1
|
Dong T, Zhang C, Wu Z, Shuai L, Fu N, Zhang Y, Zhang L, Xiong X. A biomimetic nanomedicine alleviates liver transplant-related biliary injury by sequentially inhibiting oxidative stress and regulating macrophage polarization via Nrf-2/HO-1 and JNK pathways. Mater Today Bio 2025; 32:101797. [PMID: 40343167 PMCID: PMC12059350 DOI: 10.1016/j.mtbio.2025.101797] [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: 03/02/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Liver transplantation is an effective method for treating end-stage liver disease. However, 10-20 % of liver transplantation patients develop biliary injury, the main cause of which is ischemia-reperfusion injury (IRI), which consists of oxidative stress injury in the early stage and inflammatory injury in the advanced stage. Biliary injury seriously affects patient outcomes and even leads to mortality, and there are few effective treatments for IRI. Herein, nanoparticles containing quercetin (QR) and rapamycin (RP) coated with poly (lactic-co-glycolic acid) (PLGA) and encapsulated by platelet membrane (PM) were designed to treat IRI in the liver transplantation. The specific binding of ICAM-1 expressed on the PM to integrins (e.g., LFA-1 and Mac-1) in damaged vascular endothelial cells, as well as the interaction between P-selectin on the platelet surface and PSGL-1 on the macrophage surface, allows the accumulation of these biomimetic cell membrane-encapsulated nanoparticles, and subsequently, the delivery of both drugs, to ischemia-reperfusion sites in the liver. The encapsulated QR alleviated oxidative stress injury by activating the Nrf-2/HO-1 signaling pathway in the early stage in model rats with IRI and liver transplantation models. Moreover, RP alleviated inflammatory damage in the advanced stage by suppressing the JNK signaling pathway in M1 macrophages. Thus, these biomimetic nanoparticles that intervene in IRI to alleviate both the early oxidative stress and the advanced inflammatory response constitute a novel delivery system for managing biliary injury after liver transplantation.
Collapse
Affiliation(s)
| | | | - Zhaoyi Wu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ling Shuai
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Nengsheng Fu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujun Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Leida Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiang Xiong
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
2
|
Tian Y, Zong Y, Pang Y, Zheng Z, Ma Y, Zhang C, Gao J. Platelets and diseases: signal transduction and advances in targeted therapy. Signal Transduct Target Ther 2025; 10:159. [PMID: 40374650 DOI: 10.1038/s41392-025-02198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 05/17/2025] Open
Abstract
Platelets are essential anucleate blood cells that play pivotal roles in hemostasis, tissue repair, and immune modulation. Originating from megakaryocytes in the bone marrow, platelets are small in size but possess a highly specialized structure that enables them to execute a wide range of physiological functions. The platelet cytoplasm is enriched with functional proteins, organelles, and granules that facilitate their activation and participation in tissue repair processes. Platelet membranes are densely populated with a variety of receptors, which, upon activation, initiate complex intracellular signaling cascades. These signaling pathways govern platelet activation, aggregation, and the release of bioactive molecules, including growth factors, cytokines, and chemokines. Through these mechanisms, platelets are integral to critical physiological processes such as thrombosis, wound healing, and immune surveillance. However, dysregulated platelet function can contribute to pathological conditions, including cancer metastasis, atherosclerosis, and chronic inflammation. Due to their central involvement in both normal physiology and disease, platelets have become prominent targets for therapeutic intervention. Current treatments primarily aim to modulate platelet signaling to prevent thrombosis in cardiovascular diseases or to reduce excessive platelet aggregation in other pathological conditions. Antiplatelet therapies are widely employed in clinical practice to mitigate clot formation in high-risk patients. As platelet biology continues to evolve, emerging therapeutic strategies focus on refining platelet modulation to enhance clinical outcomes and prevent complications associated with platelet dysfunction. This review explores the structure, signaling pathways, biological functions, and therapeutic potential of platelets, highlighting their roles in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Yuchen Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
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
|
4
|
Zhang Y, Jia C, Guo M, Chen Q, Wen Y, Wang T, Xie Y, Fan X, Gao J, Yarovinsky TO, Liu R, Jiang Z, Wang M, Zhou J, Che D, Fu L, Edelson R, Gu X, Hwa J, Tang WH. Platelet-Monocyte Aggregate Instigates Inflammation and Vasculopathy in Kawasaki Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406282. [PMID: 39665236 PMCID: PMC11792051 DOI: 10.1002/advs.202406282] [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: 06/07/2024] [Revised: 10/31/2024] [Indexed: 12/13/2024]
Abstract
Kawasaki disease (KD) is a severe acute febrile illness and systemic vasculitis that causes coronary artery aneurysms in young children. Platelet hyperreactivity and an aberrant immune response are key indicators of KD; however, the mechanism by which hyperactive platelets contribute to inflammation and vasculopathy in KD remains unclear. A cytokine-mediated positive feedback loop between KD platelets and monocytes is identified. KD platelet-monocyte aggregates (MPAs) are mediated by an initial interaction of P-selectin (cluster of differentiation 62P, CD62p) and its glycoprotein ligand 1 (PSGL-1). This is followed by a coordinated interaction of platelet glycoprotein (GP)Ibα with monocyte CD11b. Monocyte-activated platelets initiate transforming growth factor (TGF)β1 release, which results in nuclear localization of nuclear factor kappaB in monocytes, therefore, driving the phenotypic conversion of classical monocytes (CD14+CD16-) into proinflammatory monocytes (CD14+CD16+). The platelet-activated monocytes release interleukin-1 and tissue necrotic factor-α, which promote further platelet activation. KD-induced inflammation and vasculopathy are prevented by inhibiting the components of this positive feedback loop. Notably, mice deficient in platelet TGFβ1 show less MPA and CD14+CD16+ monocytes, along with reduced inflammation and vasculopathy. These findings reveal that platelet-monocyte interactive proteins (CD62p/PSGL-1 and (GP)Ibα/CD11b) and cytokine mediators (platelet TGFβ1) are potential biomarkers and therapeutic targets for KD vasculopathy.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Cuiping Jia
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Manli Guo
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Qian Chen
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Ying Wen
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Ting Wang
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Yinyin Xie
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Xuejiao Fan
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Jingwen Gao
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Timur O. Yarovinsky
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCT06511USA
| | - Renjing Liu
- Victor Chang Cardiac Research InstituteSydney2010Australia
| | - Zhiyong Jiang
- Department of Blood TransfusionGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Mengmeng Wang
- Department of Children's OphtalmologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Jin Zhou
- Department of Children's OphtalmologyGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Di Che
- Department of Biological Specimen BankGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Lanyan Fu
- Department of Biological Specimen BankGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Richard Edelson
- Department of DermatologySchool of MedicineYale UniversityNew HavenCT06511USA
| | - Xiaoqiong Gu
- Department of Biological Specimen BankGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - John Hwa
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCT06511USA
| | - Wai Ho Tang
- Institute of PediatricsGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
- School of Nursing and Health StudiesHong Kong Metropolitan UniversityKowloonHong Kong SARChina
| |
Collapse
|
5
|
Joyal M, Simard RD, Maharsy W, Prévost M, Nemer M, Guindon Y. Sialyl Lewis x Glycomimetics as E- and P-Selectin Antagonists Targeting Hyperinflammation. ACS Med Chem Lett 2025; 16:64-71. [PMID: 39811128 PMCID: PMC11726361 DOI: 10.1021/acsmedchemlett.4c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Inflammatory disorders, such as sepsis, pancreatitis, and severe COVID-19, often cause immune dysfunction and high mortality. These conditions trigger excessive immune cell influx, leading to cytokine storms, organ damage, and compensatory immune suppression that results in immunoparalysis, organ dysfunction, and reinfection. Controlled and reversible immunosuppression limiting immune cell recruitment to inflammation sites could reduce hyperinflammation and prevent immune exhaustion. PSGL-1 on leukocytes binds to vascular P- and E-selectins via its sialyl Lewisx pharmacophore, triggering key features of systemic inflammatory response syndrome and sepsis. We report the discovery of two immunomodulators, sialyl Lewisx glycomimetics (12 and 13), with a tetrazole carboxyl bioisostere of 3a, which binds P- and E-selectin and blocks their interaction with PSGL-1. In an in vivo hyperinflammation model, they reduced immune cell recruitment, evidenced by decreased neutrophils, CD11b+, monocytes/macrophages, and PSGL-1-positive cells at various time points. These glycomimetics may be promising leads for managing the systemic inflammatory response syndrome.
Collapse
Affiliation(s)
- Mathieu Joyal
- Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Ryan D. Simard
- Bioorganic
Chemistry Laboratory, Institut de recherches
cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
- Department
of Chemistry, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Wael Maharsy
- Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Michel Prévost
- Bioorganic
Chemistry Laboratory, Institut de recherches
cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Mona Nemer
- Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Yvan Guindon
- Bioorganic
Chemistry Laboratory, Institut de recherches
cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
- Department
of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
6
|
Yan M, Wang Z, Qiu Z, Cui Y, Xiang Q. Platelet signaling in immune landscape: comprehensive mechanism and clinical therapy. Biomark Res 2024; 12:164. [PMID: 39736771 DOI: 10.1186/s40364-024-00700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Platelets are essential for blood clotting and maintaining normal hemostasis. In pathological conditions, platelets are increasingly recognized as crucial regulatory factors in various immune-mediated inflammatory diseases. Resting platelets are induced by various factors such as immune complexes through Fc receptors, platelet-targeting autoantibodies and other platelet-activating stimuli. Platelet activation in immunological processes involves the release of immune activation stimuli, antigen presentation and interaction with immune cells. Platelets participate in both the innate immune system (neutrophils, monocytes/macrophages, dendritic cells (DCs) and Natural Killer (NK) cells and the adaptive immune system (T and B cells). Clinical therapeutic strategies include targeting platelet activation, platelet-immune cell interaction and platelet-endothelial cell interaction, which display positive development prospects. Understanding the mechanisms of platelets in immunity is important, and developing targeted modulations of these mechanisms will pave the way for promising therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhe Wang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhiwei Qiu
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
7
|
Nguyen BA, Alexander MR, Harrison DG. Immune mechanisms in the pathophysiology of hypertension. Nat Rev Nephrol 2024; 20:530-540. [PMID: 38658669 PMCID: PMC12060254 DOI: 10.1038/s41581-024-00838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Hypertension is a leading risk factor for morbidity and mortality worldwide. Despite current anti-hypertensive therapies, most individuals with hypertension fail to achieve adequate blood pressure control. Moreover, even with adequate control, a residual risk of cardiovascular events and associated organ damage remains. These findings suggest that current treatment modalities are not addressing a key element of the underlying pathology. Emerging evidence implicates immune cells as key mediators in the development and progression of hypertension. In this Review, we discuss our current understanding of the diverse roles of innate and adaptive immune cells in hypertension, highlighting key findings from human and rodent studies. We explore mechanisms by which these immune cells promote hypertensive pathophysiology, shedding light on their multifaceted involvement. In addition, we highlight advances in our understanding of autoimmunity, HIV and immune checkpoints that provide valuable insight into mechanisms of chronic and dysregulated inflammation in hypertension.
Collapse
Affiliation(s)
- Bianca A Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
8
|
Hawwari I, Rossnagel L, Rosero N, Maasewerd S, Vasconcelos MB, Jentzsch M, Demczuk A, Teichmann LL, Meffert L, Bertheloot D, Ribeiro LS, Kallabis S, Meissner F, Arditi M, Atici AE, Noval Rivas M, Franklin BS. Platelet transcription factors license the pro-inflammatory cytokine response of human monocytes. EMBO Mol Med 2024; 16:1901-1929. [PMID: 38977927 PMCID: PMC11319489 DOI: 10.1038/s44321-024-00093-3] [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: 11/14/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
In humans, blood Classical CD14+ monocytes contribute to host defense by secreting large amounts of pro-inflammatory cytokines. Their aberrant activity causes hyper-inflammation and life-threatening cytokine storms, while dysfunctional monocytes are associated with 'immunoparalysis', a state of immune hypo responsiveness and reduced pro-inflammatory gene expression, predisposing individuals to opportunistic infections. Understanding how monocyte functions are regulated is critical to prevent these harmful outcomes. We reveal platelets' vital role in the pro-inflammatory cytokine responses of human monocytes. Naturally low platelet counts in patients with immune thrombocytopenia or removal of platelets from healthy monocytes result in monocyte immunoparalysis, marked by impaired cytokine response to immune challenge and weakened host defense transcriptional programs. Remarkably, supplementing monocytes with fresh platelets reverses these conditions. We discovered that platelets serve as reservoirs of key cytokine transcription regulators, such as NF-κB and MAPK p38, and pinpointed the enrichment of platelet NF-κB2 in human monocytes by proteomics. Platelets proportionally restore impaired cytokine production in human monocytes lacking MAPK p38α, NF-κB p65, and NF-κB2. We uncovered a vesicle-mediated platelet-monocyte-propagation of inflammatory transcription regulators, positioning platelets as central checkpoints in monocyte inflammation.
Collapse
Affiliation(s)
- Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Lukas Rossnagel
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Marius Jentzsch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Agnieszka Demczuk
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lino L Teichmann
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Lisa Meffert
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lucas S Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sebastian Kallabis
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E Atici
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
9
|
Tuerhong N, Yang Y, Wang C, Huang P, Li Q. Interactions between platelets and the cancer immune microenvironment. Crit Rev Oncol Hematol 2024; 199:104380. [PMID: 38718939 DOI: 10.1016/j.critrevonc.2024.104380] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/16/2024] Open
Abstract
Cancer is a leading cause of death in both China and developed countries due to its high incidence and low cure rate. Immune function is closely linked to the development and progression of tumors. Platelets, which are primarily known for their role in hemostasis, also play a crucial part in the spread and progression of tumors through their interaction with the immune microenvironment. The impact of platelets on tumor growth and metastasis depends on the type of cancer and treatment method used. This article provides an overview of the relationship between platelets and the immune microenvironment, highlighting how platelets can either protect or harm the immune response and cancer immune escape. We also explore the potential of available platelet-targeting strategies for tumor immunotherapy, as well as the promise of new platelet-targeted tumor therapy methods through further research.
Collapse
Affiliation(s)
- Nuerye Tuerhong
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Yang Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Chenyu Wang
- The Second Clinical Medical College, Lanzhou university, No. 222 South Tianshui Road, Gansu, China
| | - Peng Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Augusto JF, Benden C, Diekmann F, Zuckermann A. The value of extracorporeal photopheresis as an immunosuppression-modifying approach in solid organ transplantation: a potential solution to an unmet medical need. Front Immunol 2024; 15:1371554. [PMID: 38846942 PMCID: PMC11154098 DOI: 10.3389/fimmu.2024.1371554] [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: 01/16/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Allograft rejection is a critical issue following solid organ transplantation (SOT). Immunosuppressive therapies are crucial in reducing risk of rejection yet are accompanied by several significant side effects, including infection, malignancy, cardiovascular diseases, and nephrotoxicity. There is a current unmet medical need with a lack of effective minimization strategies for these side effects. Extracorporeal photopheresis (ECP) has shown potential as an immunosuppression (IS)-modifying technique in several SOT types, with improvements seen in acute and recurrent rejection, allograft survival, and associated side effects, and could fulfil this unmet need. Through a review of the available literature detailing key areas in which ECP may benefit patients, this review highlights the IS-modifying potential of ECP in the four most common SOT procedures (heart, lung, kidney, and liver transplantation) and highlights existing gaps in data. Current evidence supports the use of ECP for IS modification following SOT, however there is a need for further high-quality research, in particular randomized control trials, in this area.
Collapse
Affiliation(s)
- Jean-François Augusto
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Angers, Angers, France
| | | | - Fritz Diekmann
- Renal Transplantation Unit, Department of Nephrology and Kidney Transplantation, Hospital Clinic, Barcelona, Spain
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Nicolai L, Pekayvaz K, Massberg S. Platelets: Orchestrators of immunity in host defense and beyond. Immunity 2024; 57:957-972. [PMID: 38749398 DOI: 10.1016/j.immuni.2024.04.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
12
|
Ran X, Zhang J, Wu Y, Du Y, Bao D, Pei H, Zhang Y, Zhou X, Li R, Tang X, She H, Mao Q. Prognostic gene landscapes and therapeutic insights in sepsis-induced coagulopathy. Thromb Res 2024; 237:1-13. [PMID: 38513536 DOI: 10.1016/j.thromres.2024.03.011] [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: 01/18/2024] [Revised: 02/24/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Sepsis is a common and critical condition encountered in clinical practice that can lead to multi-organ dysfunction. Sepsis-induced coagulopathy (SIC) significantly affects patient outcomes. However, the precise mechanisms remain unclear, making the identification of effective prognostic and therapeutic targets imperative. METHODS The analysis of transcriptome data from the whole blood of sepsis patients, facilitated the identification of key genes implicated in coagulation. Then we developed a prognostic model and a nomogram to predict patient survival. Consensus clustering classified sepsis patients into three subgroups for comparative analysis of immune function and immune cell infiltration. Single-cell sequencing elucidated alterations in intercellular communication between platelets and immune cells in sepsis, as well as the role of the coagulation-related gene FYN. Real-time quantitative PCR determined the mRNA levels of critical coagulation genes in septic rats' blood. Finally, administration of a FYN agonist to septic rats was observed for its effects on coagulation functions and survival. RESULTS This study identified four pivotal genes-CFD, FYN, ITGAM, and VSIG4-as significant predictors of survival in patients with sepsis. Among them, CFD, FYN, and ITGAM were underexpressed, while VSIG4 was upregulated in patients with sepsis. Moreover, a nomogram that incorporates the coagulation-related genes (CoRGs) risk score with clinical features of patients accurately predicted survival probabilities. Subgroup analysis of CoRGs expression delineated three molecular sepsis subtypes, each with distinct prognoses and immune profiles. Single-cell sequencing shed light on heightened communication between platelets and monocytes, T cells, and plasmacytoid dendritic cells, alongside reduced interactions with neutrophils in sepsis. The collagen signaling pathway was found to be essential in this dynamic. FYN may affect platelet function by modulating factors such as ELF1, PTCRA, and RASGRP2. The administration of the FYN agonist can effectively improve coagulation dysfunction and survival in septic rats. CONCLUSIONS The research identifies CoRGs as crucial prognostic markers for sepsis, highlighting the FYN gene's central role in coagulation disorders associated with the condition and suggesting novel therapeutic intervention strategies.
Collapse
Affiliation(s)
- Xiaoli Ran
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jun Zhang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yinyu Wu
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yunxia Du
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Daiqin Bao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Haoyu Pei
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yue Zhang
- Department of Medical Engineering, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaoqiong Zhou
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Rui Li
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xu Tang
- Department of Anesthesiology, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400042, China.
| | - Han She
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Qingxiang Mao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
13
|
Wu F, Ren Y, Lv W, Liu X, Wang X, Wang C, Cao Z, Liu J, Wei J, Pang Y. Generating dual structurally and functionally skin-mimicking hydrogels by crosslinking cell-membrane compartments. Nat Commun 2024; 15:802. [PMID: 38280863 PMCID: PMC10821872 DOI: 10.1038/s41467-024-45006-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
The skin is intrinsically a cell-membrane-compartmentalized hydrogel with high mechanical strength, potent antimicrobial ability, and robust immunological competence, which provide multiple protective effects to the body. Methods capable of preparing hydrogels that can simultaneously mimic the structure and function of the skin are highly desirable but have been proven to be a challenge. Here, dual structurally and functionally skin-mimicking hydrogels are generated by crosslinking cell-membrane compartments. The crosslinked network is formed via free radical polymerization using olefinic double bond-functionalized extracellular vesicles as a crosslinker. Due to the dissipation of stretching energy mediated by vesicular deformation, the obtained compartment-crosslinked network shows enhanced mechanical strength compared to hydrogels crosslinked by regular divinyl monomers. Biomimetic hydrogels also exhibit specific antibacterial activity and adequate ability to promote the maturation and activation of dendritic cells given the existence of numerous extracellular vesicle-associated bioactive substances. In addition, the versatility of this approach to tune both the structure and function of the resulting hydrogels is demonstrated through introducing a second network by catalyst-free click reaction-mediated crosslinking between alkyne-double-ended polymers and azido-decorated extracellular vesicles. This study provides a platform to develop dual structure- and function-controllable skin-inspired biomaterials.
Collapse
Affiliation(s)
- Feng Wu
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yusheng Ren
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenyan Lv
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, China
| | - Xiaobing Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, China
| | - Xinyue Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuhan Wang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenping Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyao Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jie Wei
- Shanghai Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China.
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
14
|
Robert M, Scherlinger M. Platelets are a major player and represent a therapeutic opportunity in systemic lupus erythematosus. Joint Bone Spine 2024; 91:105622. [PMID: 37495075 DOI: 10.1016/j.jbspin.2023.105622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by immune dysregulation and organ injury with a premature mortality due to cardiovascular diseases. Platelets, that are primarily known for their role in hemostasis, have been shown to play an active role in the pathogenesis and in the progression of immune-mediated inflammatory diseases. Here we summarize the evidence of their roles in SLE pathogenesis which supports the development of targeted treatments. Platelets and their precursors, the megakaryocytes, are intrinsically different in SLE patients compared with healthy controls. Different triggers related to innate and adaptive immunity activate platelets which release extracellular vesicles, soluble factors and interact with immune cells, thereby perpetuating inflammation. Platelets are involved in organ damage in SLE, especially in lupus nephritis and participate in the heightened cardiovascular mortality. They also play a clear role in antiphospholipid syndrome which can be associated with both thrombocytopenia and thrombosis. To tackle platelet activation and their interactions with immune cells now constitute promising therapeutic strategies in SLE.
Collapse
Affiliation(s)
- Marie Robert
- Service de médecine interne et immunologie clinique, centre hospitalier universitaire Édouard-Herriot, hospices civils de Lyon, Lyon, France
| | - Marc Scherlinger
- Service de rhumatologie, centre hospitalier universitaire de Strasbourg, 1, avenue Molière, 67098 Strasbourg, France; Laboratoire d'immuno-rhumatologie moléculaire, Institut national de la santé et de la recherche médicale (Inserm) UMR S 1109, Strasbourg, France; Centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), France.
| |
Collapse
|
15
|
Kauffman K, Manfra D, Nowakowska D, Zafari M, Nguyen PA, Phennicie R, Vollmann EH, O'Nuallain B, Basinski S, Komoroski V, Rooney K, Culyba EK, Wahle J, Ries C, Brehm M, Sazinsky S, Feldman I, Novobrantseva TI. PSGL-1 Blockade Induces Classical Activation of Human Tumor-associated Macrophages. CANCER RESEARCH COMMUNICATIONS 2023; 3:2182-2194. [PMID: 37819238 PMCID: PMC10601817 DOI: 10.1158/2767-9764.crc-22-0513] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/22/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
The immune suppressive microenvironment is a major culprit for difficult-to-treat solid cancers. Particularly, inhibitory tumor-associated macrophages (TAM) define the resistant nature of the tumor milieu. To define tumor-enabling mechanisms of TAMs, we analyzed molecular clinical datasets correlating cell surface receptors with the TAM infiltrate. Though P-selectin glycoprotein ligand-1 (PSGL-1) is found on other immune cells and functions as an adhesion molecule, PSGL-1 is highly expressed on TAMs across multiple tumor types. siRNA-mediated knockdown and antibody-mediated inhibition revealed a role for PSGL-1 in maintaining an immune suppressed macrophage state. PSGL-1 knockdown or inhibition enhanced proinflammatory mediator release across assays and donors in vitro. In several syngeneic mouse models, PSGL-1 blockade alone and in combination with PD-1 blockade reduced tumor growth. Using a humanized tumor model, we observed the proinflammatory TAM switch following treatment with an anti-PSGL-1 antibody. In ex vivo patient-derived tumor cultures, a PSGL-1 blocking antibody increased expression of macrophage-derived proinflammatory cytokines, as well as IFNγ, indicative of T-cell activation. Our data demonstrate that PSGL-1 blockade reprograms TAMs, offering a new therapeutic avenue to patients not responding to T-cell immunotherapies, as well as patients with tumors devoid of T cells. SIGNIFICANCE This work is a significant and actionable advance, as it offers a novel approach to treating patients with cancer who do not respond to T-cell checkpoint inhibitors, as well as to patients with tumors lacking T-cell infiltration. We expect that this mechanism will be applicable in multiple indications characterized by infiltration of TAMs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kate Rooney
- Verseau Therapeutics, Auburndale, Massachusetts
| | | | | | - Carola Ries
- Dr. Carola Ries Consulting, Penzberg, Germany
| | - Michael Brehm
- University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - Igor Feldman
- Verseau Therapeutics, Auburndale, Massachusetts
- Currently employed by Moderna Therapeutics, Cambridge, Massachusetts
| | - Tatiana I. Novobrantseva
- Verseau Therapeutics, Auburndale, Massachusetts
- Currently employed by Moderna Therapeutics, Cambridge, Massachusetts
| |
Collapse
|
16
|
Akilov OE. What Does the Future Hold for Biomarkers of Response to Extracorporeal Photopheresis for Mycosis Fungoides and Sézary Syndrome? Cells 2023; 12:2321. [PMID: 37759543 PMCID: PMC10527589 DOI: 10.3390/cells12182321] [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: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Extracorporeal photopheresis (ECP) is an FDA-approved immunotherapy for cutaneous T-cell lymphoma, which can provide a complete response in some patients. However, it is still being determined who will respond well, and predictive biomarkers are urgently needed to target patients for timely treatment and to monitor their response over time. The aim of this review is to analyze the current state of the diagnostic, prognostic, and disease state-monitoring biomarkers of ECP, and outline the future direction of the ECP biomarker discovery. Specifically, we focus on biomarkers of response to ECP in mycosis fungoides and Sézary syndrome. The review summarizes the current knowledge of ECP biomarkers, including their limitations and potential applications, and identifies key challenges in ECP biomarker discovery. In addition, we discuss emerging technologies that could revolutionize ECP biomarker discovery and accelerate the translation of biomarker research into clinical practice. This review will interest researchers and clinicians seeking to optimize ECP therapy for cutaneous T-cell lymphoma.
Collapse
Affiliation(s)
- Oleg E Akilov
- Cutaneous Lymphoma Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
17
|
Asensi Cantó P, Sanz Caballer J, Solves Alcaína P, de la Rubia Comos J, Gómez Seguí I. Extracorporeal Photopheresis in Graft-versus-Host Disease. Transplant Cell Ther 2023; 29:556-566. [PMID: 37419324 DOI: 10.1016/j.jtct.2023.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Graft-versus-host disease (GVHD) is a major cause of mortality and morbidity following allogeneic hematopoietic stem cell transplantation. Extracorporeal photopheresis (ECP), which exposes mononuclear cells to ultraviolet A irradiation in the presence of a photosensitizing agent, has shown efficacy in the treatment of GVHD. Recent observations in molecular and cell biology have revealed the mechanisms by which ECP can reverse GVHD, including lymphocyte apoptosis, differentiation of dendritic cells from circulating monocytes, and modification of the cytokine profile and T cell subpopulations. Technical innovations have made ECP accessible to a broader range of patients; however, logistical constraints may limit its use. In this review, we scrutinize the development of ECP from its origins to recent insights into the biology underlying ECP efficacy. We also review practical aspects that may complicate successful ECP treatment. Finally, we analyze how these theoretical concepts translate into clinical practice, summarizing the published experiences of leading research groups worldwide.
Collapse
Affiliation(s)
- Pedro Asensi Cantó
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.
| | - Jaime Sanz Caballer
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Pilar Solves Alcaína
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Javier de la Rubia Comos
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; School of Medicine and Dentistry, Catholic University of Valencia, Valencia, Spain
| | - Inés Gómez Seguí
- Haematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain; CIBERONC, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Abstract
Clinical flow cytometry tests for inherited and acquired platelet disorders are useful diagnostic tools but are not widely available. Flow cytometric methods are available to detect inherited glycoprotein deficiencies, granule release (secretion defects), drug-induced thrombocytopenias, presence of antiplatelet antibodies, and pharmacodynamic inhibition by antiplatelet agents. New tests take advantage of advanced multicolor cytometers and allow identification of novel platelet subsets by high-dimensional immunophenotyping. Studies are needed to evaluate the value of these new tests for diagnosis and monitoring of therapy in patients with platelet disorders.
Collapse
Affiliation(s)
- Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Benjamin E J Spurgeon
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Scherlinger M, Richez C, Tsokos GC, Boilard E, Blanco P. The role of platelets in immune-mediated inflammatory diseases. Nat Rev Immunol 2023; 23:495-510. [PMID: 36707719 PMCID: PMC9882748 DOI: 10.1038/s41577-023-00834-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 01/28/2023]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are characterized by excessive and uncontrolled inflammation and thrombosis, both of which are responsible for organ damage, morbidity and death. Platelets have long been known for their role in primary haemostasis, but they are now also considered to be components of the immune system and to have a central role in the pathogenesis of IMIDs. In patients with IMIDs, platelets are activated by disease-specific factors, and their activation often reflects disease activity. Here we summarize the evidence showing that activated platelets have an active role in the pathogenesis and the progression of IMIDs. Activated platelets produce soluble factors and directly interact with immune cells, thereby promoting an inflammatory phenotype. Furthermore, platelets participate in tissue injury and promote abnormal tissue healing, leading to fibrosis. Targeting platelet activation and targeting the interaction of platelets with the immune system are novel and promising therapeutic strategies in IMIDs.
Collapse
Affiliation(s)
- Marc Scherlinger
- Service de Rhumatologie, Centre de référence des maladies auto-immunes systémiques rares RESO, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Laboratoire d'ImmunoRhumatologie Moléculaire UMR_S 1109, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France.
| | - Christophe Richez
- Service de Rhumatologie, Centre de référence des maladies auto-immunes systémiques rares RESO, Hôpital Pellegrin, Centre Hospitalier Universitaire, Bordeaux, France
- CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, Bordeaux, France
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec, Canada
- Centre de Recherche ARThrite, Université Laval, Quebec City, Quebec, Canada
| | - Patrick Blanco
- CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, Bordeaux, France.
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Bordeaux, France.
| |
Collapse
|
20
|
Gao LC, Gong FQ. [Recent research on platelet-leukocyte aggregates and their role in the pathogenesis of Kawasaki disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:587-594. [PMID: 37382127 DOI: 10.7499/j.issn.1008-8830.2302066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Activated platelets may interact with various types of leukocytes such as monocytes, neutrophils, dendritic cells, and lymphocytes, trigger intercellular signal transduction, and thus lead to thrombosis and synthesis of massive inflammatory mediators. Elevated levels of circulating platelet-leukocyte aggregates have been found in patients with thrombotic or inflammatory diseases. This article reviews the latest research on the formation, function, and detection methods of platelet-leukocyte aggregates and their role in the onset of Kawasaki disease, so as to provide new ideas for studying the pathogenesis of Kawasaki disease.
Collapse
Affiliation(s)
- Li-Chao Gao
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine/National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Fang-Qi Gong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine/National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
21
|
Yang J, Wang X, Wu D, Yi K, Zhao Y. Yunnan Baiyao-loaded multifunctional microneedle patches for rapid hemostasis and cutaneous wound healing. J Nanobiotechnology 2023; 21:178. [PMID: 37280566 DOI: 10.1186/s12951-023-01936-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
Microneedle patches have been extensively employed for wound healing, while the lack of rapid hemostasis efficiency and multiple tissue-repair properties restrict their values in hemorrhagic wound applications. Herein, we propose a Yunnan Baiyao-loaded multifunctional microneedle patch, namely (BY + EGF)@MN, with deep tissue penetration, hemostasis efficiency and regenerative properties for hemorrhagic wound healing. The (BY + EGF)@MNs are designed with a BY-loaded Bletilla striata polysaccharide (BSP) base for rapid hemostasis and epidermal growth factor (EGF)-loaded GelMA tips for subsequent wound healing. As the BSP base can be fastly dissolved and completely release BY in 6 min to promote platelet adhesion and activate coagulation system, while the EGF can achieve a controlled and sustained release behavior in 7 days with the gradual degradation of the GelMA tips, the (BY + EGF)@MNs exhibit strong pro-coagulability and satisfactory hemostatic effect in a rat hepatic hemorrhage wound model. Based on the multifunctional characteristics, we have verified that when applied in rat cutaneous wounds, the proposed MNs can accelerate the wound healing process by enhancing neovascularization, fibroblast density, and collagen deposition. Thus, we believe that such (BY + EGF)@MNs are promising candidates for rapid hemostasis and diverse wound healing applications.
Collapse
Affiliation(s)
- Jie Yang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Xiaocheng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Dan Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Kexin Yi
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
22
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
23
|
Xiang Y, Xiang P, Zhang L, Li Y, Zhang J. A narrative review for platelets and their RNAs in cancers: New concepts and clinical perspectives. Medicine (Baltimore) 2022; 101:e32539. [PMID: 36596034 PMCID: PMC9803462 DOI: 10.1097/md.0000000000032539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent years have witnessed a growing body of evidence suggesting that platelets are involved in several stages of the metastatic process via direct or indirect interactions with cancer cells, contributing to the progression of neoplastic malignancies. Cancer cells can dynamically exchange components with platelets in and out of blood vessels, and directly phagocytose platelets to hijack their proteome, transcriptome, and secretome, or be remotely regulated by metabolites or microparticles released by platelets, resulting in phenotypic, genetic, and functional modifications. Moreover, platelet interactions with stromal and immune cells in the tumor microenvironment lead to alterations in their components, including the ribonucleic acid (RNA) profile, and complicate the impact of platelets on cancers. A deeper understanding of the roles of platelets and their RNAs in cancer will contribute to the development of anticancer strategies and the optimization of clinical management. Encouragingly, advances in high-throughput sequencing, bioinformatics data analysis, and machine learning have allowed scientists to explore the potential of platelet RNAs for cancer diagnosis, prognosis, and guiding treatment. However, the clinical application of this technique remains controversial and requires larger, multicenter studies with standardized protocols. Here, we integrate the latest evidence to provide a broader insight into the role of platelets in cancer progression and management, and propose standardized recommendations for the clinical utility of platelet RNAs to facilitate translation and benefit patients.
Collapse
Affiliation(s)
- Yunhui Xiang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Pinpin Xiang
- Department of Laboratory Medicine, Xiping Community Health Service Center of Longquanyi District Chengdu City, Chengdu, China
| | - Liuyun Zhang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanying Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Juan Zhang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- * Correspondence: Juan Zhang, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, 32# West Second Section, First Ring Road, Qingyang District, Chengdu City, Sichuan Province 610072, China (e-mail: )
| |
Collapse
|
24
|
Sun H, Du Y, Kumar R, Buchkovich N, He P. Increased circulating microparticles contribute to severe infection and adverse outcomes of COVID-19 in patients with diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H1176-H1193. [PMID: 36269646 PMCID: PMC9678425 DOI: 10.1152/ajpheart.00409.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 12/14/2022]
Abstract
Patients with diabetes infected with COVID-19 have greater mortality than those without comorbidities, but the underlying mechanisms remain unknown. This study aims to identify the mechanistic interactions between diabetes and severe COVID-19. Microparticles (MPs), the cell membrane-derived vesicles released on cell activation, are largely increased in patients with diabetes. To date, many mechanisms have been postulated for increased severity of COVID-19 in patients with underlying conditions, but the contributions of excessive MPs in patients with diabetes have been overlooked. This study characterizes plasma MPs from normal human subjects and patients with type 2 diabetes in terms of amount, cell origins, surface adhesive properties, ACE2 expression, spike protein binding capacity, and their roles in SARS-CoV-2 infection. Results showed that over 90% of plasma MPs express ACE2 that binds the spike protein of SARS-CoV-2. MPs in patients with diabetes increase 13-fold in quantity and 11-fold in adhesiveness when compared with normal subjects. Perfusion of human plasma with pseudo-typed SARS-CoV-2 virus or spike protein-bound MPs into human endothelial cell-formed microvessels-on-a chip demonstrated that MPs from patients with diabetes, not normal subjects, interact with endothelium and carry SARS-CoV-2 into cells through endocytosis, providing additional virus entry pathways and enhanced infection. Results also showed a large percentage of platelet-derived tissue factor-bearing MPs in diabetic plasma, which could contribute to thrombotic complications with SARS-CoV-2 infection. This study reveals a dual role of diabetic MPs in promoting SARS-CoV-2 entry and propagating vascular inflammation. These findings provide novel mechanistic insight into the high prevalence of COVID-19 in patients with diabetes and their propensity to develop severe vascular complications.NEW & NOTEWORTHY This study provides the first evidence that over 90% of human plasma microparticles express ACE2 that binds SARS-CoV-2 S protein with high affinity. Thus, the highly elevated adhesive circulating microparticles identified in patients with diabetes not only have greater SARS-CoV-2 binding capacity but also enable additional viral entry through virus-bound microparticle-endothelium interactions and enhanced infection. These findings reveal a novel mechanistic insight into the adverse outcomes of COVID-19 in patients with diabetes.
Collapse
Affiliation(s)
- Haoyu Sun
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Yong Du
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Rinki Kumar
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Nicholas Buchkovich
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| |
Collapse
|
25
|
Ding K, He Y, Wei J, Fu S, Wang J, Chen Z, Zhang H, Qu Y, Liang K, Gong X, Qiu L, Chen D, Xiao B, Du H. A score of DNA damage repair pathway with the predictive ability for chemotherapy and immunotherapy is strongly associated with immune signaling pathway in pan-cancer. Front Immunol 2022; 13:943090. [PMID: 36081518 PMCID: PMC9445361 DOI: 10.3389/fimmu.2022.943090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
DNA damage repair (DDR) is critical in maintaining normal cellular function and genome integrity and is associated with cancer risk, progression, and therapeutic response. However, there is still a lack of a thorough understanding of the effects of DDR genes’ expression level in cancer progression and therapeutic resistance. Therefore, we defined a tumor-related DDR score (TR-DDR score), utilizing the expression levels of 20 genes, to quantify the tumor signature of DNA damage repair pathways in tumors and explore the possible function and mechanism for the score among different cancers. The TR-DDR score has remarkably predictive power for tumor tissues. It is a more accurate indicator for the response of chemotherapy or immunotherapy combined with the tumor-infiltrating lymphocyte (TIL) and G2M checkpoint score than the pre-existing predictors (CD8 or PD-L1). This study points out that the TR-DDR score generally has positive correlations with patients of advanced-stage, genome-instability, and cell proliferation signature, while negative correlations with inflammatory response, apoptosis, and p53 pathway signature. In the context of tumor immune response, the TR-DDR score strongly positively correlates with the number of T cells (CD4+ activated memory cells, CD8+ cells, T regs, Tfh) and macrophages M1 polarization. In addition, by difference analysis and correlation analysis, COL2A1, MAGEA4, FCRL4, and ZIC1 are screened out as the potential modulating factors for the TR-DDR score. In summary, we light on a new biomarker for DNA damage repair pathways and explore its possible mechanism to guide therapeutic strategies and drug response prediction.
Collapse
Affiliation(s)
- Ke Ding
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Youhua He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- College of Life Science, Zhaoqing University, Zhaoqing, China
| | - Jiajian Wang
- Clinical Laboratory Department of Longgang District People’s Hospital of Shenzhen & The Second Affiliated Hospital of the Chinese University of Hong Kong, Shenzhen, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Haibo Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yimo Qu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Keying Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xiaocheng Gong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Li Qiu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dong Chen
- Fangrui Institute of Innovative Drugs, South China University of Technology, Guangzhou, China
| | - Botao Xiao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Botao Xiao, ; Hongli Du,
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Botao Xiao, ; Hongli Du,
| |
Collapse
|
26
|
Lee W, Suresh M. Vaccine adjuvants to engage the cross-presentation pathway. Front Immunol 2022; 13:940047. [PMID: 35979365 PMCID: PMC9376467 DOI: 10.3389/fimmu.2022.940047] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Adjuvants are indispensable components of vaccines for stimulating optimal immune responses to non-replicating, inactivated and subunit antigens. Eliciting balanced humoral and T cell-mediated immunity is paramount to defend against diseases caused by complex intracellular pathogens, such as tuberculosis, malaria, and AIDS. However, currently used vaccines elicit strong antibody responses, but poorly stimulate CD8 cytotoxic T lymphocyte (CTL) responses. To elicit potent CTL memory, vaccines need to engage the cross-presentation pathway, and this requirement has been a crucial bottleneck in the development of subunit vaccines that engender effective T cell immunity. In this review, we focus on recent insights into DC cross-presentation and the extent to which clinically relevant vaccine adjuvants, such as aluminum-based nanoparticles, water-in oil emulsion (MF59) adjuvants, saponin-based adjuvants, and Toll-like receptor (TLR) ligands modulate DC cross-presentation efficiency. Further, we discuss the feasibility of using carbomer-based adjuvants as next generation of adjuvant platforms to elicit balanced antibody- and T-cell based immunity. Understanding of the molecular mechanism of DC cross-presentation and the mode of action of adjuvants will pave the way for rational design of vaccines for infectious diseases and cancer that require balanced antibody- and T cell-based immunity.
Collapse
|
27
|
Sellars MC, Wu CJ, Fritsch EF. Cancer vaccines: Building a bridge over troubled waters. Cell 2022; 185:2770-2788. [PMID: 35835100 PMCID: PMC9555301 DOI: 10.1016/j.cell.2022.06.035] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/19/2022] [Accepted: 06/17/2022] [Indexed: 12/16/2022]
Abstract
Cancer vaccines aim to direct the immune system to eradicate cancer cells. Here we review the essential immunologic concepts underpinning natural immunity and highlight the multiple unique challenges faced by vaccines targeting cancer. Recent technological advances in mass spectrometry, neoantigen prediction, genetically and pharmacologically engineered mouse models, and single-cell omics have revealed new biology, which can help to bridge this divide. We particularly focus on translationally relevant aspects, such as antigen selection and delivery and the monitoring of human post-vaccination responses, and encourage more aggressive exploration of novel approaches.
Collapse
Affiliation(s)
- MacLean C Sellars
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
28
|
Nami M, Han P, Hanlon D, Tatsuno K, Wei B, Sobolev O, Pitruzzello M, Vassall A, Yosinski S, Edelson R, Reed M. Rapid Screen for Antiviral T-Cell Immunity with Nanowire Electrochemical Biosensors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109661. [PMID: 35165959 DOI: 10.1002/adma.202109661] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/08/2022] [Indexed: 06/14/2023]
Abstract
The ability to rapidly assess and monitor patient immune responses is critical for clinical diagnostics, vaccine design, and fundamental investigations into the presence or generation of protective immunity against infectious diseases. Recently, findings on the limits of antibody-based protection provided by B-cells have highlighted the importance of engaging pathogen-specific T-cells for long-lasting and broad protection against viruses and their emergent variants such as in SARS-CoV-2. However, low-cost and point-of-care tools for detecting engagement of T-cell immunity in patients are conspicuously lacking in ongoing efforts to assess and control population-wide disease risk. Currently available tools for human T-cell analysis are time and resource-intensive. Using multichannel silicon-nanowire field-effect transistors compatible with complementary metal-oxide-semiconductor, a device designed for rapid and label-free detection of human T-cell immune responses is developed. The generalizability of this approach is demonstrated by measuring T-cell responses against melanoma antigen MART1, common and seasonal viruses CMV, EBV, flu, as well as emergent pandemic coronavirus, SARS-CoV-2. Further, this device provides a modular and translational platform for optimizing vaccine formulations and combinations, offering quick and quantitative readouts for acquisition and persistence of T-cell immunity against variant-driven pathogens such as flu and pandemic SARS-CoV-2.
Collapse
Affiliation(s)
- Mohsen Nami
- Department of Electrical Engineering, School of Engineering and Applied Sciences, Yale University, 15 Prospect Street, New Haven, CT, 06511, USA
- Department of Neurosurgery, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Patrick Han
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
- Department of Immunobiology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Kazuki Tatsuno
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Brian Wei
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Mary Pitruzzello
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Aaron Vassall
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Shari Yosinski
- Department of Electrical Engineering, School of Engineering and Applied Sciences, Yale University, 15 Prospect Street, New Haven, CT, 06511, USA
| | - Richard Edelson
- Department of Dermatology, School of Medicine, Yale University, 333 Cedar St, New Haven, CT, 06510, USA
| | - Mark Reed
- Department of Electrical Engineering, School of Engineering and Applied Sciences, Yale University, 15 Prospect Street, New Haven, CT, 06511, USA
- Department of Applied Physics, School of Engineering and Applied Sciences, Yale University, 15 Prospect Street, New Haven, CT, 06511, USA
| |
Collapse
|
29
|
Wang J, Chen MS, Wang RS, Hu JQ, Liu S, Wang YYF, Xing XL, Zhang BW, Liu JM, Wang S. Current Advances in Structure-Function Relationships and Dose-Dependent Effects of Human Milk Oligosaccharides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6328-6353. [PMID: 35593935 DOI: 10.1021/acs.jafc.2c01365] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
HMOs (human milk oligosaccharides) are the third most important nutrient in breast milk. As complex glycans, HMOs play an important role in regulating neonatal intestinal immunity, resisting viral and bacterial infections, displaying anti-inflammatory characteristics, and promoting brain development. Although there have been some previous reports of HMOs, a detailed literature review summarizing the structure-activity relationships and dose-dependent effects of HMOs is lacking. Hence, after introducing the structures and synthetic pathways of HMOs, this review summarizes and categorizes identified structure-function relationships of HMOs. Differential mechanisms of different structural HMOs utilization by microorganisms are summarized. This review also emphasizes the recent advances in the interactions between different health benefits and the variance of dosage effect based on in vitro cell tests, animal experiments, and human intervention studies. The potential relationships between the chemical structure, the dosage selection, and the physiological properties of HMOs as functional foods are vital for further understanding of HMOs and their future applications.
Collapse
Affiliation(s)
- Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Meng-Shan Chen
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Rui-Shan Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Jia-Qiang Hu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Shuang Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Yuan-Yi-Fei Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Xiao-Long Xing
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Bo-Wei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Jing-Min Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
30
|
Zhao C, Chen Q, Li W, Zhang J, Yang C, Chen D. Multi-functional platelet membrane-camouflaged nanoparticles reduce neuronal apoptosis and regulate microglial phenotype during ischemic injury. APPLIED MATERIALS TODAY 2022; 27:101412. [DOI: 10.1016/j.apmt.2022.101412] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Chaoyue Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
- Changchun Children's Hospital, 1321Beian Road, Changchun, Jilin 130051, China
| | | | | | | | - Chunrong Yang
- Department of Pharmacy, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| |
Collapse
|
31
|
Chen X, Xu Y, Chen Q, Zhang H, Zeng Y, Geng Y, Shen L, Li F, Chen L, Chen GQ, Huang C, Liu J. The phosphatase PTEN links platelets with immune regulatory functions of mouse T follicular helper cells. Nat Commun 2022; 13:2762. [PMID: 35589797 PMCID: PMC9120038 DOI: 10.1038/s41467-022-30444-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
Beyond a function in hemostasis and thrombosis, platelets can regulate innate and adaptive immune responses. Hyperactive platelets are frequently associated with multiple human autoimmune diseases, yet their pathogenic functions in these diseases have not been fully established. Emerging studies show an essential function of the phosphatase and tensin homolog (PTEN) in maintenance of immune homeostasis. Here, we show that mice with platelet-specific deletion of Pten, develop age-related lymphoproliferative diseases and humoral autoimmunity not seen in wildtype animals. Platelet-specific Pten-deficient mice have aberrant T cell activation, excessive T follicular helper (Tfh) cell responses and accumulation of platelet aggregates in lymph nodes. Transferred Pten-deficient platelets are able to infiltrate into the peripheral lymphoid tissues and form more aggregates. Moreover, Pten-deficient platelets are hyperactive and overproduce multiple Tfh-promoting cytokines via activation of the PDK1/mTORC2-AKT-SNAP23 pathway. Pten-deficient platelets show enhanced interaction with CD4+ T cells and promote conversion of CD4+ T cells into Tfh cells. Our results implicate PTEN in platelet-mediated immune homeostasis, and provide evidence that hyperactive platelets function as an important mediator in autoimmune diseases using mouse models.
Collapse
Affiliation(s)
- Xue Chen
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China.
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qidi Chen
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Heng Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yu Zeng
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yan Geng
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Lei Shen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Fubin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Lei Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Guo-Qiang Chen
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Chuanxin Huang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
32
|
Lecot P, Ardin M, Dussurgey S, Alcazer V, Moudombi L, Pereira Abrantes M, Hubert M, Swalduz A, Hernandez‐Vargas H, Viari A, Caux C, Michallet M. Gene signature of circulating platelet‐bound neutrophils is associated with poor prognosis in cancer patients. Int J Cancer 2022; 151:138-152. [PMID: 35253899 PMCID: PMC9311065 DOI: 10.1002/ijc.33991] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 11/15/2022]
Abstract
Beyond their critical role in hemostasis, platelets physically interact with neutrophils to form neutrophil‐platelet aggregates (NPAs), enhancing neutrophil effector functions during inflammation. NPAs may also promote disease worsening in various inflammatory diseases. However, characterization of NPAs in cancer remains totally unexplored. Using ImageStreamX (ISX) imaging flow cytometer, we were not only allowed able to detect CD15+ CD14− CD36+ ITGA2B+ NPAs in both healthy donors' (HDs) and cancer patients' bloods, but we also showed that NPAs result from the binding of platelets preferentially to low‐density neutrophils (LDNs) as opposed to normal‐density neutrophils (NDNs). By reanalyzing two independent public scRNAseq data of whole blood leukocytes from cancer patients and HDs, we could identify a subset of neutrophils with high platelet gene expression that may correspond to NPAs. Moreover, we showed that cancer patients' derived NPAs possessed a distinct molecular signature compared to the other neutrophil subsets, independently of platelet genes. Gene ontology (GO) term enrichment analysis of this NPAs‐associated neutrophil transcriptomic signature revealed a significant enrichment of neutrophil degranulation, chemotaxis and trans‐endothelial migration GO terms. Lastly, using The Cancer Genome Atlas (TCGA), we could show by multivariate Cox analysis that the NPAs‐associated neutrophil transcriptomic signature was associated with a worse patient prognosis in several cancer types. These results suggest that neutrophils from NPAs are systemically primed by platelets empowering them with cancer progression capacities once at tumor site. NPAs may therefore hold clinical utility as novel noninvasive blood prognostic biomarker in cancer patients with solid tumors.
Collapse
Affiliation(s)
- Pacôme Lecot
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Maude Ardin
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Sébastien Dussurgey
- Université de Lyon, SFR Biosciences, ENS de Lyon, Inserm US8, CNRS UMS3444, UCBL ‐ 50 Avenue Tony Garnier Lyon France
| | - Vincent Alcazer
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Lyvia Moudombi
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Manuela Pereira Abrantes
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Margaux Hubert
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Aurélie Swalduz
- Department of Lung and Thoracic Medical Oncology Centre Léon Bérard Lyon France
| | - Hector Hernandez‐Vargas
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Alain Viari
- Synergie Lyon Cancer, Plateforme de Bio‐informatique ‘Gilles Thomas’ Lyon France
| | - Christophe Caux
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| | - Marie‐Cécile Michallet
- TERI (Tumor Escape, Resistance and Immunity) Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286 Lyon France
| |
Collapse
|
33
|
Mayer W, Kontekakis A, Maas C, Kuchenbecker U, Behlke S, Schennach H. Comparison of procedure times and collection efficiencies using integrated and multistep nonintegrated procedures for extracorporeal photopheresis. J Clin Apher 2022; 37:332-339. [PMID: 35225372 PMCID: PMC9542192 DOI: 10.1002/jca.21974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 01/01/2023]
Abstract
Background Extracorporeal photopheresis (ECP) is a blood‐based therapeutic procedure increasingly used for modulation of immune dysregulation in various underlying disease settings. The aim of this study was to compare the procedure times and blood collection efficiencies between the two approaches currently utilized in European centers: the integrated versus the multistep nonintegrated procedures. Methods A retrospective data analysis was conducted, comparing treatment data from patients who received ECP therapy at the Central Institute for Blood Transfusion & Department of Immunology (ZIB) of the Tirol Kliniken GmbH, where the integrated and multistep nonintegrated procedures are routinely used in an approximated setup. Results During the observation period, a total of 15 patients who were treated with alternating systems on 2 consecutive days were identified. This allowed treatment pair comparisons with minimal interpatient variabilities, similar to a cross‐over design even though analyzed retrospectively. Total average procedure times with the integrated system were 99.3 vs 122.0 minutes with the multistep nonintegrated procedures, respectively. Significant differences were observed for all steps of the ECP procedure: (a) time for buffy coat collection, 66.5 vs 74.7; (b) handling/transfer, 2.8 vs 18.7; (c) irradiation, 20.3 vs 11.7; and (d) reinfusion/handling time, 9.6 vs 16.3 minutes. The calculated collection throughput was 7.79 mL/min for the integrated and 7.84 mL/min for the multistep nonintegrated procedures, and with a white blood cell (WBC) collection efficiency of 34.2% and 21.0%, respectively. Conclusion The data presented in this study show a significant shorter overall procedure time and higher WBC collection efficiency for the integrated ECP system.
Collapse
Affiliation(s)
- Wolfgang Mayer
- Tirol Kliniken GmbH, Zentralinstitut für Bluttransfusion und Immunologische Abteilung (ZIB) Innsbruck Austria
| | | | | | | | - Susanne Behlke
- Mallinckrodt Pharmaceuticals, 3 Lotus Park, The Causeway Middlesex UK
| | - Harald Schennach
- Tirol Kliniken GmbH, Zentralinstitut für Bluttransfusion und Immunologische Abteilung (ZIB) Innsbruck Austria
| |
Collapse
|
34
|
Poli V, Di Gioia M, Sola-Visner M, Granucci F, Frelinger AL, Michelson AD, Zanoni I. Inhibition of transcription factor NFAT activity in activated platelets enhances their aggregation and exacerbates gram-negative bacterial septicemia. Immunity 2022; 55:224-236.e5. [PMID: 34995475 PMCID: PMC11318314 DOI: 10.1016/j.immuni.2021.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 09/09/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022]
Abstract
During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.
Collapse
Affiliation(s)
- Valentina Poli
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, MA, USA
| | - Marco Di Gioia
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, MA, USA
| | - Martha Sola-Visner
- Harvard Medical School, Boston Children's Hospital, Division of Newborn Medicine, Boston, MA, USA
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Alan D Michelson
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Boston, MA, USA; Harvard Medical School, Boston Children's Hospital, Division of Gastroenterology, Boston, MA, USA.
| |
Collapse
|
35
|
Abstract
Classically, platelets have been described as the cellular blood component that mediates hemostasis and thrombosis. This important platelet function has received significant research attention for >150 years. The immune cell functions of platelets are much less appreciated. Platelets interact with and activate cells of all branches of immunity in response to pathogen exposures and infection, as well as in response to sterile tissue injury. In this review, we focus on innate immune mechanisms of platelet activation, platelet interactions with innate immune cells, as well as the intersection of platelets and adaptive immunity. The immune potential of platelets is dependent in part on their megakaryocyte precursor providing them with the molecular composition to be first responders and immune sentinels in initiating and orchestrating coordinated pathogen immune responses. There is emerging evidence that extramedullary megakaryocytes may be immune differentiated compared with bone marrow megakaryocytes, but the physiological relevance of immunophenotypic differences are just beginning to be explored. These concepts are also discussed in this review. The immune functions of the megakaryocyte/platelet lineage have likely evolved to coordinate the need to repair a vascular breach with the simultaneous need to induce an immune response that may limit pathogen invasion once the blood is exposed to an external environment.
Collapse
Affiliation(s)
- Milka Koupenova
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605
| | - Alison Livada
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY 14642
- Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
36
|
O’Reilly D, Murphy CA, Drew R, El-Khuffash A, Maguire PB, Ainle FN, Mc Callion N. Platelets in pediatric and neonatal sepsis: novel mediators of the inflammatory cascade. Pediatr Res 2022; 91:359-367. [PMID: 34711945 PMCID: PMC8816726 DOI: 10.1038/s41390-021-01715-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
Sepsis, a dysregulated host response to infection, has been difficult to accurately define in children. Despite a higher incidence, especially in neonates, a non-specific clinical presentation alongside a lack of verified biomarkers has prevented a common understanding of this condition. Platelets, traditionally regarded as mediators of haemostasis and thrombosis, are increasingly associated with functions in the immune system with involvement across the spectrum of innate and adaptive immunity. The large number of circulating platelets (approx. 150,000 cells per microlitre) mean they outnumber traditional immune cells and are often the first to encounter a pathogen at a site of injury. There are also well-described physiological differences between platelets in children and adults. The purpose of this review is to place into context the platelet and its role in immunology and examine the evidence where available for its role as an immune cell in childhood sepsis. It will examine how the platelet interacts with both humoral and cellular components of the immune system and finally discuss the role the platelet proteome, releasate and extracellular vesicles may play in childhood sepsis. This review also examines how platelet transfusions may interfere with the complex relationships between immune cells in infection. IMPACT: Platelets are increasingly being recognised as important "first responders" to immune threats. Differences in adult and paediatric platelets may contribute to differing immune response to infections. Adult platelet transfusions may affect infant immune responses to inflammatory/infectious stimuli.
Collapse
Affiliation(s)
- Daniel O’Reilly
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Claire A. Murphy
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| | - Richard Drew
- grid.416068.d0000 0004 0617 7587Clinical Innovation Unit, Rotunda Hospital, Dublin, Ireland ,Irish Meningitis and Sepsis Reference Laboratory, Children’s Health Ireland at Temple Street, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Afif El-Khuffash
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| | - Patricia B. Maguire
- grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland
| | - Fionnuala Ni Ainle
- grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland ,grid.411596.e0000 0004 0488 8430Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland ,grid.416068.d0000 0004 0617 7587Department of Haematology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Medicine, University College Dublin, Dublin, Ireland
| | - Naomi Mc Callion
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| |
Collapse
|
37
|
Wang Y, Gong Y, Huang S, Xing X, Lv Z, Wang J, Yang JQ, Zhang G, Zhou Y, Han ST. Memristor-based biomimetic compound eye for real-time collision detection. Nat Commun 2021; 12:5979. [PMID: 34645801 PMCID: PMC8514515 DOI: 10.1038/s41467-021-26314-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
The lobula giant movement detector (LGMD) is the movement-sensitive, wide-field visual neuron positioned in the third visual neuropile of lobula. LGMD neuron can anticipate collision and trigger avoidance efficiently owing to the earlier occurring firing peak before collision. Vision chips inspired by the LGMD have been successfully implemented in very-large-scale-integration (VLSI) system. However, transistor-based chips and single devices to simulate LGMD neurons make them bulky, energy-inefficient and complicated. The devices with relatively compact structure and simple operation mode to mimic the escape response of LGMD neuron have not been realized yet. Here, the artificial LGMD visual neuron is implemented using light-mediated threshold switching memristor. The non-monotonic response to light flow field originated from the formation and break of Ag conductive filaments is analogue to the escape response of LGMD neuron. Furthermore, robot navigation with obstacle avoidance capability and biomimetic compound eyes with wide field-of-view (FoV) detection capability are demonstrated.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
- Hefei Innovation Research Institute, School of Microelectronics, Beihang University, 230013, Hefei, P. R. China
| | - Yue Gong
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Shenming Huang
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Xuechao Xing
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Ziyu Lv
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Junjie Wang
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Jia-Qin Yang
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Guohua Zhang
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Ye Zhou
- Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Su-Ting Han
- Institute of Microscale optoelectronics and College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, P. R. China.
| |
Collapse
|
38
|
Scherlinger M, Guillotin V, Douchet I, Vacher P, Boizard-Moracchini A, Guegan JP, Garreau A, Merillon N, Vermorel A, Ribeiro E, Machelart I, Lazaro E, Couzi L, Duffau P, Barnetche T, Pellegrin JL, Viallard JF, Saleh M, Schaeverbeke T, Legembre P, Truchetet ME, Dumortier H, Contin-Bordes C, Sisirak V, Richez C, Blanco P. Selectins impair regulatory T cell function and contribute to systemic lupus erythematosus pathogenesis. Sci Transl Med 2021; 13:13/600/eabi4994. [PMID: 34193612 DOI: 10.1126/scitranslmed.abi4994] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by a loss of tolerance toward self-nucleic acids, autoantibody production, interferon expression and signaling, and a defect in the regulatory T (Treg) cell compartment. In this work, we identified that platelets from patients with active SLE preferentially interacted with Treg cells via the P-selectin/P-selectin glycoprotein ligand-1 (PSGL-1) axis. Selectin interaction with PSGL-1 blocked the regulatory and suppressive properties of Treg cells and particularly follicular Treg cells by triggering Syk phosphorylation and an increase in intracytosolic calcium. Mechanistically, P-selectin engagement on Treg cells induced a down-regulation of the transforming growth factor-β axis, altering the phenotype of Treg cells and limiting their immunosuppressive responses. In patients with SLE, we found an up-regulation of P- and E-selectin both on microparticles and in their soluble forms that correlated with disease activity. Last, blocking P-selectin in a mouse model of SLE improved cardinal features of the disease, such as anti-dsDNA antibody concentrations and kidney pathology. Overall, our results identify a P-selectin-dependent pathway that is active in patients with SLE and validate it as a potential therapeutic avenue.
Collapse
Affiliation(s)
- Marc Scherlinger
- Department of Rheumatology, Pellegrin, Bordeaux University Hospital, 33076 Bordeaux, France.,Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Vivien Guillotin
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France.,Department of Internal Medicine, Saint André, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Isabelle Douchet
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | | | | | | | - Anne Garreau
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Nathalie Merillon
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Agathe Vermorel
- Nephrology Department, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Emmanuel Ribeiro
- Department of Internal Medicine, Saint André, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Irène Machelart
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,Department of Internal Medicine, Haut-Leveque, Bordeaux University Hospital, 33604, Pessac, France
| | - Estibaliz Lazaro
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,Department of Internal Medicine, Haut-Leveque, Bordeaux University Hospital, 33604, Pessac, France
| | - Lionel Couzi
- Nephrology Department, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Pierre Duffau
- Department of Internal Medicine, Saint André, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Thomas Barnetche
- Department of Rheumatology, Pellegrin, Bordeaux University Hospital, 33076 Bordeaux, France.,Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France
| | - Jean-Luc Pellegrin
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,Department of Internal Medicine, Haut-Leveque, Bordeaux University Hospital, 33604, Pessac, France
| | - Jean-François Viallard
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,Department of Internal Medicine, Haut-Leveque, Bordeaux University Hospital, 33604, Pessac, France
| | - Maya Saleh
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Thierry Schaeverbeke
- Department of Rheumatology, Pellegrin, Bordeaux University Hospital, 33076 Bordeaux, France.,Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France
| | - Patrick Legembre
- Contrôle de la Réponse Immune B et lymphoproliférations, CRIBL, UMR CNRS 7276, INSERM 1262, Limoges, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, Pellegrin, Bordeaux University Hospital, 33076 Bordeaux, France.,Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | | | - Cécile Contin-Bordes
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France.,Department of Immunology and Immunogenetics, Bordeaux University Hospital, 33076 Bordeaux, France
| | - Vanja Sisirak
- UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Christophe Richez
- Department of Rheumatology, Pellegrin, Bordeaux University Hospital, 33076 Bordeaux, France. .,Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France.,UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France
| | - Patrick Blanco
- Centre national de référence maladie auto-immune et systémique rares Est/Sud-Ouest (RESO), Bordeaux University Hospital, 33076 Bordeaux, France. .,UMR-CNRS 5164, ImmunoConcept, University of Bordeaux, 33076 Bordeaux, France.,Department of Immunology and Immunogenetics, Bordeaux University Hospital, 33076 Bordeaux, France
| |
Collapse
|
39
|
Aberrant protein glycosylation in cancer: implications in targeted therapy. Biochem Soc Trans 2021; 49:843-854. [PMID: 33704376 DOI: 10.1042/bst20200763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 12/28/2022]
Abstract
Aberrant cell surface glycosylation signatures are currently known to actively drive the neoplastic transformation of healthy cells. By disrupting the homeostatic functions of their protein carriers, cancer-associated glycans mechanistically underpin several molecular hallmarks of human malignancy. Furthermore, such aberrant glycan structures play key roles in the acquisition of molecular resistance to targeted therapeutic agents, which compromises their clinical efficacy, by modulating tumour cell aggressiveness and supporting the establishment of an immunosuppressive microenvironment. Recent advances in the study of the tumour cell glycoproteome have unravelled previously elusive molecular mechanisms of therapeutic resistance, guided the rational design of novel personalized therapeutic strategies, and may further improve the clinical performance of currently approved anti-cancer targeted agents. In this review, we highlight the impact of glycosylation in cancer targeted therapy, with particular focus on receptor tyrosine kinase-targeted therapy, immune checkpoints blockade therapy, and current developments on therapeutic strategies directed to glycan-binding proteins and other innovative glycan therapeutic strategies.
Collapse
|
40
|
Abstract
The immune system can recognize tumor cells to mount antigen-specific T cell response. Central to the establishment of T cell-mediated adaptive immunity are the inflammatory events that facilitate antigen presentation by stimulating the expression of MHC and costimulatory molecules and the secretion of pro-inflammatory cytokines. Such inflammatory events can be triggered upon cytotoxic treatments that induce immunogenic cancer cell death modalities. However, cancers have acquired a plethora of mechanisms to subvert, or to hide from, host-encoded immunosurveillance. Here, we discuss how tumor intrinsic oncogenic factors subvert desirable intratumoral inflammation by suppressing immunogenic cell death.
Collapse
Affiliation(s)
- Samuel T Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jonathan Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Inserm U1138, Paris, France.,Equipe 11 Labellisée Par La Ligue Nationale Contre Le Cancer, Centre De Recherche Des Cordeliers, Paris, France.,Université De Paris, Paris, France.,Sorbonne Université, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Inserm U1138, Paris, France.,Equipe 11 Labellisée Par La Ligue Nationale Contre Le Cancer, Centre De Recherche Des Cordeliers, Paris, France.,Université De Paris, Paris, France.,Sorbonne Université, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Institut Universitaire De France, Paris, France.,Pôle De Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
41
|
Puhm F, Boilard E, Machlus KR. Platelet Extracellular Vesicles: Beyond the Blood. Arterioscler Thromb Vasc Biol 2021; 41:87-96. [PMID: 33028092 PMCID: PMC7769913 DOI: 10.1161/atvbaha.120.314644] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are a means of cell-to-cell communication and can facilitate the exchange of a broad array of molecules between adjacent or distant cells. Platelets are anucleate cells derived from megakaryocytes and are primarily known for their role in maintaining hemostasis and vascular integrity. Upon activation by a variety of agonists, platelets readily generate EVs, which were initially identified as procoagulant particles. However, as both platelets and their EVs are abundant in blood, the role of platelet EVs in hemostasis may be redundant. Moreover, findings have challenged the significance of platelet-derived EVs in coagulation. Looking beyond hemostasis, platelet EV cargo is incredibly diverse and can include lipids, proteins, nucleic acids, and organelles involved in numerous other biological processes. Furthermore, while platelets cannot cross tissue barriers, their EVs can enter lymph, bone marrow, and synovial fluid. This allows for the transfer of platelet-derived content to cellular recipients and organs inaccessible to platelets. This review highlights the importance of platelet-derived EVs in physiological and pathological conditions beyond hemostasis.
Collapse
Affiliation(s)
- Florian Puhm
- Centre de recherche du CHU de Québec, Department of infectious diseases and immunity, Québec, QC, Canada
- Université Laval and Centre de recherche ARThrite, Québec, QC, Canada
| | - Eric Boilard
- Centre de recherche du CHU de Québec, Department of infectious diseases and immunity, Québec, QC, Canada
- Université Laval and Centre de recherche ARThrite, Québec, QC, Canada
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Interferon-α-Induced Dendritic Cells Generated with Human Platelet Lysate Exhibit Elevated Antigen Presenting Ability to Cytotoxic T Lymphocytes. Vaccines (Basel) 2020; 9:vaccines9010010. [PMID: 33374342 PMCID: PMC7823331 DOI: 10.3390/vaccines9010010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/06/2023] Open
Abstract
Given the recent advancements of immune checkpoint inhibitors, there is considerable interest in cancer immunotherapy provided through dendritic cell (DC)-based vaccination. Although many studies have been conducted to determine the potency of DC vaccines against cancer, the clinical outcomes are not yet optimal, and further improvement is necessary. In this study, we evaluated the potential ability of human platelet lysate (HPL) to produce interferon-α-induced DCs (IFN-DCs). In the presence of HPL, IFN-DCs (HPL-IFN-DCs) displayed high viability, yield, and purity. Furthermore, HPL-IFN-DCs displayed increased CD14, CD56, and CCR7 expressions compared with IFN-DCs produced without HPL; HPL-IFN-DCs induced an extremely higher number of antigen-specific cytotoxic T lymphocytes (CTLs) than IFN-DCs, which was evaluated with a human leukocyte antigen (HLA)-restricted melanoma antigen recognized by T cells 1 (MART-1) peptide. Additionally, the endocytic and proteolytic activities of HPL-IFN-DCs were increased. Cytokine production of interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α was also elevated in HPL-IFN-DCs, which may account for the enhanced CTL, endocytic, and proteolytic activities. Our findings suggest that ex-vivo-generated HPL-IFN-DCs are a novel monocyte-derived type of DC with high endocytic and proteolytic activities, thus highlighting a unique strategy for DC-based immunotherapies.
Collapse
|
43
|
Cai Z, Greene MI, Zhu Z, Zhang H. Structural Features and PF4 Functions that Occur in Heparin-Induced Thrombocytopenia (HIT) Complicated by COVID-19. Antibodies (Basel) 2020; 9:E52. [PMID: 33050376 PMCID: PMC7709132 DOI: 10.3390/antib9040052] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/10/2020] [Accepted: 09/29/2020] [Indexed: 01/08/2023] Open
Abstract
Platelet factor 4 (PF4, CXCL4) is a small chemokine protein released by activated platelets. Although a major physiological function of PF4 is to promote blood coagulation, this cytokine is involved in innate and adaptive immunity in events when platelets are activated in response to infections. Coronavirus disease 2019 (COVID-19) patients have abnormal coagulation activities, and severe patients develop higher D-dimer levels. D-dimers are small protein products present in the blood after blood clots are degraded by fibrinolysis. To prevent clotting, heparin is often clinically used in COVID-19 patients. Some clinical procedures for the management of COVID-19 patients may include extracorporeal membrane oxygenation (ECMO) and renal replacement therapy (CRRT), which also require the use of heparin. Anti-PF4 antibodies are frequently detected in severe patients and heparin-induced thrombocytopenia (HIT) can also be observed. PF4 and its role in HIT as well as in pathologies seen in COVID-19 patients define a potential therapeutic option of using blocking antibodies in the treatment of COVID-19.
Collapse
Affiliation(s)
| | | | | | - Hongtao Zhang
- Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19301, USA; (Z.C.); (M.I.G.); (Z.Z.)
| |
Collapse
|
44
|
Maouia A, Rebetz J, Kapur R, Semple JW. The Immune Nature of Platelets Revisited. Transfus Med Rev 2020; 34:209-220. [PMID: 33051111 PMCID: PMC7501063 DOI: 10.1016/j.tmrv.2020.09.005] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
Abstract
Platelets are the primary cellular mediators of hemostasis and this function firmly acquaints them with a variety of inflammatory processes. For example, platelets can act as circulating sentinels by expressing Toll-like receptors (TLR) that bind pathogens and this allows platelets to effectively kill them or present them to cells of the immune system. Furthermore, activated platelets secrete and express many pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. In addition, platelets can directly influence adaptive immune responses via secretion of, for example, CD40 and CD40L molecules. Platelets are also the source of most of the microvesicles in the circulation and these miniscule elements further enhance the platelet’s ability to communicate with the immune system. More recently, it has been demonstrated that platelets and their parent cells, the megakaryocytes (MK), can also uptake, process and present both foreign and self-antigens to CD8+ T-cells conferring on them the ability to directly alter adaptive immune responses. This review will highlight several of the non-hemostatic attributes of platelets that clearly and rightfully place them as integral players in immune reactions. Platelets can act as circulating sentinels by expressing pathogen-associated molecular pattern receptors that bind pathogens and induce their killing and elimination. Activated platelets secrete and express a multitude of pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. Platelets express and secrete many critical immunoregulatory molecules that significantly affect both innate and adaptive immune responses. Platelets are the primary source of microparticles in the circulation and these augment the platelet’s ability to communicate with the immune system. Platelets and megakaryocytes can act as antigen presenting cells and present both foreign- and self-peptides to T-cells.
Collapse
Affiliation(s)
- Amal Maouia
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Rick Kapur
- Sanquin Research, Department of Experimental Immunohematology, Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden.
| |
Collapse
|
45
|
Marcoux G, Laroche A, Espinoza Romero J, Boilard E. Role of platelets and megakaryocytes in adaptive immunity. Platelets 2020; 32:340-351. [PMID: 32597341 DOI: 10.1080/09537104.2020.1786043] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is comprised of two principal interconnected components called innate and adaptive immunity. While the innate immune system mounts a nonspecific response that provides protection against the spread of foreign pathogens, the adaptive immune system has developed to specifically recognize a given pathogen and lead to immunological memory. Platelets are small fragments produced from megakaryocytes in bone marrow and lungs. They circulate throughout the blood to monitor the integrity of the vasculature and to prevent bleeding. Given their large repertoire of immune receptors and inflammatory molecules, platelets and megakaryocytes can contribute to both innate and adaptive immunity. In adaptive immunity, platelets and megakaryocytes can process and present antigens to lymphocytes. Moreover, platelets, via FcγRIIA, rapidly respond to pathogens in an immune host when antibodies are present. This manuscript reviews the reported contributions of platelets and megakaryocytes with emphasis on antigen presentation and antibody response in adaptive immunity.
Collapse
Affiliation(s)
- Genevieve Marcoux
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Jenifer Espinoza Romero
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Eric Boilard
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| |
Collapse
|
46
|
Wei BM, Hanlon D, Khalil D, Han P, Tatsuno K, Sobolev O, Edelson RL. Extracorporeal Photochemotherapy: Mechanistic Insights Driving Recent Advances and Future Directions. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:145-159. [PMID: 32226344 PMCID: PMC7087063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, necessary for the initiation and maintenance of antigen-specific immunity and tolerance. Decades of research have been driven by hopes to harness the immunological capabilities of DCs and achieve physiological partnership with the immune system for therapeutic ends. Potential applications for DC-based immunotherapy include treatments for cancer, autoimmune disorders, and infectious diseases. However, DCs have poor availability in peripheral and lymphoid tissues and have poor survivability in culture, leading to the development of multiple strategies to generate and manipulate large numbers of DCs ex vivo. Among these is Extracorporeal Photopheresis (ECP), a widely used cancer immunotherapy. Recent advancements have uncovered that stimulation of monocyte-to-DC maturation via physiologic inflammatory signaling lies at the mechanistic core of ECP. Here, we describe the landscape of DC-based immunotherapy, the historical context of ECP, the current mechanistic understanding of ex vivo monocyte-to-DC maturation in ECP, and the implications of this understanding on making scientifically driven improvements to modern ECP protocols and devices.
Collapse
Affiliation(s)
- Brian M. Wei
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Douglas Hanlon
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - David Khalil
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT
| | - Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Olga Sobolev
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Richard L. Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, CT,To whom all correspondence should be addressed: Richard L. Edelson, MD, PO Box 208059, 333 Cedar St., New Haven, CT, 06520-8059; Tel: 203-785-4092, Fax: 203-737-5318,
| |
Collapse
|