosis compared to controls. Moreover, in women affected by adenomyosis, immunity was reported to be directly or indirectly linked to sex steroid hormone aberrations (notably changes in progesterone receptor in eutopic and ectopic endometrium) in three studies and to EMT in four studies. WIDER IMPLICATIONS The available literature clearly depicts immunological changes that are associated with adenomyosis. Both systemic and local immune changes have been described in women affected by adenomyosis, with the coexistence of changes in inflammatory as well as anti-inflammatory signals. It is likely that these immune changes, through an EMT mechanism, stimulate the migration of endometrial cells into the myometrium that, together with an endocrine imbalance, promote this inflammatory process. In light of the considerable impact of adenomyosis on women's health, a better understanding of the role played by the immune system in adenomyosis is likely to yield new research opportunities to better understand its pathogenesis.
Collapse
Affiliation(s)
- M Bourdon
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Gynaecology Obstetrics and Reproductive Medicine, Hopital Cochin, Paris, France
| | - P Santulli
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Gynaecology Obstetrics and Reproductive Medicine, Hopital Cochin, Paris, France
| | - M Jeljeli
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Immunology, Hopital Cochin, Paris, France
| | - S Vannuccini
- Division of Obstetrics and Gynecology, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Viale Morgagni 44, 50134 Florence, Italy.,Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - L Marcellin
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Gynaecology Obstetrics and Reproductive Medicine, Hopital Cochin, Paris, France
| | - L Doridot
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France
| | - F Petraglia
- Division of Obstetrics and Gynecology, Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Viale Morgagni 44, 50134 Florence, Italy
| | - F Batteux
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Immunology, Hopital Cochin, Paris, France
| | - C Chapron
- Université de Paris, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 27, rue du Faubourg-Saint-Jacques, 75015 Paris, France.,Department 3I "Infection, Immunité et inflammation", Institut Cochin, INSERM U1016, Paris, France.,Department of Gynaecology Obstetrics and Reproductive Medicine, Hopital Cochin, Paris, France
| |
Collapse
|
28
|
Deshmukh H, Speth C, Sheppard DC, Neurauter M, Würzner R, Lass-Flörl C, Rambach G. Aspergillus-Derived Galactosaminogalactan Triggers Complement Activation on Human Platelets. Front Immunol 2020; 11:550827. [PMID: 33123129 PMCID: PMC7573070 DOI: 10.3389/fimmu.2020.550827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Invasive fungal infections caused by Aspergillus (A.) and Mucorales species still represent life-threatening diseases in immunocompromised individuals, and deeper knowledge about fungal interactions with elements of innate immunity, such as complement and platelets, appears essential for optimized therapy. Previous studies showed that galactosaminogalactan secreted by A. fumigatus and A. flavus is deposited on platelets, thereby inducing their activation. Since the altered platelet surface is a putative trigger for complement activation, we aimed to study the interplay of platelets with complement in the presence of fungal GAG. Culture supernatants (SN) of A. fumigatus and A. flavus both induced not only GAG deposition but also subsequent deposition of complement C3 fragments on the platelet surface. The SN of a Δuge3 mutant of A. fumigatus, which is unable to synthesize GAG, did not induce complement deposition on platelets, nor did the SN of other Aspergillus species and all tested Mucorales. Detailed analysis revealed that GAG deposition itself triggered the complement cascade rather than the GAG-induced phosphatidylserine exposure. The lectin pathway of complement could be shown to be crucially involved in this process. GAG-induced complement activation on the platelet surface was revealed to trigger processes that might contribute to the pathogenesis of invasive aspergillosis by A. fumigatus or A. flavus. Both pro-inflammatory anaphylatoxins C3a and C5a arose when platelets were incubated with SN of these fungal species; these processes might favor excessive inflammation after fungal infection. Furthermore, platelets were stimulated to shed microparticles, which are also known to harbor pro-inflammatory and pro-coagulant properties. Not only did early processes of the complement cascade proceed on platelets, but also the formation of the terminal complement C5b-9 complex was detected on platelets after incubation with fungal SN. Subsequently, reduced viability of the platelets could be shown, which might contribute to the lowered platelet numbers found in infected patients. In summary, fungal GAG initiates an interplay between complement and platelets that can be supposed to contribute to excessive inflammation, thrombocytopenia, and thrombosis, which are important hallmarks of fatal invasive mycoses.
Collapse
Affiliation(s)
- Hemalata Deshmukh
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Speth
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Donald C Sheppard
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Magdalena Neurauter
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Günter Rambach
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| |
Collapse
|
29
|
Trotta A, Milillo MA, Serafino A, Castillo LA, Birnberg Weiss F, Delpino MV, Giambartolomei GH, Fernández GC, Barrionuevo P. Brucella abortus-infected platelets modulate the activation of neutrophils. Immunol Cell Biol 2020; 98:743-756. [PMID: 32623755 DOI: 10.1111/imcb.12373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 01/18/2023]
Abstract
Brucellosis is a contagious disease caused by bacteria of the genus Brucella. Platelets (PLTs) have been widely involved in the modulation of the immune response. We have previously reported the modulation of Brucella abortus-mediated infection of monocytes. As a result, PLTs cooperate with monocytes and increase their inflammatory capacity, promoting the resolution of the infection. Extending these results, in this study we demonstrate that patients with brucellosis present slightly elevated levels of complexes between PLTs and both monocytes and neutrophils. We then assessed whether PLTs were capable of modulating functional aspects of neutrophils. The presence of PLTs throughout neutrophil infection increased the production of interleukin-8, CD11b surface expression and reactive oxygen species formation, whereas it decreased the expression of CD62L, indicating an activated status of these cells. We next analyzed whether this modulation was mediated by released factors. To discriminate between these options, neutrophils were treated with supernatants collected from B. abortus-infected PLTs. Our results show that CD11b expression was induced by soluble factors of PLTs but direct contact between cell populations was needed to enhance the respiratory burst. Additionally, B. abortus-infected PLTs recruit polymorphonuclear (PMN) cells to the site of infection. Finally, the presence of PLTs did not modify the initial invasion of PMN cells by B. abortus but improved the control of the infection at extended times. Altogether, our results demonstrate that PLTs interact with neutrophils and promote a proinflammatory phenotype which could also contribute to the resolution of the infection.
Collapse
Affiliation(s)
- Aldana Trotta
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - M Ayelén Milillo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Agustina Serafino
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Luis A Castillo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Federico Birnberg Weiss
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - M Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Buenos Aires, Argentina
| | | | - Gabriela C Fernández
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| |
Collapse
|
30
|
Vieira ML, Nascimento ALTO. Virulent Leptospira interrogans Induce Cytotoxic Effects in Human Platelets in vitro Through Direct Interactions. Front Microbiol 2020; 11:572972. [PMID: 33117318 PMCID: PMC7552899 DOI: 10.3389/fmicb.2020.572972] [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: 06/15/2020] [Accepted: 09/07/2020] [Indexed: 12/27/2022] Open
Abstract
Leptospirosis is a prevalent zoonotic disease, caused by bacteria of the genus Leptospira. Leptospirosis frequently leads to hemostatic disturbances, and the severe cases are marked by hemorrhages and low platelet number in circulation, which is associated with the patients’ poor outcomes. Nevertheless, Leptospira-platelet interactions remain poorly explored. In this study, we performed a series of in vitro experiments evaluating whether leptospires induce human platelet aggregation, activation, and morphological changes. Platelets were incubated with virulent L. interrogans and the platelet outcomes were assessed by aggregometry, flow cytometry, and scanning and transmission electron microscopy. Our results show that leptospires alone do not induce platelet aggregation and activation, and induce platelet cytotoxic effects instead, by clearly inducing platelet disruption and detachment. We show for the first time that virulent leptospires do interact directly with platelets, an event that could trigger pathophysiological effects during the infection. This study might serve as a basis for the development of novel treatments for the disease.
Collapse
Affiliation(s)
- Monica Larucci Vieira
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Laboratório de Desenvolvimento de Vacinas, Butantan Institute, São Paulo, Brazil
| | | |
Collapse
|
31
|
Karabulut B, Alatas SO. Diagnostic Value of Neutrophil to Lymphocyte Ratio and Mean Platelet Volume on Early Onset Neonatal Sepsis on Term Neonate. J Pediatr Intensive Care 2020; 10:143-147. [PMID: 33884215 DOI: 10.1055/s-0040-1715104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/21/2020] [Indexed: 12/29/2022] Open
Abstract
By setting out from increased neutrophil count, decreased lymphocyte count, and increased mean platelet volume (MPV), which is a result of the effect of inflammation on blood cells, we aimed to investigate whether neutrophil to lymphocyte ratio (NLP) and MPV can be used as an auxiliary parameter for the diagnosis of early-onset neonatal sepsis (EOS). This study was conducted by analyzing term neonates with EOS and physiological jaundice who were admitted to the neonatal intensive care unit of Izmir Katip Celebi University Ataturk Training and Research Hospital. A total of 63 neonate files were examined to include 30 term neonates with EOS, and 77 neonate files were examined to include 30 term neonates with physiological jaundice as a control group. NLR had an area under the curve (AUC) of 0.891 for prediction of EOS. At a cut-off level of 1.42, NLR had a likelihood ratio (LR) of 5.5, sensitivity of 88%, a specificity of 84%, a positive predictive value (PPV) of 84.6%, and a negative predictive value (NPV) of 87.5%. MPV had an AUC of 0.666 for the prediction of EOS and at a cut-off level of 9.3 fL, MPV had an LR of 1.23, sensitivity of 84%, a specificity of 32%, a PPV of 55.2%, and an NPV of 66.6%. In conclusion, this study provides evidence that NLR and MPV can be used in addition to conventional parameters in the diagnosis of EOS.
Collapse
Affiliation(s)
- Birol Karabulut
- Department of Pediatrics, Division of Neonatology, Izmir Katip Celebi University Ataturk Training and Research Hospital, Izmir, Karsıyaka, Turkey
| | - Silem Ozdem Alatas
- Department of Pediatrics, Division of Neonatology, Izmir Katip Celebi University Ataturk Training and Research Hospital, Izmir, Karsıyaka, Turkey
| |
Collapse
|
32
|
Yeoh CB, Lee KJ, Rieth EF, Mapes R, Tchoudovskaia AV, Fischer GW, Tollinche LE. COVID-19 in the Cancer Patient. Anesth Analg 2020; 131:16-23. [PMID: 32543802 PMCID: PMC7179051 DOI: 10.1213/ane.0000000000004884] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2020] [Indexed: 12/15/2022]
Abstract
The novel coronavirus disease 2019 (COVID-19) was first reported in China in December 2019. Since then, it has spread across the world to become one of the most serious life-threatening pandemics since the influenza pandemic of 1918. This review article will focus on the specific risks and nuanced considerations of COVID-19 in the cancer patient. Important perioperative management recommendations during this outbreak are emphasized, in addition to discussion of current treatment techniques and strategies available in the battle against COVID-19.
Collapse
Affiliation(s)
- Cindy B. Yeoh
- From the Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathleen J. Lee
- Department of Anesthesiology and Pain Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Elizabeth F. Rieth
- From the Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Renee Mapes
- Department of Anesthesiology and Pain Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Anna V. Tchoudovskaia
- From the Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gregory W. Fischer
- From the Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luis E. Tollinche
- From the Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
33
|
Jeyaraman M, Ranjan R, Kumar R, Arora A, Chaudhary D, Ajay SS, Jain R. Cellular Therapy: Shafts of Light Emerging for COVID-19. Stem Cell Investig 2020; 7:11. [PMID: 32695804 PMCID: PMC7367471 DOI: 10.21037/sci-2020-022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
The COVID-19 pandemic has presented with debilitating respiratory consequences especially more pronounced in high risk individuals. Individuals with underlying systemic diseases are more prone and vulnerable to suffer severe consequences of SARS-CoV-2 infectivity. The pathophysiological changes identified cytokine storm mechanism for out setting the series of adverse clinical conditions. Thereby, associating it with high mortality rates. This warrants urgent consideration of divergent modalities such as the cellular therapy. Cellular therapy (CT) is a new medical paradigm wherein cellular material is administered to patients for therapeutic purposes. In this regard, mesenchymal stem cells (MSCs) have yielded the most promising results among stromal vascular fraction (SVF); placental cells; natural killer (NK) cell and platelet lysate respectively. Following the administration of the CT as per preferred route, these play pivotal role in modifying the microenvironment of the lung tissue with their distinct sets of mechanism. Evidences have shown how their immunomodulatory action repairs and prevents lung injury which in turn improvise the compliance of lungs. In this review article we have discussed these emerging novel approaches and their target step serving as a ray of hope to combat severe form of COVID-19. Currently these aren't approved for preventing or treating COVID-19 cases, however clinical trials are afoot to dispense the utmost understanding in terms of efficacy and safety concerns.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rakesh Kumar
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Arunabh Arora
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Dushyant Chaudhary
- Department of Orthopaedics, School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | | | - Rashmi Jain
- School of Medical Sciences & Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
34
|
Rambach G, Fleischer V, Harpf V, Lackner M, Meinitzer A, Maier H, Engesser J, Lass-Flörl C, Speth C. Comparative immunopathogenesis in a murine model of inhalative infection with the mucormycetes Lichtheimia corymbifera and Rhizopus arrhizus. PLoS One 2020; 15:e0234063. [PMID: 32555589 PMCID: PMC7299637 DOI: 10.1371/journal.pone.0234063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/18/2020] [Indexed: 12/27/2022] Open
Abstract
Pathogenic mucormycetes induce diseases with considerable morbidity and mortality in immunocompromised patients. Virulence data comparing different Mucorales species and various underlying risk factors are limited. We therefore compared the pathogenesis of inhalative infection by Rhizopus (R.) arrhizus and Lichtheimia (L.) corymbifera in murine models for predominant risk factors for onset of infection. Mice with diabetes or treated with cyclophosphamide or cortisone acetate were challenged via the intranasal route with an isolate of R. arrhizus or L. corymbifera, respectively. Clinical, immunological and inflammation parameters as well as efficacy of posaconazole prophylaxis were monitored over 14 days. Whereas immunocompetent mice showed no clinical symptoms after mucormycete infection, mice treated with either cyclophosphamide (CP) or cortisone acetate (CA) were highly susceptible. Animals infected with the isolate of R. arrhizus showed prolonged survival and lower mortality, compared to those exposed to the L. corymbifera isolate. This lower virulence of R. arrhizus was risk factor-dependent, since diabetic mice died only after infection with Rhizopus, whereas all Lichtheimia-infected diabetic animals survived. Under posaconazole prophylaxis, both mucormycetes were able to establish breakthrough infections in CA- and CP-treated mice, but the course of infection was significantly delayed. Detailed analysis revealed that susceptibility of CA- and CP-treated mice could not be mimicked by exclusive lack or downmodulation of neutrophils, platelets or complement, but can be supposed to be the consequence of a broad immunosuppressive effect induced by the drugs. Both Lichtheimia corymbifera and Rhizopus arrhizus induce invasive mycoses in immunocompromised hosts after inhalative infection. Key parameters such as virulence and immunopathogenesis vary strongly according to fungal species and underlying risk group. Selected neutropenia is no sufficient risk factor for onset of inhalative mucormycosis.
Collapse
Affiliation(s)
- Günter Rambach
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Verena Fleischer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Verena Harpf
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hans Maier
- INNPATH GmbH–Institute of Pathology, Innsbruck, Austria
| | - Johannes Engesser
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Cornelia Speth
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
- * E-mail:
| |
Collapse
|
35
|
Page MJ, Pretorius E. A Champion of Host Defense: A Generic Large-Scale Cause for Platelet Dysfunction and Depletion in Infection. Semin Thromb Hemost 2020; 46:302-319. [PMID: 32279287 PMCID: PMC7339151 DOI: 10.1055/s-0040-1708827] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thrombocytopenia is commonly associated with sepsis and infections, which in turn are characterized by a profound immune reaction to the invading pathogen. Platelets are one of the cellular entities that exert considerable immune, antibacterial, and antiviral actions, and are therefore active participants in the host response. Platelets are sensitive to surrounding inflammatory stimuli and contribute to the immune response by multiple mechanisms, including endowing the endothelium with a proinflammatory phenotype, enhancing and amplifying leukocyte recruitment and inflammation, promoting the effector functions of immune cells, and ensuring an optimal adaptive immune response. During infection, pathogens and their products influence the platelet response and can even be toxic. However, platelets are able to sense and engage bacteria and viruses to assist in their removal and destruction. Platelets greatly contribute to host defense by multiple mechanisms, including forming immune complexes and aggregates, shedding their granular content, and internalizing pathogens and subsequently being marked for removal. These processes, and the nature of platelet function in general, cause the platelet to be irreversibly consumed in the execution of its duty. An exaggerated systemic inflammatory response to infection can drive platelet dysfunction, where platelets are inappropriately activated and face immunological destruction. While thrombocytopenia may arise by condition-specific mechanisms that cause an imbalance between platelet production and removal, this review evaluates a generic large-scale mechanism for platelet depletion as a repercussion of its involvement at the nexus of responses to infection.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
36
|
Deshmukh H, Rambach G, Sheppard DC, Lee M, Hagleitner M, Hermann M, Würzner R, Lass-Flörl C, Speth C. Galactosaminogalactan secreted from Aspergillus fumigatus and Aspergillus flavus induces platelet activation. Microbes Infect 2020; 22:331-339. [PMID: 31962135 DOI: 10.1016/j.micinf.2019.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 01/09/2023]
Abstract
Platelets are meanwhile recognized as versatile elements within the immune system and appear to play a key role in the innate immune response to pathogens including fungi. Previous experiments revealed platelet activation by direct contact with the hyphal-associated polysaccharide galactosaminogalactan (GAG). Since secreted fungal products may also be relevant and trigger immune reactions or thrombosis, we screened culture supernatants (SN) of human-pathogenic fungi for their capacity to activate platelets. For that purpose, platelets were incubated with SN from various fungal species; platelet activation and GAG deposition on the surface of platelets were detected by flow cytometry and electron and confocal microscopy, Culture supernatants of Aspergillus fumigatus and flavus isolates were potent platelet stimulators in a dose- and time-dependent manner, while SN of other Aspergillus species and all tested mucormycete species did not significantly induce platelet activation. The capacity of culture SN to activate platelets was dependent on fungal production of GAG and deposition of secreted GAG on the platelet surface; supernatants from mucormycetes or mutants of A. fumigatus lacking GAG secretion did not affect platelet activity. These results suggest that invading fungi can stimulate platelets not only locally through direct interactions with fungal hyphae, but can also act over a certain distance through secreted GAG.
Collapse
Affiliation(s)
- Hemalata Deshmukh
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Donald C Sheppard
- Department of Microbiology and Immunology, McGill University, Montréal, H3A 0G4, Canada
| | - Mark Lee
- Department of Microbiology and Immunology, McGill University, Montréal, H3A 0G4, Canada
| | - Magdalena Hagleitner
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, 6020, Innsbruck, Austria
| | - Reinhard Würzner
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
37
|
Platelet function in patients with septic shock. Thromb Res 2020; 185:33-42. [DOI: 10.1016/j.thromres.2019.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/14/2019] [Accepted: 11/11/2019] [Indexed: 12/11/2022]
|
38
|
Kerris EWJ, Hoptay C, Calderon T, Freishtat RJ. Platelets and platelet extracellular vesicles in hemostasis and sepsis. J Investig Med 2019; 68:813-820. [PMID: 31843956 DOI: 10.1136/jim-2019-001195] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2019] [Indexed: 01/09/2023]
Abstract
Platelets, cell fragments traditionally thought of as important only for hemostasis, substantially and dynamically contribute to the immune system's response to infection. In addition, there is increasing evidence that externally active platelet entities, including platelet granules and platelet extracellular vesicles (PEVs), play a role not only in hemostasis, but also in inflammatory actions previously ascribed to platelets themselves. Given the functions of platelets and PEVs during inflammation and infection, their role in sepsis is being investigated. Sepsis is a condition marked by the dysregulation of the body's normal activation of the immune system in response to a pathogen. The mechanisms for controlling infection locally become detrimental to the host if they are applied systemically. Similar to cells traditionally ascribed to the immune system, including neutrophils, lymphocytes, and macrophages, platelets are instrumental in helping a host clear an infection, but are also implicated in the uncontrolled amplification of the immune response that leads to sepsis. Clearly, the function of platelets is more complicated than its simple structure and primary role in hemostasis initially suggest. This review provides an overview of platelet and platelet extracellular vesicle structure and function, highlighting the complex role platelets and PEVs play in the body in the context of infection and sepsis.
Collapse
Affiliation(s)
- Elizabeth W J Kerris
- Division of Critical Care Medicine, Children's National Hospital, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Claire Hoptay
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Thais Calderon
- Department of Medical Education, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Robert J Freishtat
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA.,Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
| |
Collapse
|
39
|
Lee JK, Mereuta L, Luchian T, Park Y. Antimicrobial peptide HPA3NT3-A2 effectively inhibits biofilm formation in mice infected with drug-resistant bacteria. Biomater Sci 2019; 7:5068-5083. [PMID: 31555777 DOI: 10.1039/c9bm01051c] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial biofilms formed through secretion of extracellular polymeric substances (EPS) have been implicated in many serious infections and can increase antibiotic resistance by a factor of more than 1000. Here, we examined the abilities of the antimicrobial peptide HPA3NT3-A2 to inhibit and reduce biofilm formation, eliminate EPS, and suppress inflammation in mice infected with clinical isolates of drug-resistant Pseudomonas aeruginosa strains. HPA3NT3-A2 was developed from a desirable analogue peptide, HPA3NT3, derived from residues 2-20 of the Helicobacter pylori ribosomal protein L1. HPA3NT3-A2 showed stronger activity against planktonic cells (MIC: 8 μM) compared to ciprofloxacin or tobramycin (>512 μM), and a favorable minimum biofilm inhibition and elimination concentration. This peptide also neutralized LPS; decreased levels of EPS; inhibited the production of pro-inflammatory cytokines in the lung, kidney, and spleen; decreased white blood cell counts; and increased survival among infected mice.
Collapse
Affiliation(s)
- Jong-Kook Lee
- Research Center for Proteinaceous Materials (RCPM), Chosun University, Gwangju 501-759, Korea
| | | | | | | |
Collapse
|
40
|
Padmakumar M, Van Raes E, Van Geet C, Freson K. Blood platelet research in autism spectrum disorders: In search of biomarkers. Res Pract Thromb Haemost 2019; 3:566-577. [PMID: 31624776 PMCID: PMC6781926 DOI: 10.1002/rth2.12239] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a clinically heterogeneous neurodevelopmental disorder that is caused by gene-environment interactions. To improve its diagnosis and treatment, numerous efforts have been undertaken to identify reliable biomarkers for autism. None of them have delivered the holy grail that represents a reproducible, quantifiable, and sensitive biomarker. Though blood platelets are mainly known to prevent bleeding, they also play pivotal roles in cancer, inflammation, and neurological disorders. Platelets could serve as a peripheral biomarker or cellular model for autism as they share common biological and molecular characteristics with neurons. In particular, platelet-dense granules contain neurotransmitters such as serotonin and gamma-aminobutyric acid. Molecular players controlling granule formation and secretion are similarly regulated in platelets and neurons. The major platelet integrin receptor αIIbβ3 has recently been linked to ASD as a regulator of serotonin transport. Though many studies revealed associations between platelet markers and ASD, there is an important knowledge gap in linking these markers with autism and explaining the altered platelet phenotypes detected in autism patients. The present review enumerates studies of different biomarkers detected in ASD using platelets and highlights the future needs to bring this research to the next level and advance our understanding of this complex disorder.
Collapse
Affiliation(s)
- Manisha Padmakumar
- Department of Cardiovascular SciencesCenter for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Eveline Van Raes
- Department of Cardiovascular SciencesCenter for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Chris Van Geet
- Department of Cardiovascular SciencesCenter for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Kathleen Freson
- Department of Cardiovascular SciencesCenter for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| |
Collapse
|
41
|
Chen H, Lin C, Fan Z, Yu W, Cao M, Ke C, Jiao X. Serum cytokines and clinical features in patients with fever and thrombocytopenia syndrome. Clin Chim Acta 2019; 494:22-30. [PMID: 30831067 PMCID: PMC7124278 DOI: 10.1016/j.cca.2019.02.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 01/11/2019] [Accepted: 02/28/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE To explore the clinical, microbiological and immunological features of patients with fever and thrombocytopenia. METHODS Patients with unexplained fever and thrombocytopenia were enrolled. Viruses were detected using real-time PCR, and bacteria were measured by culturing methods. Serum cytokines, platelet antibody IgG (PA-IgG) and Helicobacter pylori (HP) were detected using ELISA. RESULTS Pathogens were detected in 74.68% of patients, which included single fungal/viral/bacterial infection and multiple infection. The pathogens could not be unidentified in 25.32% of cases. Cytokines including Interleukin (IL)-6, IL-10, interferon-γ(IFN-γ), platelet activating factor (PAF) and PA-IgG were significantly higher in patients as compared to healthy controls (P < .01 or P < .05). Principal component analyses extracted four groups of parameters that have a strong positive predicting value, revealing that disease status evaluation would be more accurate if we combined the platelet parameters and inflammatory biomarkers. While event-free survival (EFS) that indicates the time of platelet elevated after therapy was the highest in patients with single bacterial or fungal infection, EFS was affected by the levels of cytokines and PA-IgG. CONCLUSIONS Differences in immune function may be the main factors affecting the prognosis of patients with fever and thrombocytopenia, while treatment based on precise etiological diagnosis is important for therapeutic efficacy.
Collapse
Affiliation(s)
- Huanzhu Chen
- Cell Biology, Genetics Department, Shantou University Medical College, Shantou 515041, China; Department of Biochemistry, Medical College of Jiaying University, Meizhou 514031, China
| | - Chun Lin
- First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zhiqiang Fan
- Cell Biology, Genetics Department, Shantou University Medical College, Shantou 515041, China
| | - Wenjun Yu
- Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Manxiong Cao
- First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Changwen Ke
- Cell Biology, Genetics Department, Shantou University Medical College, Shantou 515041, China
| | - Xiaoyang Jiao
- Cell Biology, Genetics Department, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
42
|
Salamah MF, Ravishankar D, Vaiyapuri R, Moraes LA, Patel K, Perretti M, Gibbins JM, Vaiyapuri S. The formyl peptide fMLF primes platelet activation and augments thrombus formation. J Thromb Haemost 2019; 17:1120-1133. [PMID: 31033193 PMCID: PMC6617722 DOI: 10.1111/jth.14466] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 04/18/2019] [Indexed: 01/07/2023]
Abstract
Essentials The role of formyl peptide receptor 1 (FPR1) and its ligand, fMLF, in the regulation of platelet function, hemostasis, and thrombosis is largely unknown. Fpr1-deficient mice and selective inhibitors for FPR1 were used to investigate the function of fMLF and FPR1 in platelets. N-formyl-methionyl-leucyl-phenylalanine primes platelet activation and augments thrombus formation, mainly through FPR1 in platelets. Formyl peptide receptor 1 plays a pivotal role in the regulation of platelet function. BACKGROUND Formyl peptide receptors (FPRs) play pivotal roles in the regulation of innate immunity and host defense. The FPRs include three family members: FPR1, FPR2/ALX, and FPR3. The activation of FPR1 by its high-affinity ligand, N-formyl-methionyl-leucyl-phenylalanine (fMLF) (a bacterial chemoattractant peptide), triggers intracellular signaling in immune cells such as neutrophils and exacerbates inflammatory responses to accelerate the clearance of microbial infection. Notably, fMLF has been demonstrated to induce intracellular calcium mobilization and chemotaxis in platelets that are known to play significant roles in the regulation of innate immunity and inflammatory responses. Despite a plethora of research focused on the roles of FPR1 and its ligands such as fMLF on the modulation of immune responses, their impact on the regulation of hemostasis and thrombosis remains unexplored. OBJECTIVE To determine the effects of fMLF on the modulation of platelet reactivity, hemostasis, and thrombus formation. METHODS Selective inhibitors for FPR1 and Fpr1-deficient mice were used to determine the effects of fMLF and FPR1 on platelets using various platelet functional assays. RESULTS N-formyl-methionyl-leucyl-phenylalanine primes platelet activation through inducing distinctive functions and enhances thrombus formation under arterial flow conditions. Moreover, FPR1 regulates normal platelet function as its deficiency in mouse or blockade in human platelets using a pharmacological inhibitor resulted in diminished agonist-induced platelet activation. CONCLUSION Since FPR1 plays critical roles in numerous disease conditions, its influence on the modulation of platelet activation and thrombus formation may provide insights into the mechanisms that control platelet-mediated complications under diverse pathological settings.
Collapse
Affiliation(s)
| | | | | | | | - Ketan Patel
- School of Biological SciencesUniversity of ReadingReadingUK
| | - Mauro Perretti
- William Harvey Research Institute, Queen Mary University of LondonLondonUK
| | | | | |
Collapse
|
43
|
Speth C, Rambach G, Lass-Flörl C, Howell PL, Sheppard DC. Galactosaminogalactan (GAG) and its multiple roles in Aspergillus pathogenesis. Virulence 2019; 10:976-983. [PMID: 30667338 PMCID: PMC8647848 DOI: 10.1080/21505594.2019.1568174] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aspergillus spp and particularly the species Aspergillus fumigatus are the causative agents of invasive aspergillosis, a progressive necrotizing pneumonia that occurs in immunocompromised patients. The limited efficacy of currently available antifungals has led to interest in a better understanding of the molecular mechanisms underlying the pathogenesis of invasive aspergillosis in order to identify new therapeutic targets for this devastating disease. The Aspergillus exopolysaccharide galactosaminogalactan (GAG) plays an important role in the pathogenesis of experimental invasive aspergillosis. The present review article summarizes our current understanding of GAG composition and synthesis and the molecular mechanisms whereby GAG promotes virulence. Promising directions for future research and the prospect of GAG as both a therapy and therapeutic target are reviewed.
Collapse
Affiliation(s)
- Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Donald C Sheppard
- Departments of Medicine and of Microbiology and Immunology, McGill University, Montréal, Canada.,Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Canada
| |
Collapse
|
44
|
Minasyan H, Flachsbart F. Blood coagulation: a powerful bactericidal mechanism of human innate immunity. Int Rev Immunol 2019; 38:3-17. [DOI: 10.1080/08830185.2018.1533009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hayk Minasyan
- Private laboratory, Immunology Microbiology, Yerevan, Armenia
| | | |
Collapse
|
45
|
Rahman A, Tiwari A, Narula J, Hickling T. Importance of Feedback and Feedforward Loops to Adaptive Immune Response Modeling. CPT Pharmacometrics Syst Pharmacol 2018; 7:621-628. [PMID: 30198637 PMCID: PMC6202469 DOI: 10.1002/psp4.12352] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/15/2018] [Indexed: 12/15/2022] Open
Abstract
The human adaptive immune system is a very complex network of different types of cells, cytokines, and signaling molecules. This complex network makes it difficult to understand the system level regulations. To properly explain the immune system, it is necessary to explicitly investigate the presence of different feedback and feedforward loops (FFLs) and their crosstalks. Considering that these loops increase the complexity of the system, the mathematical modeling has been proved to be an important tool to explain such complex biological systems. This review focuses on these regulatory loops and discusses their importance on systems modeling of the immune system.
Collapse
|
46
|
Abstract
Neonatal sepsis (NS) continues to be a diagnostic challenge and a prime cause of mortality. Forage for a lucid, cost-effective yet highly sensitive and specific marker in diagnosing this entity is an incessant task. This study aimed to evaluate the predictive value of mean platelet volume (MPV) in diagnosing NS. Neonates diagnosed with sepsis from January 2016 to March 2016 were included in the study. The subjects were stratified into the following: (i) culture-proven sepsis (group I); (ii) culture-negative clinical sepsis (group II); and (iii) control group (group III). Several hematologic markers such as hemoglobin, total leukocyte count, platelet count, MPV, plateletcrit, platelet distribution width, immature-to-mature neutrophil ratio, toxic change, serum urea, bilirubin, and C-reactive protein were analyzed. The results were compared among the groups, and their efficacy in diagnosing NS was appraised. The study involved 210 neonates, of which, groups I, II, and III constituted 64, 75, and 71 cases, respectively. The mean MPV among groups I, II, and III was 9.56, 8.86, and 8.58 fL, respectively (P<0.05). Strikingly higher values of platelet count, immature-to-mature neutrophil ratio, MPV, plateletcrit, and C-reactive protein were found in group I in contrast to those in groups II and III (P<0.05). The baseline MPV of patients with culture-proven sepsis was comparatively higher than controls and was found to be statistically significant. Hence, MPV can be a simple, economical, and specific predictor of NS.
Collapse
|
47
|
Eberl C, Speth C, Jacobsen ID, Hermann M, Hagleitner M, Deshmukh H, Ammann CG, Lass-Flörl C, Rambach G. Candida: Platelet Interaction and Platelet Activity in vitro. J Innate Immun 2018; 11:52-62. [PMID: 30176656 PMCID: PMC6738150 DOI: 10.1159/000491030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
Over the last 2 decades, platelets have been recognized as versatile players of innate immunity. The interaction of platelets with fungal pathogens and subsequent processes may critically influence the clinical outcome of invasive mycoses. Since the role of platelets in Candida infections is poorly characterized and controversially discussed, we studied interactions of human platelets with yeast cells, (pseudo-)hyphae, biofilms and secretory products of human pathogenic Candida species applying platelet rich plasma and a whole blood model. Incubation of Candida with platelets resulted in moderate mutual interaction with some variation between different species. The rate of platelets binding to -Candida (pseudo-) hyphae and candidal biofilm was comparably low as that to the yeast form. Candida-derived secretory products did not affect platelet activity - neither stimulatory nor inhibitory. The small subset of platelets that bound to Candida morphotypes was consequently activated. However, this did not result in reduced growth or viability of the different Candida species. A whole blood model simulating in vivo conditions confirmed platelet activation in the subpopulation of Candida-bound platelets. Thus, the inability of platelets to efficiently react on Candida presence might favor fungal survival in the blood and contribute to high morbidity of Candida sepsis.
Collapse
Affiliation(s)
- Claudia Eberl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck,
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
- Institute for Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Hagleitner
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hemalata Deshmukh
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph G Ammann
- Experimental Orthopaedics, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
48
|
Petrea C, Crăiţoiu Ş, Vrapciu A, Mănoiu V, Rusu M. The telopode- and filopode-projecting heterogeneous stromal cells of the human sclera niche. Ann Anat 2018; 218:129-140. [DOI: 10.1016/j.aanat.2017.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/13/2017] [Accepted: 12/15/2017] [Indexed: 12/23/2022]
|
49
|
Low platelet count as risk factor for infections in patients with primary immune thrombocytopenia: a retrospective evaluation. Ann Hematol 2018; 97:1701-1706. [PMID: 29777278 PMCID: PMC6097778 DOI: 10.1007/s00277-018-3367-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 05/07/2018] [Indexed: 12/13/2022]
Abstract
Infectious complications are common and sometimes life threatening in patients with immune thrombocytopenia (ITP), mainly due to the immune-suppressive therapy. Recent evidence suggests a potential role of platelets in the inflammation process. In this clinical study, we further investigated the role of thrombocytopenia on infections in patients with primary ITP. We retrospectively evaluated data from the recently published large randomized clinical trial of a cohort of 195 patients with primary ITP, who were randomized for prednisone or high-dose dexamethasone. From 158 patients (81%), data on platelet count and infections within the first month of treatment were collected. In this period, 24% of the ITP patients had an infection. Patients with infection had significant lower platelet counts during the first month of treatment leading to a significant lower therapy response at 1 month and a significant longer hospital stay (14.0 versus 9.8 days). Additionally, Cox regression analysis showed that an increase in platelet count of 20 × 109/L led to a reduction of 52% in infections in the next week, showing low platelet count is a significant risk factor for infection. Platelet transfusion led to an increase in platelet count in ITP patients without infection, but not in patients with infection. In conclusion, infections are common in patients with primary ITP leading to significant worse response rates and a longer hospital stay. Interestingly, low platelet count was independently correlated with an increased risk of infection.
Collapse
|
50
|
Trotta A, Velásquez LN, Milillo MA, Delpino MV, Rodríguez AM, Landoni VI, Giambartolomei GH, Pozner RG, Barrionuevo P. Platelets Promote Brucella abortus Monocyte Invasion by Establishing Complexes With Monocytes. Front Immunol 2018; 9:1000. [PMID: 29867977 PMCID: PMC5949576 DOI: 10.3389/fimmu.2018.01000] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/23/2018] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is an infectious disease elicited by bacteria of the genus Brucella. Platelets have been extensively described as mediators of hemostasis and responsible for maintaining vascular integrity. Nevertheless, they have been recently involved in the modulation of innate and adaptive immune responses. Although many interactions have been described between Brucella abortus and monocytes/macrophages, the role of platelets during monocyte/macrophage infection by these bacteria remained unknown. The aim of this study was to investigate the role of platelets in the immune response against B. abortus. We first focused on the possible interactions between B. abortus and platelets. Bacteria were able to directly interact with platelets. Moreover, this interaction triggered platelet activation, measured as fibrinogen binding and P-selectin expression. We further investigated whether platelets were involved in Brucella-mediated monocyte/macrophage early infection. The presence of platelets promoted the invasion of monocytes/macrophages by B. abortus. Moreover, platelets established complexes with infected monocytes/macrophages as a result of a carrier function elicited by platelets. We also evaluated the ability of platelets to modulate functional aspects of monocytes in the context of the infection. The presence of platelets during monocyte infection enhanced IL-1β, TNF-α, IL-8, and MCP-1 secretion while it inhibited the secretion of IL-10. At the same time, platelets increased the expression of CD54 (ICAM-1) and CD40. Furthermore, we showed that soluble factors released by B. abortus-activated platelets, such as soluble CD40L, platelet factor 4, platelet-activating factor, and thromboxane A2, were involved in CD54 induction. Overall, our results indicate that platelets can directly sense and react to B. abortus presence and modulate B. abortus-mediated infection of monocytes/macrophages increasing their pro-inflammatory capacity, which could promote the resolution of the infection.
Collapse
Affiliation(s)
- Aldana Trotta
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Lis N. Velásquez
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - M. Ayelén Milillo
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - M. Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana M. Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica I. Landoni
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Guillermo H. Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Roberto G. Pozner
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|