1
|
Liu Q, Niu Y, Pei Z, Yang Y, Xie Y, Wang M, Wang J, Wu M, Zheng J, Yang P, Hao H, Pang Y, Bao L, Dai Y, Niu Y, Zhang R. Gas6-Axl signal promotes indoor VOCs exposure-induced pulmonary fibrosis via pulmonary microvascular endothelial cells-fibroblasts cross-talk. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134786. [PMID: 38824778 DOI: 10.1016/j.jhazmat.2024.134786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Volatile organic compounds (VOCs) as environmental pollutants were associated with respiratory diseases. Pulmonary fibrosis (PF) was characterized by an increase of extracellular matrix, leading to deterioration of lung function. The adverse effects on lung and the potential mechanism underlying VOCs induced PF had not been elucidated clearly. In this study, the indoor VOCs exposure mouse model along with an ex vivo biosensor assay was established. Based on scRNA-seq analysis, the adverse effects on lung and potential molecular mechanism were studied. Herein, the results showed that VOCs exposure from indoor decoration contributed to decreased lung function and facilitated pulmonary fibrosis in mice. Then, the whole lung cell atlas after VOCs exposure and the heterogeneity of fibroblasts were revealed. We explored the molecular interactions among various pulmonary cells, suggesting that endothelial cells contributed to fibroblasts activation in response to VOCs exposure. Mechanistically, pulmonary microvascular endothelial cells (MPVECs) secreted Gas6 after VOCs-induced PANoptosis phenotype, bound to the Axl in fibroblasts, and then activated fibroblasts. Moreover, Atf3 as the key gene negatively regulated PANoptosis phenotype to ameliorate fibrosis induced by VOCs exposure. These novel findings provided a new perspective about MPVECs could serve as the initiating factor of PF induced by VOCs exposure.
Collapse
Affiliation(s)
- Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Zijie Pei
- Department of Thoracic Surgery, the 2nd Hospital of Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yizhe Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yujia Xie
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Jingyuan Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Jie Zheng
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Peihao Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Haiyan Hao
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Province Center for Disease Control and Prevention, Shijiazhuang 050021, Hebei, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Lei Bao
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yufei Dai
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yujie Niu
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China.
| |
Collapse
|
2
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
3
|
Kandouz M. Cell Death, by Any Other Name…. Cells 2024; 13:325. [PMID: 38391938 PMCID: PMC10886887 DOI: 10.3390/cells13040325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Studies trying to understand cell death, this ultimate biological process, can be traced back to a century ago. Yet, unlike many other fashionable research interests, research on cell death is more alive than ever. New modes of cell death are discovered in specific contexts, as are new molecular pathways. But what is "cell death", really? This question has not found a definitive answer yet. Nevertheless, part of the answer is irreversibility, whereby cells can no longer recover from stress or injury. Here, we identify the most distinctive features of different modes of cell death, focusing on the executive final stages. In addition to the final stages, these modes can differ in their triggering stimulus, thus referring to the initial stages. Within this framework, we use a few illustrative examples to examine how intercellular communication factors in the demise of cells. First, we discuss the interplay between cell-cell communication and cell death during a few steps in the early development of multicellular organisms. Next, we will discuss this interplay in a fully developed and functional tissue, the gut, which is among the most rapidly renewing tissues in the body and, therefore, makes extensive use of cell death. Furthermore, we will discuss how the balance between cell death and communication is modified during a pathological condition, i.e., colon tumorigenesis, and how it could shed light on resistance to cancer therapy. Finally, we briefly review data on the role of cell-cell communication modes in the propagation of cell death signals and how this has been considered as a potential therapeutic approach. Far from vainly trying to provide a comprehensive review, we launch an invitation to ponder over the significance of cell death diversity and how it provides multiple opportunities for the contribution of various modes of intercellular communication.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Department of Pathology, School of Medicine, Wayne State University, 540 East Canfield Avenue, Detroit, MI 48201, USA;
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
4
|
Urban MW, Charsar BA, Heinsinger NM, Markandaiah SS, Sprimont L, Zhou W, Brown EV, Henderson NT, Thomas SJ, Ghosh B, Cain RE, Trotti D, Pasinelli P, Wright MC, Dalva MB, Lepore AC. EphrinB2 knockdown in cervical spinal cord preserves diaphragm innervation in a mutant SOD1 mouse model of ALS. eLife 2024; 12:RP89298. [PMID: 38224498 PMCID: PMC10945582 DOI: 10.7554/elife.89298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motor neuron loss. Importantly, non-neuronal cell types such as astrocytes also play significant roles in disease pathogenesis. However, mechanisms of astrocyte contribution to ALS remain incompletely understood. Astrocyte involvement suggests that transcellular signaling may play a role in disease. We examined contribution of transmembrane signaling molecule ephrinB2 to ALS pathogenesis, in particular its role in driving motor neuron damage by spinal cord astrocytes. In symptomatic SOD1G93A mice (a well-established ALS model), ephrinB2 expression was dramatically increased in ventral horn astrocytes. Reducing ephrinB2 in the cervical spinal cord ventral horn via viral-mediated shRNA delivery reduced motor neuron loss and preserved respiratory function by maintaining phrenic motor neuron innervation of diaphragm. EphrinB2 expression was also elevated in human ALS spinal cord. These findings implicate ephrinB2 upregulation as both a transcellular signaling mechanism in mutant SOD1-associated ALS and a promising therapeutic target.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Brittany A Charsar
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Nicolette M Heinsinger
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Shashirekha S Markandaiah
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Lindsay Sprimont
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Wei Zhou
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Eric V Brown
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Nathan T Henderson
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Samantha J Thomas
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Biswarup Ghosh
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Rachel E Cain
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Megan C Wright
- Department of Biology, Arcadia UniversityGlensideUnited States
| | - Matthew B Dalva
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
- Department of Cell and Molecular Biology, Tulane Brain Institute, Tulane UniversityNew OrleansUnited States
| | - Angelo C Lepore
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
5
|
Urban MW, Charsar BA, Heinsinger NM, Markandaiah SS, Sprimont L, Zhou W, Brown EV, Henderson NT, Thomas SJ, Ghosh B, Cain RE, Trotti D, Pasinelli P, Wright MC, Dalva MB, Lepore AC. EphrinB2 knockdown in cervical spinal cord preserves diaphragm innervation in a mutant SOD1 mouse model of ALS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.538887. [PMID: 37215009 PMCID: PMC10197713 DOI: 10.1101/2023.05.10.538887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motor neuron loss. Importantly, non-neuronal cell types such as astrocytes also play significant roles in disease pathogenesis. However, mechanisms of astrocyte contribution to ALS remain incompletely understood. Astrocyte involvement suggests that transcellular signaling may play a role in disease. We examined contribution of transmembrane signaling molecule ephrinB2 to ALS pathogenesis, in particular its role in driving motor neuron damage by spinal cord astrocytes. In symptomatic SOD1-G93A mice (a well-established ALS model), ephrinB2 expression was dramatically increased in ventral horn astrocytes. Reducing ephrinB2 in the cervical spinal cord ventral horn via viral-mediated shRNA delivery reduced motor neuron loss and preserved respiratory function by maintaining phrenic motor neuron innervation of diaphragm. EphrinB2 expression was also elevated in human ALS spinal cord. These findings implicate ephrinB2 upregulation as both a transcellular signaling mechanism in mutant SOD1-associated ALS and a promising therapeutic target.
Collapse
|
6
|
Verma M, Chopra M, Kumar H. Unraveling the Potential of EphA4: A Breakthrough Target and Beacon of Hope for Neurological Diseases. Cell Mol Neurobiol 2023; 43:3375-3391. [PMID: 37477786 DOI: 10.1007/s10571-023-01390-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
Erythropoietin-producing hepatocellular carcinoma A4 (EphA4) is a transmembrane receptor protein which is a part of the most prominent family of receptor tyrosine kinases (RTKs). It serves a crucial role in both physiological, biological, and functional states binding with their ligand like Ephrins. Its abundance in the majority of the body's systems has been reported. Moreover, it draws much attention in the CNS since it influences axonal and vascular guidance. Also, it has a widespread role at the pathological state of various CNS disorders. Reports suggest it obstructs axonal regeneration in various neurodegenerative diseases and neurological disorders. Although, neuro-regeneration is still an open challenge to the modern drug discovery community. Hence, in this review, we will provide information about the role of EphA4 in neurological diseases by which it may emerge as a therapeutic target for CNS disease. We will also provide a glance at numerous signaling pathways that activate or inhibit the EphA4-associated biological processes contributing to the course of neurodegenerative diseases. Thus, this work might serve as a basis for futuristic studies that are related to the target-based drug discovery in the field of neuro-regeneration. Pathological and physiological events associated with EphA4 and Ephrin upregulation and interaction.
Collapse
Affiliation(s)
- Meenal Verma
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Manjeet Chopra
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Hemant Kumar
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
7
|
García-Ceca J, Montero-Herradón S, González A, Plaza R, Zapata AG. Altered thymocyte development observed in EphA4-deficient mice courses with changes in both thymic epithelial and extracellular matrix organization. Cell Mol Life Sci 2022; 79:583. [PMID: 36334147 PMCID: PMC9637064 DOI: 10.1007/s00018-022-04610-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022]
Abstract
Eph receptors and their ligands, Ephrins, are involved in the thymocyte-thymic epithelial cell (TEC) interactions, key for the functional maturation of both thymocytes and thymic epithelium. Several years ago, we reported that the lack of EphA4, a Eph of the subfamily A, coursed with reduced proportions of double positive (DP) thymocytes apparently due to an altered thymic epithelial stroma [Munoz et al. in J Immunol 177:804–813, 2006]. In the present study, we reevaluate the lymphoid, epithelial, and extracellular matrix (ECM) phenotype of EphA4−/− mice grouped into three categories with respect to their proportions of DP thymocytes. Our results demonstrate a profound hypocellularity, specific alterations of T cell differentiation that affected not only DP thymocytes, but also double negative and single positive T cell subsets, as well as the proportions of positively and negatively selected thymocytes. In correlation, thymic histological organization changed markedly, especially in the cortex, as well as the proportions of both Ly51+UEA-1− cortical TECs and Ly51−UEA-1+ medullary TECs. The alterations observed in the expression of ECM components (Fibronectin, Laminin, Collagen IV), integrin receptors (VLA-4, VLA-6), chemokines (CXCL12, CCL25, CCL21) and their receptors (CXCR4, CCR7, CCR9) and in vitro transwell assays on the capacity of migration of WT and mutant thymocytes suggest that the lack of EphA4 alters T-cell differentiation by presumably affecting cell adhesion between TECs and T-TEC interactions rather than by thymocyte migration.
Collapse
Affiliation(s)
- Javier García-Ceca
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040, Madrid, Spain.,Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Sara Montero-Herradón
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040, Madrid, Spain.,Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Ana González
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040, Madrid, Spain
| | - Rosa Plaza
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040, Madrid, Spain. .,Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain.
| |
Collapse
|
8
|
Ng XW, Chung YH, Piston DW. Intercellular Communication in the Islet of Langerhans in Health and Disease. Compr Physiol 2021; 11:2191-2225. [PMID: 34190340 PMCID: PMC8985231 DOI: 10.1002/cphy.c200026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Blood glucose homeostasis requires proper function of pancreatic islets, which secrete insulin, glucagon, and somatostatin from the β-, α-, and δ-cells, respectively. Each islet cell type is equipped with intrinsic mechanisms for glucose sensing and secretory actions, but these intrinsic mechanisms alone cannot explain the observed secretory profiles from intact islets. Regulation of secretion involves interconnected mechanisms among and between islet cell types. Islet cells lose their normal functional signatures and secretory behaviors upon dispersal as compared to intact islets and in vivo. In dispersed islet cells, the glucose response of insulin secretion is attenuated from that seen from whole islets, coordinated oscillations in membrane potential and intracellular Ca2+ activity, as well as the two-phase insulin secretion profile, are missing, and glucagon secretion displays higher basal secretion profile and a reverse glucose-dependent response from that of intact islets. These observations highlight the critical roles of intercellular communication within the pancreatic islet, and how these communication pathways are crucial for proper hormonal and nonhormonal secretion and glucose homeostasis. Further, misregulated secretions of islet secretory products that arise from defective intercellular islet communication are implicated in diabetes. Intercellular communication within the islet environment comprises multiple mechanisms, including electrical synapses from gap junctional coupling, paracrine interactions among neighboring cells, and direct cell-to-cell contacts in the form of juxtacrine signaling. In this article, we describe the various mechanisms that contribute to proper islet function for each islet cell type and how intercellular islet communications are coordinated among the same and different islet cell types. © 2021 American Physiological Society. Compr Physiol 11:2191-2225, 2021.
Collapse
Affiliation(s)
- Xue W Ng
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - Yong H Chung
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| |
Collapse
|
9
|
Li J, Ma N, Chen J, Yan D, Zhang Q, Shi J. EphA4 receptor regulates outwardly rectifying chloride channel in CA1 hippocampal neurons after ischemia-reperfusion. Neuroreport 2019; 30:980-984. [PMID: 31469726 PMCID: PMC6735946 DOI: 10.1097/wnr.0000000000001311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022]
Abstract
CA1 hippocampal neurons are sensitive to ischemia. The erythropoietin-producing hepatocellular carcinoma (Eph) receptors are a cell-cell contact signaling pathway for regulating neuron function and death. However, the mechanisms of EphA receptor in neuron death after ischemia remain unclear. In this study, we present evidence that outwardly rectifying chloride channels reside in CA1 hippocampal neurons. EphA4 receptor increased chloride channel currents. Moreover, the EphA4 receptor no longer had significant effects on enhanced channel currents following ischemia-reperfusion. Inhibition of EphA4 receptor with EphA4-Fc significantly decreased the channel currents after ischemia-reperfusion. These results suggest that the increased effect of the EphA4 receptor on the outwardly rectifying chloride channel activity in CA1 hippocampal neurons may provide better treatment for ischemic brain injury.
Collapse
Affiliation(s)
- Jianguo Li
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Na Ma
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jing Chen
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Deping Yan
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Qian Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jinchao Shi
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
10
|
Teng S, Palmieri A, Maita I, Zheng C, Das G, Park J, Zhou R, Alder J, Thakker-Varia S. Inhibition of EphA/Ephrin-A signaling using genetic and pharmacologic approaches improves recovery following traumatic brain injury in mice. Brain Inj 2019; 33:1385-1401. [DOI: 10.1080/02699052.2019.1641622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Shavonne Teng
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Alicia Palmieri
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Isabella Maita
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Cynthia Zheng
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Gitanjali Das
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Juyeon Park
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Renping Zhou
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Janet Alder
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Smita Thakker-Varia
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
11
|
García-Aranda M, Redondo M. Targeting Protein Kinases to Enhance the Response to anti-PD-1/PD-L1 Immunotherapy. Int J Mol Sci 2019; 20:E2296. [PMID: 31075880 PMCID: PMC6540309 DOI: 10.3390/ijms20092296] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022] Open
Abstract
The interaction between programmed cell death protein (PD-1) and its ligand (PD-L1) is one of the main pathways used by some tumors to escape the immune response. In recent years, immunotherapies based on the use of antibodies against PD-1/PD-L1 have been postulated as a great promise for cancer treatment, increasing total survival compared to standard therapy in different tumors. Despite the hopefulness of these results, a significant percentage of patients do not respond to such therapy or will end up evolving toward a progressive disease. Besides their role in PD-L1 expression, altered protein kinases in tumor cells can limit the effectiveness of PD-1/PD-L1 blocking therapies at different levels. In this review, we describe the role of kinases that appear most frequently altered in tumor cells and that can be an impediment for the success of immunotherapies as well as the potential utility of protein kinase inhibitors to enhance the response to such treatments.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Research Unit, Hospital Costa del Sol. Autovía A7, km 187. Marbella, 29603 Málaga, Spain.
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), 28029 Madrid, Spain.
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain.
| | - Maximino Redondo
- Research Unit, Hospital Costa del Sol. Autovía A7, km 187. Marbella, 29603 Málaga, Spain.
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), 28029 Madrid, Spain.
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain.
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Universidad de Málaga, Campus Universitario de Teatinos, 29010 Málaga, Spain.
| |
Collapse
|