1
|
Majumder D. Ischemic Stroke: Pathophysiology and Evolving Treatment Approaches. Neurosci Insights 2024; 19:26331055241292600. [PMID: 39444789 PMCID: PMC11497522 DOI: 10.1177/26331055241292600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Stroke remains a leading cause of mortality and disability, with ischemic stroke being the most common type. It occurs due to reduced cerebral blood flow, leading to a cascade of events initiated by oxygen and nutrient deprivation, triggering excitotoxicity, oxidative stress, and inflammation and finally culminating in neuronal injury and death. Key molecular players in ischemic stroke include glutamate receptors, acid-sensing ion channels, and purinergic receptors, exacerbating cellular damage through calcium influx, oxidative stress, and mitochondrial dysfunction. Understanding these mechanisms has shaped therapeutic strategies, such as neuroprotective agents and stem cell therapies. Current treatments such as tissue plasminogen activator (tPA) emphasize timely intervention, yet challenges persist in patient-specific variability and accessibility. This review provides an overview of ischemic stroke pathophysiology, emphasizing cellular responses to ischemia and current and future therapeutic approaches including stem cell therapies aimed at mitigating stroke-induced disabilities and improving long-term outcomes.
Collapse
|
2
|
Kostecki KL, Iida M, Crossman BE, Salgia R, Harari PM, Bruce JY, Wheeler DL. Immune Escape Strategies in Head and Neck Cancer: Evade, Resist, Inhibit, Recruit. Cancers (Basel) 2024; 16:312. [PMID: 38254801 PMCID: PMC10814769 DOI: 10.3390/cancers16020312] [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: 11/28/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Head and neck cancers (HNCs) arise from the mucosal lining of the aerodigestive tract and are often associated with alcohol use, tobacco use, and/or human papillomavirus (HPV) infection. Over 600,000 new cases of HNC are diagnosed each year, making it the sixth most common cancer worldwide. Historically, treatments have included surgery, radiation, and chemotherapy, and while these treatments are still the backbone of current therapy, several immunotherapies have recently been approved by the Food and Drug Administration (FDA) for use in HNC. The role of the immune system in tumorigenesis and cancer progression has been explored since the early 20th century, eventually coalescing into the current three-phase model of cancer immunoediting. During each of the three phases-elimination, equilibrium, and escape-cancer cells develop and utilize multiple strategies to either reach or remain in the final phase, escape, at which point the tumor is able to grow and metastasize with little to no detrimental interference from the immune system. In this review, we summarize the many strategies used by HNC to escape the immune system, which include ways to evade immune detection, resist immune cell attacks, inhibit immune cell functions, and recruit pro-tumor immune cells.
Collapse
Affiliation(s)
- Kourtney L. Kostecki
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Bridget E. Crossman
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
| | - Ravi Salgia
- Department of Medical Oncology and Experimental Therapeutics, Comprehensive Cancer Center, City of Hope, Duarte, CA 91010, USA;
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
| | - Justine Y. Bruce
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deric L. Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA; (K.L.K.); (M.I.); (B.E.C.)
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA;
| |
Collapse
|
3
|
Kumar R, Patil G, Dayal S. NLRP3-Induced NETosis: A Potential Therapeutic Target for Ischemic Thrombotic Diseases? Cells 2023; 12:2709. [PMID: 38067137 PMCID: PMC10706381 DOI: 10.3390/cells12232709] [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] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Ischemic thrombotic disease, characterized by the formation of obstructive blood clots within arteries or veins, is a condition associated with life-threatening events, such as stroke, myocardial infarction, deep vein thrombosis, and pulmonary embolism. The conventional therapeutic strategy relies on treatments with anticoagulants that unfortunately pose an inherent risk of bleeding complications. These anticoagulants primarily target clotting factors, often overlooking upstream events, including the release of neutrophil extracellular traps (NETs). Neutrophils are integral components of the innate immune system, traditionally known for their role in combating pathogens through NET formation. Emerging evidence has now revealed that NETs contribute to a prothrombotic milieu by promoting platelet activation, increasing thrombin generation, and providing a scaffold for clot formation. Additionally, NET components enhance clot stability and resistance to fibrinolysis. Clinical and preclinical studies have underscored the mechanistic involvement of NETs in the pathogenesis of thrombotic complications, since the clots obtained from patients and experimental models consistently exhibit the presence of NETs. Given these insights, the inhibition of NETs or NET formation is emerging as a promising therapeutic approach for ischemic thrombotic diseases. Recent investigations also implicate a role for the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome as a mediator of NETosis and thrombosis, suggesting that NLRP3 inhibition may also hold potential for mitigating thrombotic events. Therefore, future preclinical and clinical studies aimed at identifying and validating NLRP3 inhibition as a novel therapeutic intervention for thrombotic disorders are imperative.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Visakhapatnam 530045, India
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
4
|
Kroll RG, Powell C, Chen J, Snider NT, St. Hilaire C, Reddy A, Kim J, Pinsky DJ, Murthy VL, Sutton NR. Circulating Ectonucleotidases Signal Impaired Myocardial Perfusion at Rest and Stress. J Am Heart Assoc 2023; 12:e027920. [PMID: 37119076 PMCID: PMC10227209 DOI: 10.1161/jaha.122.027920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/20/2023] [Indexed: 04/30/2023]
Abstract
Background Ectonucleotidases maintain vascular homeostasis by metabolizing extracellular nucleotides, modulating inflammation and thrombosis, and potentially, myocardial flow through adenosine generation. Evidence implicates dysfunction or deficiency of ectonucleotidases CD39 or CD73 in human disease; the utility of measuring levels of circulating ectonucleotidases as plasma biomarkers of coronary artery dysfunction or disease has not been previously reported. Methods and Results A total of 529 individuals undergoing clinically indicated positron emission tomography stress testing between 2015 and 2019 were enrolled in this single-center retrospective analysis. Baseline demographics, clinical data, nuclear stress test, and coronary artery calcium score variables were collected, as well as a blood sample. CD39 and CD73 levels were assessed as binary (detectable, undetectable) or continuous variables using ELISAs. Plasma CD39 was detectable in 24% of White and 8% of Black study participants (P=0.02). Of the clinical history variables examined, ectonucleotidase levels were most strongly associated with underlying liver disease and not other traditional coronary artery disease risk factors. Intriguingly, detection of circulating ectonucleotidase was inversely associated with stress myocardial blood flow (2.3±0.8 mL/min per g versus 2.7 mL/min per g±1.1 for detectable versus undetectable CD39 levels, P<0.001) and global myocardial flow reserve (Pearson correlation between myocardial flow reserve and log(CD73) -0.19, P<0.001). A subanalysis showed these differences held true independent of liver disease. Conclusions Vasodilatory adenosine is the expected product of local ectonucleotidase activity, yet these data support an inverse relationship between plasma ectonucleotidases, stress myocardial blood flow (CD39), and myocardial flow reserve (CD73). These findings support the conclusion that plasma levels of ectonucleotidases, which may be shed from the endothelial surface, contribute to reduced stress myocardial blood flow and myocardial flow reserve.
Collapse
Affiliation(s)
- Rachel G. Kroll
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Corey Powell
- Consulting for Statistics, Computing, and Analytics ResearchUniversity of MichiganAnn ArborMI
| | - Jun Chen
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Natasha T. Snider
- Department of Cell Biology and PhysiologyUniversity of North Carolina at Chapel HillChapel HillNC
| | - Cynthia St. Hilaire
- Division of Cardiology, Departments of Medicine and BioengineeringVascular Medicine Institute, University of PittsburghPittsburghPAUSA
| | - Akshay Reddy
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Judy Kim
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - David J. Pinsky
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMI
| | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Nadia R. Sutton
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTN
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN
| |
Collapse
|
5
|
Wang Y, Zhu Y, Wang J, Dong L, Liu S, Li S, Wu Q. Purinergic signaling: A gatekeeper of blood-brain barrier permeation. Front Pharmacol 2023; 14:1112758. [PMID: 36825149 PMCID: PMC9941648 DOI: 10.3389/fphar.2023.1112758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
This review outlined evidence that purinergic signaling is involved in the modulation of blood-brain barrier (BBB) permeability. The functional and structural integrity of the BBB is critical for maintaining the homeostasis of the brain microenvironment. BBB integrity is maintained primarily by endothelial cells and basement membrane but also be regulated by pericytes, neurons, astrocytes, microglia and oligodendrocytes. In this review, we summarized the purinergic receptors and nucleotidases expressed on BBB cells and focused on the regulation of BBB permeability by purinergic signaling. The permeability of BBB is regulated by a series of purinergic receptors classified as P2Y1, P2Y4, P2Y12, P2X4, P2X7, A1, A2A, A2B, and A3, which serve as targets for endogenous ATP, ADP, or adenosine. P2Y1 and P2Y4 antagonists could attenuate BBB damage. In contrast, P2Y12-mediated chemotaxis of microglial cell processes is necessary for rapid closure of the BBB after BBB breakdown. Antagonists of P2X4 and P2X7 inhibit the activation of these receptors, reduce the release of interleukin-1 beta (IL-1β), and promote the function of BBB closure. In addition, the CD39/CD73 nucleotidase axis participates in extracellular adenosine metabolism and promotes BBB permeability through A1 and A2A on BBB cells. Furthermore, A2B and A3 receptor agonists protect BBB integrity. Thus, the regulation of the BBB by purinergic signaling is complex and affects the opening and closing of the BBB through different pathways. Appropriate selective agonists/antagonists of purinergic receptors and corresponding enzyme inhibitors could modulate the permeability of the BBB, effectively delivering therapeutic drugs/cells to the central nervous system (CNS) or limiting the entry of inflammatory immune cells into the brain and re-establishing CNS homeostasis.
Collapse
Affiliation(s)
| | | | - Junmeng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Longcong Dong
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuqing Liu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | | |
Collapse
|
6
|
Malekpour K, Hazrati A, Soudi S, Hashemi SM. Mechanisms behind therapeutic potentials of mesenchymal stem cell mitochondria transfer/delivery. J Control Release 2023; 354:755-769. [PMID: 36706838 DOI: 10.1016/j.jconrel.2023.01.059] [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: 12/11/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/29/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) perform their therapeutic effects through various mechanisms, including their ability to differentiate, producing different growth factors, immunomodulatory factors, and extracellular vesicles (EVs). In addition to the mentioned mechanisms, a new aspect of the therapeutic potential of MSCs has recently been noticed, which occurs through mitochondrial transfer. Various methods of MSCs mitochondria transfer have been used in studies to benefit from their therapeutic potential. Among these methods, mitochondrial transfer after MSCs transplantation in cell-to-cell contact, EVs-mediated transfer of mitochondria, and the use of MSCs isolated mitochondria (MSCs-mt) are well studied. Pathological conditions can affect the cells in the damaged microenvironment and lead to cells mitochondrial damage. Since the defect in the mitochondrial function of the cell leads to a decrease in ATP production and the subsequent cell death, restoring the mitochondrial content, functions, and hemostasis can affect the functions of the damaged cell. Various studies show that the transfer of MSCs mitochondria to other cells can affect vital processes such as proliferation, differentiation, cell metabolism, inflammatory responses, cell senescence, cell stress, and cell migration. These changes in cell attributes and behavior are very important for therapeutic purposes. For this reason, their investigation can play a significant role in the direction of the researchers'.
Collapse
Affiliation(s)
- Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran..
| |
Collapse
|
7
|
Noll JM, Augello CJ, Kürüm E, Pan L, Pavenko A, Nam A, Ford BD. Spatial Analysis of Neural Cell Proteomic Profiles Following Ischemic Stroke in Mice Using High-Plex Digital Spatial Profiling. Mol Neurobiol 2022; 59:7236-7252. [PMID: 36151369 PMCID: PMC9616789 DOI: 10.1007/s12035-022-03031-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/09/2022] [Indexed: 10/14/2022]
Abstract
Stroke is ranked as the fifth leading cause of death and the leading cause of adult disability in the USA. The progression of neuronal damage after stroke is recognized to be a complex integration of glia, neurons, and the surrounding extracellular matrix, therefore potential treatments must target the detrimental effects created by these interactions. In this study, we examined the spatial cellular and neuroinflammatory mechanisms occurring early after ischemic stroke utilizing Nanostring Digital Spatial Profiling (DSP) technology. Male C57bl/6 mice were subjected to photothrombotic middle cerebral artery occlusion (MCAO) and sacrificed at 3 days post-ischemia. Spatial distinction of the ipsilateral hemisphere was studied according to the regions of interest: the ischemic core, peri-infarct tissues, and peri-infarct normal tissue (PiNT) in comparison to the contralateral hemisphere. We demonstrated that the ipsilateral hemisphere initiates distinct spatial regulatory proteomic profiles with DSP technology that can be identified consistently with the immunohistochemical markers, FJB, GFAP, and Iba-1. The core border profile demonstrated an induction of neuronal death, apoptosis, autophagy, immunoreactivity, and early degenerative proteins. Most notably, the core border resulted in a decrease of the neuronal proteins Map2 and NeuN; an increase in the autophagy proteins BAG3 and CTSD; an increase in the microglial and peripheral immune invasion proteins Iba1, CD45, CD11b, and CD39; and an increase in the neurodegenerative proteins BACE1, APP, amyloid β 1-42, ApoE, and hyperphosphorylated tau protein S-199. The peri-infarct region demonstrated increased astrocytic, immunoreactivity, apoptotic, and neurodegenerative proteomic profiles, with an increase in BAG3, GFAP, and hyperphosphorylated tau protein S-199. The PiNT region displayed minimal changes compared to the contralateral cortex with only an increase in GFAP. In this study, we showed that mechanisms known to be associated with stroke, such as apoptosis and inflammation, occur in distinct spatial domains of the injured brain following ischemia. We also demonstrated the dysregulation of specific autophagic pathways that may lead to neurodegeneration in peri-infarct brain tissues. Taken together, these data suggest that identifying post-ischemic mechanisms occurring in a spatiotemporal manner may lead to more precise targets for successful therapeutic interventions to treat stroke.
Collapse
Affiliation(s)
- Jessica M Noll
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, 900 University Ave, Riverside, CA, 92521, USA
| | - Catherine J Augello
- Division of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Esra Kürüm
- Department of Statistics, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Liuliu Pan
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Anna Pavenko
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Andy Nam
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Byron D Ford
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
8
|
Baxevanis CN, Gritzapis AD, Voutsas IF, Batsaki P, Goulielmaki M, Adamaki M, Zoumpourlis V, Fortis SP. T-Cell Repertoire in Tumor Radiation: The Emerging Frontier as a Radiotherapy Biomarker. Cancers (Basel) 2022; 14:cancers14112674. [PMID: 35681654 PMCID: PMC9179913 DOI: 10.3390/cancers14112674] [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: 05/06/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Radiotherapy constitutes an essential component of the treatment for malignant disease. Besides its direct effect on cancer cells, namely, DNA damage and cell death, ionizing irradiation also mediates indirect antitumor effects that are mostly mediated by the immune system. Investigations into the processes underlying the interaction between radiotherapy and the immune system have uncovered mechanisms that can be exploited to promote the antitumor efficacy of radiotherapy both locally in the irradiated primary tumor and also at distant lesions in non-irradiated tumors. Because of its capacity to stimulate antitumor immunity, radiotherapy is also applied in combination with immune-checkpoint-inhibition-based immunotherapy. This review discusses the important pathways that govern the synergistic interactions between ionizing radiation and antitumor immune reactivity. Unravelling these involved mechanisms is mandatory for the successful application of anticancer radiotherapy and immunotherapy. We also place emphasis on the need for biomarkers that will aid in the selection of patients most likely to benefit from such combined treatments. Abstract Radiotherapy (RT) is a therapeutic modality that aims to eliminate malignant cells through the induction of DNA damage in the irradiated tumor site. In addition to its cytotoxic properties, RT also induces mechanisms that result in the promotion of antitumor immunity both locally within the irradiation field but also at distant tumor lesions, a phenomenon that is known as the “abscopal” effect. Because the immune system is capable of sensing the effects of RT, several treatment protocols have been assessing the synergistic role of radiotherapy combined with immunotherapy, collectively referred to as radioimmunotherapy. Herein, we discuss mechanistic insights underlying RT-based immunomodulation, which also enhance our understanding of how RT regulates antitumor T-cell-mediated immunity. Such knowledge is essential for the discovery of predictive biomarkers and for the improvement of clinical trials investigating the efficacy of radio-immunotherapeutic modalities in cancer patients.
Collapse
Affiliation(s)
- Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Ioannis F. Voutsas
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Panagiota Batsaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Vassilios Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece; (M.A.); (V.Z.)
| | - Sotirios P. Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 11522 Athens, Greece; (C.N.B.); (A.D.G.); (I.F.V.); (P.B.); (M.G.)
- Correspondence: ; Tel.: +30-2106409462
| |
Collapse
|
9
|
Zhang C, Frye MD, Riordan J, Sharma A, Manohar S, Salvi R, Sun W, Hu BH. Loss of CX3CR1 augments neutrophil infiltration into cochlear tissues after acoustic overstimulation. J Neurosci Res 2021; 99:2999-3020. [PMID: 34520571 DOI: 10.1002/jnr.24925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/16/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022]
Abstract
The cochlea, the sensory organ for hearing, has a protected immune environment, segregated from the systemic immune system by the blood-labyrinth barrier. Previous studies have revealed that acute acoustic injury causes the infiltration of circulating leukocytes into the cochlea. However, the molecular mechanisms controlling immune cell trafficking are poorly understood. Here, we report the role of CX3CR1 in regulating the entry of neutrophils into the cochlea after acoustic trauma. We employed B6.129P-Cx3cr1tm1Litt /J mice, a transgenic strain that lacks the gene, Cx3cr1, for coding the fractalkine receptor. Our results demonstrate that lack of Cx3cr1 results in the augmentation of neutrophil infiltration into cochlear tissues after exposure to an intense noise of 120 dB SPL for 1 hr. Neutrophil distribution in the cochlea is site specific, and the infiltration level is positively associated with noise intensity. Moreover, neutrophils are short lived and macrophage phagocytosis plays a role in neutrophil clearance, consistent with typical neutrophil dynamics in inflamed non-cochlear tissues. Importantly, our study reveals the potentiation of noise-induced hearing loss and sensory cell loss in Cx3cr1-/- mice. In wild-type control mice (Cx3cr1+/+ ) exposed to the same noise, we also found neutrophils. However, neutrophils were present primarily inside the microvessels of the cochlea, with only a few in the cochlear tissues. Collectively, our data implicate CX3CR1-mediated signaling in controlling neutrophil migration from the circulation into cochlear tissues and provide a better understanding of the impacts of neutrophils on cochlear responses to acoustic injury.
Collapse
Affiliation(s)
- Celia Zhang
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA.,Department of Audiology, School of Health Sciences, University of the Pacific, San Francisco, CA, USA
| | - Mitchell D Frye
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Juliana Riordan
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Ashu Sharma
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, The State University of New York, Buffalo, NY, USA
| | | | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Wei Sun
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Bo Hua Hu
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
10
|
Di Virgilio F, Vultaggio-Poma V, Sarti AC. P2X receptors in cancer growth and progression. Biochem Pharmacol 2020; 187:114350. [PMID: 33253643 DOI: 10.1016/j.bcp.2020.114350] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that ion channels have a crucial role in tumors, either as promoters of cancer cell growth, or modulators of immune cell functions, or both. Among ion channels, P2X receptors have a special status because they are gated by ATP, a common and abundant component of the tumor microenvironment. Furthermore, one P2X receptor, i.e. P2X7, may also function as a conduit for ATP release, thus fuelling the increased extracellular ATP level in the tumor interstitium. These findings show that P2X receptors and extracellular ATP are indissoluble partners and key regulators of tumor growth, and suggest the exploitation of the extracellular ATP-P2X partnership to develop innovative therapeutic approaches to cancer.
Collapse
|
11
|
Endogenous antisense RNA curbs CD39 expression in Crohn's disease. Nat Commun 2020; 11:5894. [PMID: 33208731 PMCID: PMC7676266 DOI: 10.1038/s41467-020-19692-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
CD39 is an ectonucleotidase that initiates conversion of extracellular nucleotides into immunosuppressive adenosine. CD39 is expressed by regulatory T (Treg)-cells, where it mediates immunosuppression, and by a subset of T-helper (Th) 17-cells, where it limits pathogenicity. CD39 is regulated via single-nucleotide-polymorphisms and upon activation of aryl-hydrocarbon-receptor and oxygen-mediated pathways. Here we report a mechanism of CD39 regulation that relies on the presence of an endogenous antisense RNA, transcribed from the 3′-end of the human CD39/ENTPD1 gene. CD39-specific antisense is increased in Treg and Th17-cells of Crohn’s disease patients over controls. It largely localizes in the cell nucleus and regulates CD39 by interacting with nucleolin and heterogeneous-nuclear-ribonucleoprotein-A1. Antisense silencing results in CD39 upregulation in vitro and amelioration of disease activity in a trinitro-benzene-sulfonic-acid model of colitis in humanized NOD/scid/gamma mice. Inhibition/blockade of antisense might represent a therapeutic strategy to restore CD39 along with immunohomeostasis in Crohn’s disease. CD39 is an ectonucleotidase associated with immunoregulatory function. Here authors show regulation of CD39 expression by an endogenous antisense RNA moiety transcribed from the 3‘ end of CD39/ENTPD1 which when itself is silenced results in amelioration of pathology in an animal model of colitis.
Collapse
|
12
|
Anyanwu AC, Kanthi Y, Fukase K, Liao H, Mimura T, Desch KC, Gruca M, Kaskar S, Sheikh-Aden H, Chi L, Zhao R, Yadav V, Wakefield TW, Hyman MC, Pinsky DJ. Tuning the Thromboinflammatory Response to Venous Flow Interruption by the Ectonucleotidase CD39. Arterioscler Thromb Vasc Biol 2020; 39:e118-e129. [PMID: 30816804 DOI: 10.1161/atvbaha.119.312407] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Objective- Leukocyte flux contributes to thrombus formation in deep veins under pathological conditions, but mechanisms that inhibit venous thrombosis are incompletely understood. Ectonucleotide di(tri)phosphohydrolase 1 ( ENTPD1 or Cd39), an ectoenzyme that catabolizes extracellular adenine nucleotides, is embedded on the surface of endothelial cells and leukocytes. We hypothesized that under venous stasis conditions, CD39 regulates inflammation at the vein:blood interface in a murine model of deep vein thrombosis. Approach and Results- CD39-null mice developed significantly larger venous thrombi under venous stasis, with more leukocyte recruitment compared with wild-type mice. Gene expression profiling of wild-type and Cd39-null mice revealed 76 differentially expressed inflammatory genes that were significantly upregulated in Cd39-deleted mice after venous thrombosis, and validation experiments confirmed high expression of several key inflammatory mediators. P-selectin, known to have proximal involvement in venous inflammatory and thrombotic events, was upregulated in Cd39-null mice. Inferior vena caval ligation resulted in thrombosis and a corresponding increase in both P-selectin and VWF (von Willebrand Factor) levels which were strikingly higher in mice lacking the Cd39 gene. These mice also manifest an increase in circulating platelet-leukocyte heteroaggregates suggesting heterotypic crosstalk between coagulation and inflammatory systems, which is amplified in the absence of CD39. Conclusions- These data suggest that CD39 mitigates the venous thromboinflammatory response to flow interruption.
Collapse
Affiliation(s)
- Anuli C Anyanwu
- From the Department of Molecular and Integrative Physiology (A.C.A., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor.,Section of Cardiology, Ann Arbor Veterans Health System, Michigan (Y.K.)
| | - Keigo Fukase
- Department of Cardiovascular Surgery, Awaji Medical Center, Hyogo, Japan (K.F.)
| | - Hui Liao
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Tekashi Mimura
- Department of Surgical Oncology, Hiroshima University, Japan (T.M.)
| | - Karl C Desch
- Department of Pediatrics (K.C.D.), University of Michigan Medical Center, Ann Arbor
| | - Martin Gruca
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Saabir Kaskar
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Hussein Sheikh-Aden
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Liguo Chi
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Raymond Zhao
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Vinita Yadav
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| | - Thomas W Wakefield
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories Ann Arbor, MI (T.W.W.)
| | - Matthew C Hyman
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia (M.C.H.)
| | - David J Pinsky
- From the Department of Molecular and Integrative Physiology (A.C.A., D.J.P.), University of Michigan Medical Center, Ann Arbor.,Division of Cardiovascular Medicine, Frankel Cardiovascular Center (Y.K., H.L., M.G., S.K., H.S.-A., L.C., R.Z., V.Y., D.J.P.), University of Michigan Medical Center, Ann Arbor
| |
Collapse
|
13
|
Herrera FG, Irving M, Kandalaft LE, Coukos G. Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol 2019; 20:e417-e433. [DOI: 10.1016/s1470-2045(19)30401-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
|
14
|
De Marchi E, Orioli E, Pegoraro A, Sangaletti S, Portararo P, Curti A, Colombo MP, Di Virgilio F, Adinolfi E. The P2X7 receptor modulates immune cells infiltration, ectonucleotidases expression and extracellular ATP levels in the tumor microenvironment. Oncogene 2019; 38:3636-3650. [PMID: 30655604 PMCID: PMC6756114 DOI: 10.1038/s41388-019-0684-y] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/06/2018] [Accepted: 12/23/2018] [Indexed: 01/22/2023]
Abstract
In the tumor microenvironment (TME) ATP and its receptor P2X7 exert a pivotal influence on cancer growth and tumor-host interactions. Here we analyzed the different effect of P2X7 genetic deficiency versus its antagonism on response against P2X7-expressing implanted tumors. We focused on immune cell expression of ATP degrading enzymes CD39 and CD73 and in vivo measured TME's ATP. The immune infiltrate of tumors growing in P2X7 null mice shows a decrease in CD8+ cells and an increased number of Tregs, overexpressing the fitness markers OX40, PD-1, and CD73. A similar Treg phenotype is also present in the spleen of tumor-bearing P2X7 null mice and it is paralleled by a decrease in proinflammatory cytokines and an increase in TGF-β. Differently, systemic administration of the P2X7 blocker A740003 in wild-type mice left unaltered the number of tumor-infiltrating CD8+ and Treg lymphocytes but increased CD4+ effector cells and decreased their expression of CD39 and CD73. P2X7 blockade did not affect spleen immune cell composition or ectonucleotidase expression but increased circulating INF-γ. Augmented CD73 in P2X7 null mice was mirrored by a decrease in TME ATP concentration and nucleotide reduced secretion from immune cells. On the contrary, TME ATP levels remained unaltered upon P2X7 antagonism, owing to release of ATP from cancerous cells and diminished ectonucleotidase expression by CD4+ and dendritic cells. These data point at P2X7 receptor as a key determinant of TME composition due to its combined action on immune cell infiltrate, ectonucleotidases, and ATP release.
Collapse
MESH Headings
- 5'-Nucleotidase/metabolism
- Adenosine Triphosphate/metabolism
- Animals
- Antigens, Differentiation/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/pathology
- Cell Line, Tumor
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Programmed Cell Death 1 Receptor/metabolism
- Purinergic P2X Receptor Antagonists/pharmacology
- Receptors, Purinergic P2X7/genetics
- Receptors, Purinergic P2X7/metabolism
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Elisa Orioli
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Anna Pegoraro
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Sabina Sangaletti
- Department of Experimental Oncology, Molecular Immunology Unit, Istituto Nazionale dei Tumori (IRCCS), Via Amadeo, 42, 20133, Milan, Italy
| | - Paola Portararo
- Department of Experimental Oncology, Molecular Immunology Unit, Istituto Nazionale dei Tumori (IRCCS), Via Amadeo, 42, 20133, Milan, Italy
| | - Antonio Curti
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, via Massarenti, 9, 40138, Bologna, Italy
| | - Mario Paolo Colombo
- Department of Experimental Oncology, Molecular Immunology Unit, Istituto Nazionale dei Tumori (IRCCS), Via Amadeo, 42, 20133, Milan, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy.
| |
Collapse
|
15
|
Yadav V, Chi L, Zhao R, Tourdot BE, Yalavarthi S, Jacobs BN, Banka A, Liao H, Koonse S, Anyanwu AC, Visovatti SH, Holinstat MA, Kahlenberg JM, Knight JS, Pinsky DJ, Kanthi Y. Ectonucleotidase tri(di)phosphohydrolase-1 (ENTPD-1) disrupts inflammasome/interleukin 1β-driven venous thrombosis. J Clin Invest 2019; 129:2872-2877. [PMID: 30990798 DOI: 10.1172/jci124804] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Deep vein thrombosis (DVT), caused by alterations in venous homeostasis is the third most common cause of cardiovascular mortality; however, key molecular determinants in venous thrombosis have not been fully elucidated. Several lines of evidence indicate that DVT occurs at the intersection of dysregulated inflammation and coagulation. The enzyme ectonucleoside tri(di)phosphohydrolase (ENTPD1, also known as CD39) is a vascular ecto-apyrase on the surface of leukocytes and the endothelium that inhibits intravascular inflammation and thrombosis by hydrolysis of phosphodiester bonds from nucleotides released by activated cells. Here, we evaluated the contribution of CD39 to venous thrombosis in a restricted-flow model of murine inferior vena cava stenosis. CD39-deficiency conferred a >2-fold increase in venous thrombogenesis, characterized by increased leukocyte engagement, neutrophil extracellular trap formation, fibrin, and local activation of tissue factor in the thrombotic milieu. This was orchestrated by increased phosphorylation of the p65 subunit of NFκB, activation of the NLRP3 inflammasome, and interleukin-1β (IL-1β) release in CD39-deficient mice. Substantiating these findings, an IL-1β-neutralizing antibody attenuated the thrombosis risk in CD39-deficient mice. These data demonstrate that IL-1β is a key accelerant of venous thrombo-inflammation, which can be suppressed by CD39. CD39 inhibits in vivo crosstalk between inflammation and coagulation pathways, and is a critical vascular checkpoint in venous thrombosis.
Collapse
Affiliation(s)
- Vinita Yadav
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center
| | - Liguo Chi
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center
| | - Raymond Zhao
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center
| | | | | | - Benjamin N Jacobs
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alison Banka
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center.,Department of Chemical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Hui Liao
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center
| | - Sharon Koonse
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center
| | - Anuli C Anyanwu
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | - David J Pinsky
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center.,Section of Cardiology, Ann Arbor Veterans Health System, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Huang Y, Gu Z, Fan Y, Zhai G, Zhao X, Sun Q, Shi Y, Lin G. Inhibition of the adenosinergic pathway: the indispensable part of oncological therapy in the future. Purinergic Signal 2019; 15:53-67. [PMID: 30809739 PMCID: PMC6439062 DOI: 10.1007/s11302-018-9641-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/04/2018] [Indexed: 02/08/2023] Open
Abstract
In recent years, immunotherapy has produced many unexpected breakthroughs in oncological therapy; however, it still has many deficiencies. For example, the number of patients who are unresponsive to anti-programmed death-ligand 1 (PD-L1), anti-cytotoxic T-like antigen-4 (CTLA4), and anti-programmed death-1 (PD1) therapies cannot be ignored, and the search for an undiscovered immunosuppressive pathway is imminent. Five decades ago, researchers found that activation of the adenosinergic pathway was negatively correlated with prognosis in many cancers. This review describes the entire process of the adenosinergic pathway in the tumor microenvironment and the mechanism of immunosuppression, which promotes tumor metastasis and drug resistance. Additionally, the review explores factors that regulate this pathway, including signaling factors secreted by the tumor microenvironment and certain anti-tumor drugs. Additionally, the combination of adenosinergic pathway inhibitors with chemotherapy, checkpoint blockade therapy, and immune cell-based therapy is summarized. Finally, certain issues regarding treatment via inhibition of this pathway and the use of targeted nanoparticles to reduce adverse reactions in patients are put forward in this review. Graphical Abstract The inhibitors of adenosinergic pathway loaded nanoparticles enter tumor tissue through EPR effect, and inhibit adenosinergic pathway to enhance or restore the effect of immune checkpoint blockade therapy, chemotherapies and immune cell-based therapy. Note: EPR means enhanced penetration and retention, × means blockade.
Collapse
Affiliation(s)
- Yi Huang
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Zili Gu
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Yang Fan
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Guangxi Zhai
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, Second Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Qifeng Sun
- Department of Thoracic Surgery, Second Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Yanbin Shi
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
17
|
Cicko S, Köhler TC, Ayata CK, Müller T, Ehrat N, Meyer A, Hossfeld M, Zech A, Di Virgilio F, Idzko M. Extracellular ATP is a danger signal activating P2X7 receptor in a LPS mediated inflammation (ARDS/ALI). Oncotarget 2018; 9:30635-30648. [PMID: 30093975 PMCID: PMC6078145 DOI: 10.18632/oncotarget.25761] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/19/2018] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threating lung condition resulting from a direct and indirect injury to the lungs [1, 2]. Pathophysiologically it is characterized by an acute alveolar damage, an increased permeability of the microvascular-barrier, leading to protein-rich pulmonary edema and subsequent impairment of arterial oxygenation and respiratory failure [1]. This study examined the role of extracellular ATP in recruiting inflammatory cells to the lung after induction of acute lung injury with lipopolysaccharide (LPS). However, the precise mechanism is poorly understood. Our objective was to investigate the functional role of the P2X7 receptor in the pathogenesis of acute respiratory distress syndrome (ARDS/ acute lung injury (ALI)) in vitro and in vivo. We show that intratracheally applied LPS causes an acute accumulation of ATP in the BALF (bronchoalveolar lavage) and lungs of mice. Prophylactic and therapeutic inhibition of P2X7R signalling by a specific antagonist and knock-out experiments was able to ameliorate the inflammatory response demonstrated by reduced ATP-levels, number of neutrophils and concentration of pro-inflammatory cytokine levels in the BALF. Experiments with chimeric mice showed that P2X7R expression on immune cells was responsible for the observed effect. Consistently, the inflammatory response is diminished only by a cell-type specific knockdown of P2X7 receptor on non-stationary immune cells. Since the results of BALF from patients with acute ARDS or pneumonia simulated the in vivo data after LPS exposure, the P2X7 receptor may be a new therapeutic target for treatment in acute respiratory distress syndrome (ARDS/ALI).
Collapse
Affiliation(s)
- Sanja Cicko
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | | | - Cemil Korcan Ayata
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | - Tobias Müller
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany.,Division of Pneumology, University Hospital RWTH Aachen, Aachen, Germany
| | - Nicolas Ehrat
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | - Anja Meyer
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | - Madelon Hossfeld
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | - Andreas Zech
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| | - Francesco Di Virgilio
- Department of Experimental and Diagnostic Medicine, University of Ferrara, Ferrara, Italy
| | - Marco Idzko
- University Hospital Freiburg, Department of Pneumology, Freiburg, Germany
| |
Collapse
|
18
|
Gülke E, Gelderblom M, Magnus T. Danger signals in stroke and their role on microglia activation after ischemia. Ther Adv Neurol Disord 2018; 11:1756286418774254. [PMID: 29854002 PMCID: PMC5968660 DOI: 10.1177/1756286418774254] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/10/2018] [Indexed: 12/26/2022] Open
Abstract
Ischemic stroke is a major cause of death. Besides the direct damage resulting from oxygen and glucose deprivation, sterile inflammation plays a pivotal role in increasing cellular death. Damaged-associated molecular patterns (DAMPs) are passively released from dying cells and activate the innate immune system. Thus, they take part in the direct and rapid activation of the inflammatory response after stroke onset. In this review the role of the most important DAMPs, high mobility group box 1, heat and cold shock proteins, purines, and peroxiredoxins, are addressed. Moreover, intracellular pathways activated by DAMPs in microglia are illuminated.
Collapse
Affiliation(s)
- Eileen Gülke
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | | |
Collapse
|
19
|
Tang JD, Lampe KJ. From de novo peptides to native proteins: advancements in biomaterial scaffolds for acute ischemic stroke repair. Biomed Mater 2018; 13:034103. [DOI: 10.1088/1748-605x/aaa4c3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
PINSKY DAVIDJ. CD39 AS A CRITICAL ECTONUCLEOTIDASE DEFENSE AGAINST PATHOLOGICAL VASCULAR REMODELING. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2018; 129:132-139. [PMID: 30166707 PMCID: PMC6116580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A common thread underlying vascular or tissue injury is the loss of plasmalemmal integrity and the passive (or even active) spillage of intracellular contents into the circulation. Purinergic nucleotides, which serve as energy shuttling moieties within cells, are among the contents released into the bloodstream, where they signal danger and trigger thrombosis and inflammation. To regain vascular homeostasis, vascular cells have evolved highly conserved mechanisms to transact the catalytic degradation of extracellular nucleotides such as adenosine triphosphate (ATP) and adenosine diphosphate (ADP). CD39, the main endothelial ectonucleotidase which cleaves ATP and ADP, plays an essential role in ridding the bloodstream of these danger signals, thereby sustaining vascular homeostasis. Studies herein describe the upregulation of endothelial CD39 gene by steady laminar shear forces, and conversely, its downregulation under turbulent flow conditions. CD39 appears to be a critical ectonucleotidase which suppresses atherogenesis under experimental hyperlipidemic conditions in mice, and which also significantly mitigates pathologic vascular remodeling and development of pulmonary arterial hypertension in mice placed under chronic hypoxic conditions. Together, these data reveal that CD39 opposes pathologic vascular remodeling under hyperlipidemic or hypoxic conditions. CD39 can therefore be viewed as a critical vascular homeostatic regulator to sustain vascular quiescence and to protect against pathological vascular remodeling in diseases as diverse as atherosclerosis and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- DAVID J. PINSKY
- Correspondence and reprint requests: David J. Pinsky, MD, University of Michigan Health Systems,
1500 E. Medical Center Drive, Suite 2141, Ann Arbor, Michigan 48109-5853734-936-3500
| |
Collapse
|
21
|
High Level P2X7-Mediated Signaling Impairs Function of Hematopoietic Stem/Progenitor Cells. Stem Cell Rev Rep 2017; 12:305-14. [PMID: 27059869 DOI: 10.1007/s12015-016-9651-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nucleotides, which bind to P2 receptors, have emerged as a family of mediators in intercellular communication. P2X7 is a member of the P2X family ligand-gated ion channels respond to extracellular ATP. High level expression of P2X7 was detected in leukemia samples, especially in relapsed cases. However, the role of P2X7 mediated signaling in hematopoietic stem/progenitor cells (HSPCs) as well as its potential role in leukemogenesis have not been established. In this study, the expression of P2X7 in hematopoietic cells in different lineages and stages was analyzed. Over-expression of P2X7 in HSPCs was carried out by retrovirus infection to study the impact on HSPCs. The results showed that low level expression of P2X7 was detected in HSPCs. Over-expression of P2X7 in HSPCs resulted in decreased colony forming ability in vitro and engraftment potential in vivo. These results suggested that high level purinergic signaling by P2X7 impaired function of HSPCs.
Collapse
|
22
|
Microglial-mediated PDGF-CC activation increases cerebrovascular permeability during ischemic stroke. Acta Neuropathol 2017; 134:585-604. [PMID: 28725968 PMCID: PMC5587628 DOI: 10.1007/s00401-017-1749-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022]
Abstract
Treatment of acute ischemic stroke with the thrombolytic tissue plasminogen activator (tPA) can significantly improve neurological outcomes; however, thrombolytic therapy is associated with an increased risk of intra-cerebral hemorrhage (ICH). Previously, we demonstrated that during stroke tPA acting on the parenchymal side of the neurovascular unit (NVU) can increase blood–brain barrier (BBB) permeability and ICH through activation of latent platelet-derived growth factor-CC (PDGF-CC) and signaling by the PDGF receptor-α (PDGFRα). However, in vitro, activation of PDGF-CC by tPA is very inefficient and the mechanism of PDGF-CC activation in the NVU is not known. Here, we show that the integrin Mac-1, expressed on brain microglia/macrophages (denoted microglia throughout), acts together with the endocytic receptor LRP1 in the NVU to promote tPA-mediated activation of PDGF-CC. Mac-1-deficient mice (Mac-1−/−) are protected from tPA-induced BBB permeability but not from permeability induced by intracerebroventricular injection of active PDGF-CC. Immunofluorescence analysis demonstrates that Mac-1, LRP1, and the PDGFRα all localize to the NVU of arterioles, and following middle cerebral artery occlusion (MCAO) Mac-1−/− mice show significantly less PDGFRα phosphorylation, BBB permeability, and infarct volume compared to wild-type mice. Bone-marrow transplantation studies indicate that resident CD11b+ cells, but not bone-marrow-derived leukocytes, mediate the early activation of PDGF-CC by tPA after MCAO. Finally, using a model of thrombotic stroke with late thrombolysis, we show that wild-type mice have an increased incidence of spontaneous ICH following thrombolysis with tPA 5 h after MCAO, whereas Mac-1−/− mice are resistant to the development of ICH even with late tPA treatment. Together, these results indicate that Mac-1 and LRP1 act as co-factors for the activation of PDGF-CC by tPA in the NVU, and suggest a novel mechanism for tightly regulating PDGFRα signaling in the NVU and controlling BBB permeability.
Collapse
|
23
|
Khoshnam SE, Winlow W, Farzaneh M. The Interplay of MicroRNAs in the Inflammatory Mechanisms Following Ischemic Stroke. J Neuropathol Exp Neurol 2017; 76:548-561. [DOI: 10.1093/jnen/nlx036] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
24
|
Petrovic-Djergovic D, Goonewardena SN, Pinsky DJ. Inflammatory Disequilibrium in Stroke. Circ Res 2017; 119:142-58. [PMID: 27340273 DOI: 10.1161/circresaha.116.308022] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/25/2016] [Indexed: 01/01/2023]
Abstract
Over the past several decades, there have been substantial advances in our knowledge of the pathophysiology of stroke. Understanding the benefits of timely reperfusion has led to the development of thrombolytic therapy as the cornerstone of current management of ischemic stroke, but there remains much to be learned about mechanisms of neuronal ischemic and reperfusion injury and associated inflammation. For ischemic stroke, novel therapeutic targets have continued to remain elusive. When considering modern molecular biological techniques, advanced translational stroke models, and clinical studies, a consistent pattern emerges, implicating perturbation of the immune equilibrium by stroke in both central nervous system injury and repair responses. Stroke triggers activation of the neuroimmune axis, comprised of multiple cellular constituents of the immune system resident within the parenchyma of the brain, leptomeninges, and vascular beds, as well as through secretion of biological response modifiers and recruitment of immune effector cells. This neuroimmune activation can directly impact the initiation, propagation, and resolution phases of ischemic brain injury. To leverage a potential opportunity to modulate local and systemic immune responses to favorably affect the stroke disease curve, it is necessary to expand our mechanistic understanding of the neuroimmune axis in ischemic stroke. This review explores the frontiers of current knowledge of innate and adaptive immune responses in the brain and how these responses together shape the course of ischemic stroke.
Collapse
Affiliation(s)
- Danica Petrovic-Djergovic
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor
| | - Sascha N Goonewardena
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor
| | - David J Pinsky
- From the Departments of Internal Medicine (D.P.-D., S.N.G., D.J.P.) and Molecular and Integrative Physiology (D.J.P.), University of Michigan, Ann Arbor.
| |
Collapse
|
25
|
Baek AE, Sutton NR, Petrovic-Djergovic D, Liao H, Ray JJ, Park J, Kanthi Y, Pinsky DJ. Ischemic Cerebroprotection Conferred by Myeloid Lineage-Restricted or Global CD39 Transgene Expression. Circulation 2017; 135:2389-2402. [PMID: 28377485 DOI: 10.1161/circulationaha.116.023301] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/22/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cerebral tissue damage after an ischemic event can be exacerbated by inflammation and thrombosis. Elevated extracellular ATP and ADP levels are associated with cellular injury, inflammation, and thrombosis. Ectonucleoside triphosphate diphosphohydrolase-1 (CD39), an enzyme expressed on the plasmalemma of leukocytes and endothelial cells, suppresses platelet activation and leukocyte infiltration by phosphohydrolyzing ATP/ADP. To investigate the effects of increased CD39 in an in vivo cerebral ischemia model, we developed a transgenic mouse expressing human CD39 (hCD39). METHODS A floxed-stop sequence was inserted between the promoter and the hCD39 transcriptional start site, generating a mouse in which the expression of hCD39 can be controlled tissue-specifically using Cre recombinase mice. We generated mice that express hCD39 globally or in myeloid-lineage cells only. Cerebral ischemia was induced by middle cerebral artery occlusion. Infarct volumes were quantified by MRI after 48 hours. RESULTS Both global and transgenic hCD39- and myeloid lineage CD39-overexpressing mice (transgenic, n=9; myeloid lineage, n=6) demonstrated significantly smaller cerebral infarct volumes compared with wild-type mice. Leukocytes from ischemic and contralateral hemispheres were analyzed by flow cytometry. Although contralateral hemispheres had equal numbers of macrophages and neutrophils, ischemic hemispheres from transgenic mice had less infiltration (n=4). Transgenic mice showed less neurological deficit compared with wild-type mice (n=6). CONCLUSIONS This is the first report of transgenic overexpression of CD39 in mice imparting a protective phenotype after stroke, with reduced leukocyte infiltration, smaller infarct volumes, and decreased neurological deficit. CD39 overexpression, either globally or in myeloid lineage cells, quenches postischemic leukosequestration and reduces stroke-induced neurological injury.
Collapse
Affiliation(s)
- Amy E Baek
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Nadia R Sutton
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Danica Petrovic-Djergovic
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Hui Liao
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Jessica J Ray
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Joan Park
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - Yogendra Kanthi
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.)
| | - David J Pinsky
- From Department of Molecular and Integrative Physiology (A.E.B., D.J.P.) and Department of Internal Medicine (N.R.S., D.P.-D, H.L., J.R., Y.K., D.J.P.), Division of Cardiovascular Medicine University of Michigan Medical Center, Ann Arbor; and Section of Cardiology, VA Ann Arbor Healthcare System, MI (J.P.).
| |
Collapse
|
26
|
Wennerberg E, Lhuillier C, Vanpouille-Box C, Pilones KA, García-Martínez E, Rudqvist NP, Formenti SC, Demaria S. Barriers to Radiation-Induced In Situ Tumor Vaccination. Front Immunol 2017; 8:229. [PMID: 28348554 PMCID: PMC5346586 DOI: 10.3389/fimmu.2017.00229] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/17/2017] [Indexed: 12/11/2022] Open
Abstract
The immunostimulatory properties of radiation therapy (RT) have recently generated widespread interest due to preclinical and clinical evidence that tumor-localized RT can sometimes induce antitumor immune responses mediating regression of non-irradiated metastases (abscopal effect). The ability of RT to activate antitumor T cells explains the synergy of RT with immune checkpoint inhibitors, which has been well documented in mouse tumor models and is supported by observations of more frequent abscopal responses in patients refractory to immunotherapy who receive RT during immunotherapy. However, abscopal responses following RT remain relatively rare in the clinic, and antitumor immune responses are not effectively induced by RT against poorly immunogenic mouse tumors. This suggests that in order to improve the pro-immunogenic effects of RT, it is necessary to identify and overcome the barriers that pre-exist and/or are induced by RT in the tumor microenvironment. On the one hand, RT induces an immunogenic death of cancer cells associated with release of powerful danger signals that are essential to recruit and activate dendritic cells (DCs) and initiate antitumor immune responses. On the other hand, RT can promote the generation of immunosuppressive mediators that hinder DCs activation and impair the function of effector T cells. In this review, we discuss current evidence that several inhibitory pathways are induced and modulated in irradiated tumors. In particular, we will focus on factors that regulate and limit radiation-induced immunogenicity and emphasize current research on actionable targets that could increase the effectiveness of radiation-induced in situ tumor vaccination.
Collapse
Affiliation(s)
- Erik Wennerberg
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY , USA
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY , USA
| | | | - Karsten A Pilones
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY , USA
| | - Elena García-Martínez
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA; Department of Hematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
| | | | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY , USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine , New York, NY , USA
| |
Collapse
|
27
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
28
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017. [PMID: 28258700 DOI: 10.1111/imr.12528] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC, Canada.,Faculté de Pharmacie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
29
|
Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 2017; 276:121-144. [PMID: 28258700 PMCID: PMC5338647 DOI: 10.1111/imr.12528] [Citation(s) in RCA: 659] [Impact Index Per Article: 82.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancers are able to grow by subverting immune suppressive pathways, to prevent the malignant cells as being recognized as dangerous or foreign. This mechanism prevents the cancer from being eliminated by the immune system and allows disease to progress from a very early stage to a lethal state. Immunotherapies are newly developing interventions that modify the patient's immune system to fight cancer, by either directly stimulating rejection-type processes or blocking suppressive pathways. Extracellular adenosine generated by the ectonucleotidases CD39 and CD73 is a newly recognized "immune checkpoint mediator" that interferes with anti-tumor immune responses. In this review, we focus on CD39 and CD73 ectoenzymes and encompass aspects of the biochemistry of these molecules as well as detailing the distribution and function on immune cells. Effects of CD39 and CD73 inhibition in preclinical and clinical studies are discussed. Finally, we provide insights into potential clinical application of adenosinergic and other purinergic-targeting therapies and forecast how these might develop in combination with other anti-cancer modalities.
Collapse
Affiliation(s)
- Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Maria Serena Longhi
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - Simon C. Robson
- Divisions of Gastroenterology and Transplantation, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, USA. 02215
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal et Institut du Cancer de Montréal, Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
| |
Collapse
|
30
|
Marutani T, Hattori T, Tsutsumi K, Koike Y, Harada A, Noguchi K, Kiso Y, Mukai H. Mitochondrial protein-derived cryptides: Are endogenous N-formylated peptides including mitocryptide-2 components of mitochondrial damage-associated molecular patterns? Biopolymers 2017; 106:580-7. [PMID: 26600263 DOI: 10.1002/bip.22788] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 11/08/2022]
Abstract
Recently, much attention has been paid to "nonclassical" bioactive peptides, which are fragmented peptides simultaneously produced during maturation and degradation of various functional proteins. We identified many fragmented peptides derived from various mitochondrial proteins including mitocryptide-1 and mitocryptide-2 that efficiently activate neutrophils. These endogenous, functionally active, fragmented peptides are referred to as "cryptides." Among them, mitocryptide-2 is an N-formylated cryptide cleaved from mitochondrial cytochrome b that is encoded in mitochondrial DNA (mtDNA). It is known that 13 proteins encoded in mtDNA are translated in mitochondria as N-formylated forms, suggesting the existence of endogenous N-formylated peptides other than mitocryptide-2. Here, we investigated the effects of N-formylated peptides presumably cleaved from mtDNA-encoded proteins other than cytochrome b on the functions of neutrophilic cells to elucidate possible regulation by endogenous N-formylated cryptides. Four N-formylated cryptides derived from cytochrome c oxidase subunit I and NADH dehydrogenase subunits 4, 5, and 6 among 12 peptides from mtDNA-encoded proteins efficiently induced not only migration but also β-hexosaminidase release, which is an indicator of neutrophilic phagocytosis, in HL-60 cells differentiated into neutrophilic cells. These activities were comparable to or higher than those induced by mitocryptide-2. Although endogenous N-formylated peptides that are contained in mitochondrial damage-associated molecular patterns (DAMPs) have yet to be molecularly identified, they have been implicated in innate immunity. Thus, N-formylated cryptides including mitocryptide-2 are first-line candidates for the contents of mitochondrial DAMPs to promote innate immune responses. © 2015 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 580-587, 2016.
Collapse
Affiliation(s)
- Takayuki Marutani
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Tatsuya Hattori
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Koki Tsutsumi
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Yusuke Koike
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Akihiko Harada
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Kosuke Noguchi
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Yoshiaki Kiso
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Hidehito Mukai
- Laboratory of Peptide Science, Graduate School of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| |
Collapse
|
31
|
Sutton NR, Hayasaki T, Hyman MC, Anyanwu AC, Liao H, Petrovic-Djergovic D, Badri L, Baek AE, Walker N, Fukase K, Kanthi Y, Visovatti SH, Horste EL, Ray JJ, Goonewardena SN, Pinsky DJ. Ectonucleotidase CD39-driven control of postinfarction myocardial repair and rupture. JCI Insight 2017; 2:e89504. [PMID: 28097233 PMCID: PMC5213916 DOI: 10.1172/jci.insight.89504] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
Mechanical complications of myocardial infarction (MI) are often fatal. Little is known about endogenous factors that predispose to myocardial rupture after MI. Ectonucleoside triphosphate diphosphohydrolase (CD39) could be a critical mediator of propensity to myocardial rupture after MI due to its role in modulating inflammation and thrombosis. Using a model of permanent coronary artery ligation, rupture was virtually abrogated in cd39-/- mice versus cd39+/+ controls, with elevated fibrin and collagen deposition and marked neutrophil and macrophage influx. Macrophages were found to display increased surface expression of CD301 and CD206, marking a reparative phenotype, driven by increased extracellular ATP and IL-4 in the infarcted myocardium of cd39-/- mice. A myeloid-specific CD39-knockout mouse also demonstrated protection from rupture, with an attenuated rupture phenotype, suggesting that complete ablation of CD39 provides the greatest degree of protection in this model. Absence of CD39, either globally or in a myeloid lineage-restricted fashion, skews the phenotype toward alternatively activated (reparative) macrophage infiltration following MI. These studies reveal a previously unrecognized and unexpected role of endogenous CD39 to skew macrophage phenotype and promote a propensity to myocardial rupture after MI.
Collapse
Affiliation(s)
- Nadia R. Sutton
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Takanori Hayasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Matthew C. Hyman
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center
| | - Anuli C. Anyanwu
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center
| | - Hui Liao
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | | | - Linda Badri
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Amy E. Baek
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center
| | - Natalie Walker
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Keigo Fukase
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
- Section of Cardiology, Veterans Affairs Medical Center, Ann Arbor, Michigan, USA
| | - Scott H. Visovatti
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Ellen L. Horste
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Jessica J. Ray
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
| | - Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
- Section of Cardiology, Veterans Affairs Medical Center, Ann Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center
| |
Collapse
|
32
|
McNamara HA, Cockburn IA. The three Rs: Recruitment, Retention and Residence of leukocytes in the liver. Clin Transl Immunology 2016; 5:e123. [PMID: 28435674 PMCID: PMC5384287 DOI: 10.1038/cti.2016.84] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/20/2022] Open
Abstract
The composition of leukocytes in the liver is highly distinct from that of the blood and lymphoid organs. In particular, the liver is highly enriched in non-conventional T cells such as natural killer T (NKT) cells, γδ T cells and mucosal-associated invariant T cells. In addition, there are significant populations of tissue-resident NK cells (or innate lymphoid cells (ILC1)) and memory CD8+ T cells. These cells are joined in conditions of inflammation by neutrophils, monocytes and macrophages. In recent years a multitude of studies have generated insights into how these cells arrest, move and remain resident in the liver. This new understanding has largely been due to the use of intra-vital microscopy to track immune cells in the liver, coupled with gene expression profiling and parabiosis techniques. These studies have revealed that leukocyte recruitment in the liver does not correspond to the classical paradigm of the leukocyte adhesion cascade. Rather, both lymphoid and myeloid cells have been found to adhere in the liver sinusoids in a platelet-dependent manner. Leukocytes have also been observed to patrol the hepatic sinusoids using a characteristic crawling motility. Moreover, T cells have been observed surveying hepatocytes for antigen through the unique fenestrated endothelium of the liver sinusoids, potentially negating the need for extravasation. In this review we highlight some of these recent discoveries and examine the different molecular interactions required for the recruitment, retention and-in some cases-residence of diverse leukocyte populations within the liver.
Collapse
Affiliation(s)
- Hayley A McNamara
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
33
|
Middleton EA, Weyrich AS, Zimmerman GA. Platelets in Pulmonary Immune Responses and Inflammatory Lung Diseases. Physiol Rev 2016; 96:1211-59. [PMID: 27489307 PMCID: PMC6345245 DOI: 10.1152/physrev.00038.2015] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Platelets are essential for physiological hemostasis and are central in pathological thrombosis. These are their traditional and best known activities in health and disease. In addition, however, platelets have specializations that broaden their functional repertoire considerably. These functional capabilities, some of which are recently discovered, include the ability to sense and respond to infectious and immune signals and to act as inflammatory effector cells. Human platelets and platelets from mice and other experimental animals can link the innate and adaptive limbs of the immune system and act across the immune continuum, often also linking immune and hemostatic functions. Traditional and newly recognized facets of the biology of platelets are relevant to defensive, physiological immune responses of the lungs and to inflammatory lung diseases. The emerging view of platelets as blood cells that are much more diverse and versatile than previously thought further predicts that additional features of the biology of platelets and of megakaryocytes, the precursors of platelets, will be discovered and that some of these will also influence pulmonary immune defenses and inflammatory injury.
Collapse
Affiliation(s)
- Elizabeth A Middleton
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Andrew S Weyrich
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Guy A Zimmerman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
34
|
Visovatti SH, Hyman MC, Goonewardena SN, Anyanwu AC, Kanthi Y, Robichaud P, Wang J, Petrovic-Djergovic D, Rattan R, Burant CF, Pinsky DJ. Purinergic dysregulation in pulmonary hypertension. Am J Physiol Heart Circ Physiol 2016; 311:H286-98. [PMID: 27208163 PMCID: PMC4967198 DOI: 10.1152/ajpheart.00572.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 05/18/2016] [Indexed: 11/22/2022]
Abstract
Despite the fact that nucleotides and adenosine help regulate vascular tone through purinergic signaling pathways, little is known regarding their contributions to the pathobiology of pulmonary arterial hypertension, a condition characterized by elevated pulmonary vascular resistance and remodeling. Even less is known about the potential role that alterations in CD39 (ENTPD1), the ectonucleotidase responsible for the conversion of the nucleotides ATP and ADP to AMP, may play in pulmonary arterial hypertension. In this study we identified decreased CD39 expression on the pulmonary endothelium of patients with idiopathic pulmonary arterial hypertension. We next determined the effects of CD39 gene deletion in mice exposed to normoxia or normobaric hypoxia (10% oxygen). Compared with controls, hypoxic CD39(-/-) mice were found to have a markedly elevated ATP-to-adenosine ratio, higher pulmonary arterial pressures, more right ventricular hypertrophy, more arterial medial hypertrophy, and a pro-thrombotic phenotype. In addition, hypoxic CD39(-/-) mice exhibited a marked increase in lung P2X1 receptors. Systemic reconstitution of ATPase and ADPase enzymatic activities through continuous administration of apyrase decreased pulmonary arterial pressures in hypoxic CD39(-/-) mice to levels found in hypoxic CD39(+/+) controls. Treatment with NF279, a potent and selective P2X1 receptor antagonist, lowered pulmonary arterial pressures even further. Our study is the first to implicate decreased CD39 and resultant alterations in circulating purinergic signaling ligands and cognate receptors in the pathobiology of pulmonary arterial hypertension. Reconstitution and receptor blocking experiments suggest that phosphohydrolysis of purinergic nucleotide tri- and diphosphates, or blocking of the P2X1 receptor could serve as treatment for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Scott H Visovatti
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan;
| | - Matthew C Hyman
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Cardiovascular Medicine, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan; and
| | - Anuli C Anyanwu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Cardiovascular Medicine, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan; and
| | - Patrick Robichaud
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jintao Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Danica Petrovic-Djergovic
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rahul Rattan
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - David J Pinsky
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
35
|
Novitskaya T, Chepurko E, Covarrubias R, Novitskiy S, Ryzhov SV, Feoktistov I, Gumina RJ. Extracellular nucleotide regulation and signaling in cardiac fibrosis. J Mol Cell Cardiol 2016; 93:47-56. [PMID: 26891859 DOI: 10.1016/j.yjmcc.2016.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/03/2016] [Accepted: 02/10/2016] [Indexed: 02/06/2023]
Abstract
Following myocardial infarction, purinergic nucleotides and nucleosides are released via non-specific and specific mechanisms in response to cellular activation, stress, or injury. These extracellular nucleotides are potent mediators of physiologic and pathologic responses, contributing to the inflammatory and fibrotic milieu within the injured myocardium. Via autocrine or paracrine signaling, cell-specific effects occur through differentially expressed purinergic receptors of the P2X, P2Y, and P1 families. Nucleotide activation of the ionotropic (ligand-gated) purine receptors (P2X) and several of the metabotropic (G-protein-coupled) purine receptors (P2Y) or adenosine activation of the P1 receptors can have profound effects on inflammatory cell function, fibroblast function, and cardiomyocyte function. Extracellular nucleotidases that hydrolyze released nucleotides regulate the magnitude and duration of purinergic signaling. While there are numerous studies on the role of the purinergic signaling pathway in cardiovascular disease, the extent to which the purinergic signaling pathway modulates cardiac fibrosis is incompletely understood. Here we provide an overview of the current understanding of how the purinergic signaling pathway modulates cardiac fibroblast function and myocardial fibrosis.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Elena Chepurko
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Roman Covarrubias
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Sergey Novitskiy
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Igor Feoktistov
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Pathology, Immunology and Microbiology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
36
|
Burnstock G. P2X ion channel receptors and inflammation. Purinergic Signal 2016; 12:59-67. [PMID: 26739702 DOI: 10.1007/s11302-015-9493-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/23/2015] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation limits tissue damage in response to pathogens or injury and promotes repair. There are two stages of inflammation, initiation and resolution. P2X receptors are gaining attention in relation to immunology and inflammation. The P2X7 receptor in particular appears to be an essential immunomodulatory receptor, although P2X1 and P2X4 receptors also appear to be involved. ATP released from damaged or infected cells causes inflammation by release of inflammatory cytokines via P2X7 receptors and acts as a danger signal by occupying upregulated P2X receptors on immune cells to increase immune responses. The purinergic involvement in inflammation is being explored for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK. .,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
37
|
Lazar Z, Müllner N, Lucattelli M, Ayata CK, Cicko S, Yegutkin GG, De Cunto G, Müller T, Meyer A, Hossfeld M, Sorichter S, Horvath I, Virchow CJ, Robson SC, Lungarella G, Idzko M. NTPDase1/CD39 and aberrant purinergic signalling in the pathogenesis of COPD. Eur Respir J 2016; 47:254-63. [PMID: 26541524 DOI: 10.1183/13993003.02144-2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 08/05/2015] [Indexed: 01/06/2023]
Abstract
Purinergic receptor activation via extracellular ATP is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). Nucleoside triphosphate diphosphohydrolase-1/CD39 hydrolyses extracellular ATP and modulates P2 receptor signalling.We aimed to investigate the expression and function of CD39 in the pathogenesis of cigarette smoke-induced lung inflammation in patients and preclinical mouse models. CD39 expression and soluble ATPase activity were quantified in sputum and bronchoalveolar lavage fluid (BALF) cells in nonsmokers, smokers and COPD patients or mice with cigarette smoke-induced lung inflammation. In mice, pulmonary ATP and cytokine concentrations, inflammation and emphysema were analysed in the presence or absence of CD39.Following acute cigarette smoke exposure CD39 was upregulated in BALF cells in smokers with further increases in COPD patients. Acute cigarette smoke exposure induced CD39 upregulation in murine lungs and BALF cells, and ATP degradation was accelerated in airway fluids. CD39 inhibition and deficiency led to augmented lung inflammation; treatment with ATPase during cigarette smoke exposure prevented emphysema.Pulmonary CD39 expression and activity are increased in COPD. CD39 deficiency leads to enhanced emphysema in mice, while external administration of a functional CD39 analogue partially rescues the phenotype. The compensatory upregulation of pulmonary CD39 might serve as a protective mechanism in cigarette smoke-induced lung damage.
Collapse
Affiliation(s)
- Zsofia Lazar
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany Dept of Pulmonology, Semmelweis University, Budapest, Hungary These authors contributed equally
| | - Nina Müllner
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany These authors contributed equally
| | - Monica Lucattelli
- Dept of Life Sciences, University of Siena, Siena, Italy These authors contributed equally
| | - Cemil Korcan Ayata
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany These authors contributed equally
| | - Sanja Cicko
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| | | | | | - Tobias Müller
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| | - Anja Meyer
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| | - Madelon Hossfeld
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| | - Stephan Sorichter
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| | - Ildiko Horvath
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | | | - Simon C Robson
- Division of Gastroenterology, Dept of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Marco Idzko
- Dept of Pulmonology, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
38
|
Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Pugliese AM. Purinergic signalling in brain ischemia. Neuropharmacology 2015; 104:105-30. [PMID: 26581499 DOI: 10.1016/j.neuropharm.2015.11.007] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022]
Abstract
Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia a primary damage due to the early massive increase of extracellular glutamate is followed by activation of resident immune cells, i.e microglia, and production or activation of inflammation mediators. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. Extracellular concentrations of ATP and adenosine in the brain increase dramatically during ischemia in concentrations able to stimulate their respective specific P2 and P1 receptors. Both ATP P2 and adenosine P1 receptor subtypes exert important roles in ischemia. Although adenosine exerts a clear neuroprotective effect through A1 receptors during ischemia, the use of selective A1 agonists is hampered by undesirable peripheral effects. Evidence up to now in literature indicate that A2A receptor antagonists provide protection centrally by reducing excitotoxicity, while agonists at A2A (and possibly also A2B) and A3 receptors provide protection by controlling massive infiltration and neuroinflammation in the hours and days after brain ischemia. Among P2X receptors most evidence indicate that P2X7 receptor contribute to the damage induced by the ischemic insult due to intracellular Ca(2+) loading in central cells and facilitation of glutamate release. Antagonism of P2X7 receptors might represent a new treatment to attenuate brain damage and to promote proliferation and maturation of brain immature resident cells that can promote tissue repair following cerebral ischemia. Among P2Y receptors, antagonists of P2Y12 receptors are of value because of their antiplatelet activity and possibly because of additional anti-inflammatory effects. Moreover strategies that modify adenosine or ATP concentrations at injury sites might be of value to limit damage after ischemia. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | - Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alessia Melani
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Irene Fusco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
39
|
Abstract
Danger molecules are the first signals released from dying tissue after stroke. These danger signals bind to receptors on immune cells that will result in their activation and the release of inflammatory and neurotoxic mediators, resulting in amplification of the immune response and subsequent enlargement of the damaged brain volume. The release of danger signals is a central event that leads to a multitude of signals and cascades in the affected and neighbouring tissue, therefore providing a potential target for therapy.
Collapse
|
40
|
Pannexin 1 channels regulate leukocyte emigration through the venous endothelium during acute inflammation. Nat Commun 2015; 6:7965. [PMID: 26242575 PMCID: PMC4824045 DOI: 10.1038/ncomms8965] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 06/30/2015] [Indexed: 12/18/2022] Open
Abstract
Inflammatory cell recruitment to local sites of tissue injury and/or infection is controlled by a plethora of signalling processes influencing cell-to-cell interactions between the vascular endothelial cells (ECs) in post-capillary venules and circulating leukocytes. Recently, ATP-sensitive P2Y purinergic receptors have emerged as downstream regulators of EC activation in vascular inflammation. However, the mechanism(s) regulating cellular ATP release in this response remains elusive. Here we report that the ATP-release channel Pannexin1 (Panx1) opens downstream of EC activation by TNF-α. This process involves activation of type-1 TNF receptors, recruitment of Src family kinases (SFK) and SFK-dependent phosphorylation of Panx1. Using an inducible, EC-specific Panx1 knockout mouse line, we report a previously unidentified role for Panx1 channels in promoting leukocyte adhesion and emigration through the venous wall during acute systemic inflammation, placing Panx1 channels at the centre of cytokine crosstalk with purinergic signalling in the endothelium. Endothelial cell activation by inflammation requires extracellular ATP release. Here the authors show that TNF-α induces Src-family kinase-dependent ATP release by Pannexin1 channels in endothelial cells, and that Pannexin1 is required for leukocyte adhesion and emigration into the inflamed tissue.
Collapse
|
41
|
Feng W, Wang L, Zheng G. Expression and function of P2 receptors in hematopoietic stem and progenitor cells. Stem Cell Investig 2015; 2:14. [PMID: 27358882 DOI: 10.3978/j.issn.2306-9759.2015.07.01] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/28/2015] [Indexed: 12/15/2022]
Abstract
Nucleotides have unambiguously emerged as a family of mediators of intercellular communication, which bind to a class of plasma membrane receptors, P2 receptors, to trigger intercellular signaling. P2 receptors can be further divided into P2X and P2Y subfamilies based on structure and function. Different hematopoietic cells express diverse spectrums of P2 receptors at different levels, including hematopoietic stem and progenitor cells (HSPCs). Extracellular adenosine triphosphate (ATP) exerts different effects on HSPCs, regulating cell proliferation, differentiation, migration, and chemotaxis, release of cytokines or lysosomal constituents, and generation of reactive oxygen or nitrogen species. The relationship between abnormal P2 receptor function and human diseases attracts more and more attention. This review summarizes the expression and function of P2 receptors in HSPCs and the relationship to hematopoietic diseases.
Collapse
Affiliation(s)
- Wenli Feng
- 1 State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China ; 2 Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lina Wang
- 1 State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China ; 2 Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guoguang Zheng
- 1 State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300020, China ; 2 Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
42
|
Kanthi Y, Hyman MC, Liao H, Baek AE, Visovatti SH, Sutton NR, Goonewardena SN, Neral MK, Jo H, Pinsky DJ. Flow-dependent expression of ectonucleotide tri(di)phosphohydrolase-1 and suppression of atherosclerosis. J Clin Invest 2015; 125:3027-36. [PMID: 26121751 DOI: 10.1172/jci79514] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/21/2015] [Indexed: 01/18/2023] Open
Abstract
The ability of cells to detect and respond to nucleotide signals in the local microenvironment is essential for vascular homeostasis. The enzyme ectonucleotide tri(di)phosphohydrolase-1 (ENTPD1, also known as CD39) on the surface of leukocytes and endothelial cells metabolizes locally released, intravascular ATP and ADP, thereby eliminating these prothrombotic and proinflammatory stimuli. Here, we evaluated the contribution of CD39 to atherogenesis in the apolipoprotein E-deficient (ApoE-deficient) mouse model of atherosclerosis. Compared with control ApoE-deficient animals, plaque burden was markedly increased along with circulating markers of platelet activation in Cd39+/-Apoe-/- mice fed a high-fat diet. Plaque analysis revealed stark regionalization of endothelial CD39 expression and function in Apoe-/- mice, with CD39 prominently expressed in atheroprotective, stable flow regions and diminished in atheroprone areas subject to disturbed flow. In mice, disturbed flow as the result of partial carotid artery ligation rapidly suppressed endothelial CD39 expression. Moreover, unidirectional laminar shear stress induced atheroprotective CD39 expression in human endothelial cells. CD39 induction was dependent upon the vascular transcription factor Krüppel-like factor 2 (KLF2) binding near the transcriptional start site of CD39. Together, these data establish CD39 as a regionalized regulator of atherogenesis that is driven by shear stress.
Collapse
|
43
|
Fuentes E, Palomo I. Extracellular ATP metabolism on vascular endothelial cells: A pathway with pro-thrombotic and anti-thrombotic molecules. Vascul Pharmacol 2015; 75:1-6. [PMID: 25989108 DOI: 10.1016/j.vph.2015.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 04/16/2015] [Accepted: 05/09/2015] [Indexed: 12/20/2022]
Abstract
Vascular endothelial contributes to the metabolism and interconversion of extracellular adenine nucleotides via ecto-ATPase/ADPase (CD39) and ecto-5'nucleotidase (CD73) activities. These enzymes collectively dephosphorylate ATP, ADP, and AMP with the production of additional adenosine. In the vascular system, adenine nucleotides (ATP and ADP) and nucleoside adenosine represent an important class of extracellular molecules involved in modulating the processes linked to vascular thrombosis exerting various effects in platelets. Yet, the mechanisms by which the extracellular ATP metabolism in the local environment trigger pro-thrombotic and anti-thrombotic states are yet to be fully elucidated. In this article, the relative contribution of extracellular ATP metabolism in platelet regulation is explored.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| |
Collapse
|
44
|
Calder BW, Matthew Rhett J, Bainbridge H, Fann SA, Gourdie RG, Yost MJ. Inhibition of connexin 43 hemichannel-mediated ATP release attenuates early inflammation during the foreign body response. Tissue Eng Part A 2015; 21:1752-62. [PMID: 25760687 DOI: 10.1089/ten.tea.2014.0651] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In the last 50 years, the use of medical implants has increased dramatically. Failure of implanted devices and biomaterials is a significant source of morbidity and increasing healthcare expenditures. An important cause of implant failure is the host inflammatory response. Recent evidence implicates extracellular ATP as an important inflammatory signaling molecule. A major pathway for release of cytoplasmic ATP into the extracellular space is through connexin hemichannels, which are the unpaired constituents of gap junction intercellular channels. Blockade of hemichannels of the connexin 43 (Cx43) isoform has been shown to reduce inflammation and improve healing. We have developed a Cx43 mimetic peptide (JM2) that targets the microtubule-binding domain of Cx43. The following report investigates the role of the Cx43 microtubule-binding domain in extracellular ATP release by Cx43 hemichannels and how this impacts early inflammatory events of the foreign body reaction. METHODS In vitro Cx43 hemichannel-mediated ATP release by cultured human microvascular endothelial cells subjected to hypocalcemic and normocalcemic conditions was measured after application of JM2 and the known hemichannel blocker, flufenamic acid. A submuscular silicone implant model was used to investigate in vivo ATP signaling during the early foreign body response. Implants were coated with control pluronic vehicle or pluronic carrying JM2, ATP, JM2+ATP, or known hemichannel blockers and harvested at 24 h for analysis. RESULTS JM2 significantly inhibited connexin hemichannel-mediated ATP release from cultured endothelial cells. Importantly, the early inflammatory response to submuscular silicone implants was inhibited by JM2. The reduction in inflammation by JM2 was reversed by the addition of exogenous ATP to the pluronic vehicle. CONCLUSIONS These data indicate that ATP released through Cx43 hemichannels into the vasculature is an important signal driving the early inflammatory response to implanted devices. A vital aspect of this work is that it demonstrates that targeted molecular therapeutics, such as JM2, provide the capacity to regulate inflammation in a clinically relevant system.
Collapse
Affiliation(s)
- Bennett W Calder
- 1Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Joshua Matthew Rhett
- 1Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Bainbridge
- 1Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen A Fann
- 1Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Robert G Gourdie
- 2Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion School of Medicine and Research Institute, Roanoke, Virginia
| | - Michael J Yost
- 1Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
45
|
Yoshida O, Dou L, Kimura S, Yokota S, Isse K, Robson SC, Geller DA, Thomson AW. CD39 deficiency in murine liver allografts promotes inflammatory injury and immune-mediated rejection. Transpl Immunol 2015; 32:76-83. [PMID: 25661084 PMCID: PMC4368493 DOI: 10.1016/j.trim.2015.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/15/2015] [Accepted: 01/26/2015] [Indexed: 01/06/2023]
Abstract
Adenosine triphosphate (ATP), an essential metabolic energy source, is released following cell apoptosis or necrosis. It acts as a damage-associated molecule pattern to stimulate innate immune cells. The ectonucleotidase CD39 regulates immune activation by hydrolysis of extracellular ATP. We have shown previously that CD39 expression by donor livers helps protect syngeneic grafts with extended (24 hr) cold preservation time from ischemia reperfusion injury. Given its immune regulatory properties, we hypothesized that CD39 expression in donor livers might modulate transplant tolerance that occurs following mouse allogeneic liver transplantation (LTx). Livers from C57BL/6 (B6) wild-type (WT) or CD39 KO mice were transplanted into normal C3H recipients with minimal (approximately 1 hr) cold ischemia. Serum alanine aminotransferase levels at day 4 post LTx were significantly higher in animals given CD39KO compared with WT livers. Moreover, IFN-γ production by liver-infiltrating CD8+ T cells at day 4 was significantly higher in CD39KO than in WT grafts. Furthermore, splenic T cells from CD39KO liver recipients exhibited greater proliferative responses to donor alloantigens than those from mice given WT grafts. By contrast, there was a concomitant significant reduction in the frequency of regulatory T cells (Treg) in CD39KO than in WT livers. Whereas WT liver allografts survived > 100 days, no CD39KO grafts survived beyond 40 days (median survival time [MST]: WT: >100 days vs CD39KO: 8 days; p<0.01). In addition, soluble CD39 administration significantly prolonged CD39KO liver allograft survival (MST: 27.5 days). These novel data suggest that CD39 expression in liver allografts modulates tissue injury, inflammation, anti-donor effector T cell responses and Treg infiltration and can suppress transplant rejection.
Collapse
Affiliation(s)
- Osamu Yoshida
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Lei Dou
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Shoko Kimura
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Shinichiro Yokota
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Kumiko Isse
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Simon C Robson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - David A Geller
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh Medical Center, Liver Cancer Center, Pittsburgh, PA, USA.
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
46
|
Cattaneo M. The platelet P2 receptors in inflammation. Hamostaseologie 2015; 35:262-6. [PMID: 25579761 DOI: 10.5482/hamo-14-09-0044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/18/2014] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED In addition to their well characterized and established role in haemostasis and thrombosis, platelets contribute to the pathogenesis of inflammation. Adenine nucleotides are signalling molecules that regulate the function of virtually every cell in the body, by interacting with P2 receptors. Their important role in inflammation is well established. In the last few years, the pro-inflammatory roles of adenine nucleotides interacting with their platelet P2 receptors has emerged. In particular, it was shown that the platelet P2Y12 receptor for ADP significantly contributed to the pro-inflammatory effects of cysteinyl leukotrienes (CysLT) in experimental models of asthma in mice. More importantly, it was recently shown that P2Y12 variants were associated with lung function in a large family-based asthma cohort and that the P2Y12 antagonist prasugrel tended to decrease bronchial hyper-reactivity to mannitol in patients with allergic bronchial asthma in a randomized, placebo controlled trial. CONCLUSION These data strongly suggest that P2Y12 may represent an important pharmacological target for the treatment of patients with allergic bronchial asthma.
Collapse
Affiliation(s)
- M Cattaneo
- Marco Cattaneo, MD, Divisione di Medicina Generale III, Azienda Ospedaliera San Paolo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via di Rudinì, 8, 20142 Milano, Italy, Tel. +39/02 50 32 30-95, Fax -89
| |
Collapse
|
47
|
Abstract
Extracellular nucleotides play a critical role in vascular thrombosis and inflammation. Alterations in purinergic extracellular nucleotide concentrations activate pathways that result in platelet degranulation and aggregation, and endothelial and leukocyte activation and recruitment. CD39, the dominant vascular nucleotidase, hydrolyzes ATP and ADP to provide the substrate for generation of the anti-inflammatory and antithrombotic mediator adenosine. The purinergic signaling system, with CD39 at its center, plays an important role in modulating vascular homeostasis and the response to vascular injury, as seen in clinically relevant diseases such as stroke, ischemia-reperfusion injury, and pulmonary hypertension. A growing body of knowledge of the purinergic signaling pathway implicates CD39 as a critical modulator of vascular thrombosis and inflammation. Therapeutic strategies targeting CD39 offer promising opportunities in the management of vascular thromboinflammatory diseases.
Collapse
|
48
|
Cui M, Ding H, Chen F, Zhao Y, Yang Q, Dong Q. Mdivi-1 Protects Against Ischemic Brain Injury via Elevating Extracellular Adenosine in a cAMP/CREB-CD39-Dependent Manner. Mol Neurobiol 2014; 53:240-253. [PMID: 25428621 DOI: 10.1007/s12035-014-9002-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/12/2014] [Indexed: 01/15/2023]
Abstract
This study aimed to examine whether the neuroprotective effects of Mdivi-1 are attributable to extracellular ATP and adenosine. Mdivi-1 was administered prior to or post middle cerebral artery occlusion (MCAO). The extracellular adenosine was measured by in vivo microdialysis and high-pressure liquid chromatography (HPLC) in MCAO mouse model. Western blot was done to determine the influence of Mdivi-1 on the expression of CD39 and CREB phosphorylation both in vivo and in the cultured astrocytes. Intracellular cAMP and protein kinase A (PKA) activity were detected in primary astrocytes. Results showed that Mdivi-1 significantly reduced infarct volume and neurological scores when administered either prior to or post MCAO. Interestingly, pretreatment with Mdivi-1 resulted in marked increase of extracellular adenosine and concomitant decrease in ATP. The expression of CD39, but not CD73, was upregulated by Mdivi-1, which was associated with the elevated phosphorylated cAMP response element-binding protein (CREB), a transcription factor potentially regulating CD39 expression. In primary astrocytes, Mdivi-1 treatment induced increases in intracellular cAMP, PKA activity and CREB phosphorylation, and PKA-specific inhibitor completely reversed Mdivi-1-induced CD39 expression. Our results demonstrate that Mdivi-1 protects against ischemic brain injury through increasing extracellular adenosine, a process involving elevated CD39 expression that is likely modulated by cAMP/PKA/CREB cascade. Figure Potential mechanisms by which Mdivi-1 mediates the neuroprotection on cerebral ischemic stroke. Results from the present study indicate that Mdivi-1 protects against ischemic brain injury through increasing extracellular adenosine, a process involving elevated CD39 expression that is likely modulated by the cAMP/PKA/CREB cascades.
Collapse
Affiliation(s)
- Mei Cui
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| | - Hongyan Ding
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Fangzhe Chen
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Yanxin Zhao
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| | - Qi Yang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| |
Collapse
|
49
|
Umansky V, Shevchenko I, Bazhin AV, Utikal J. Extracellular adenosine metabolism in immune cells in melanoma. Cancer Immunol Immunother 2014; 63:1073-80. [PMID: 24756420 PMCID: PMC11029545 DOI: 10.1007/s00262-014-1553-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/11/2014] [Indexed: 01/04/2023]
Abstract
Malignant melanoma is characterized by the development of chronic inflammation in the tumor microenvironment, which leads to a strong immunosuppression associated with a rapid tumor progression. Adenosine is considered as one of the main immunosuppressive factors in the tumor environment. It is produced via enzymatic hydrolysis of extracellular ATP by ectonucleotidases CD39 and CD73 localized on cell surface. Using the ret transgenic mouse melanoma model that closely mimics human melanoma, we demonstrated an increased frequency of ectonucleotidase-positive myeloid-derived suppressor cells (MDSCs) in melanoma lesions and lymphoid organs. Furthermore, we observed that conventional CD4(+)FoxP3(-) and CD8(+) T cells infiltrating melanoma lesions of ret transgenic mice were distinctly enriched in the CD39(+)CD73(+) subpopulation that co-expressed also PD-1. Ectonucleotidase expression was also up-regulated in CD4(+) and CD8(+) T cells upon activation. In addition, these ectoenzymes were largely found to be expressed on memory T cell compartment (in particular, on effector memory cells). Our data suggest that extracellular adenosine produced by regulatory T cells (Tregs) and MDSCs can suppress T cell effector functions through paracrine signaling. Another mechanism involves its production also by effector T cells and an inhibition of their anti-tumor reactivity via autocrine signaling as a part of the negative feedback loop. This mode of adenosine signaling could be also used by Tregs and MDSCs to enhance their immunosuppressive activity.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany,
| | | | | | | |
Collapse
|
50
|
Sullivan JA, Jankowska-Gan E, Shi L, Roenneburg D, Hegde S, Greenspan DS, Wilkes DS, Denlinger LC, Burlingham WJ. Differential requirement for P2X7R function in IL-17 dependent vs. IL-17 independent cellular immune responses. Am J Transplant 2014; 14:1512-22. [PMID: 24866539 PMCID: PMC4295495 DOI: 10.1111/ajt.12741] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/24/2014] [Accepted: 03/10/2014] [Indexed: 01/25/2023]
Abstract
IL17-dependent autoimmunity to collagen type V (Col V) has been associated with lung transplant obliterative bronchiolitis. Unlike the T helper 1 (Th1)-dependent immune responses to Tetanus Toxoid (TT), the Th17 response to Col V in lung transplant patients and its Th1/17 variant observed in coronary artery disease patients requires IL-1β, tumor necrosis factor α and CD14(+) cells. Given the involvement of the P2X7 receptor (P2X7R) in monocyte IL-1β responses, we investigated its role in Th17-, Th1/17- and Th1-mediated proinflammatory responses. Transfer of antigen-pulsed peripheral blood mononucleated cells (PBMCs) from Col V-reactive patients into SCID mouse footpads along with P2X7R antagonists revealed a selective inhibition of Col V-, but not TT-specific swelling responses. P2X7R inhibitors blocked IL-1β induction from monocytes, including both Col V-α1 peptide-induced (T-dependent), as well as native Col V-induced (T-independent) responses. Significantly higher P2X7R expression was found on CXCR3(neg) CCR4(+)/6(+) CD4(+) [Th17] versus CXCR3(+)CCR4/6(neg) CD4(+) [Th1] subsets in PBMCs, suggesting that the paradigm of selective dependence on P2X7R might extend beyond Col V autoimmunity. Indeed, P2X7R inhibitors suppressed not only anti-Col V, but also Th1/17-mediated alloimmunity, in a heart transplant patient without affecting anti-viral Epstein-Barr virus responses. These results suggest that agents targeting the P2X7R might effectively treat Th17-related transplant pathologies, while maintaining Th1-immunity to infection.
Collapse
Affiliation(s)
- JA Sullivan
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - E Jankowska-Gan
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - L Shi
- Department of Medicine, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - D Roenneburg
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | | | - DS Greenspan
- Department of Cell & Regenerative Biology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - DS Wilkes
- Department of Medicine, University of Indiana, 340 W 10th St Suite 6200 Indianapolis, IN 46202
| | - LC Denlinger
- Department of Medicine, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - WJ Burlingham
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792,To whom correspondence should be addressed: 600 Highland Avenue, Room G4/702, Madison, WI 53792. Tel: (608) 263-0119 Fax: (608) 262-6280
| |
Collapse
|