1
|
Xiong W, Tang J, Yu H, Luo Y, Yu M, Li Y. Emodin inhibits M1 macrophage activation that related to acute and chronic kidney injury through EGFR/MAPK pathway. Funct Integr Genomics 2024; 24:131. [PMID: 39078513 DOI: 10.1007/s10142-024-01407-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Macrophages are the main inflammatory cells involved in kidney injury and play a significant role in the development of acute kidney injury (AKI) and progression of chronic kidney disease (CKD). Emodin is believed to stabilize macrophage homeostasis under pathological conditions. The objective of this study aimed to explore the underlying mechanisms and effects of Emodin on M1 macrophages. METHODS Network pharmacology methods were used to predict target proteins associated with renal injury and identify the pathways affected by emodin. RAW264.7 macrophages were induced into M1 polarization using LPS and then treated with emodin at 20, 40, and 80 µM. The effects of emodin on cell viability, cytokines (IL-1β, IL-6, TNF-α), M1 macrophage markers (F4/80 + CD86+), and the EGFR/MAPK pathway were evaluated. Additionally, we transfected RAW264.7 cells with an EGFR shRNA interference lentivirus to assess its effects on RAW264.7 cells function and MAPK pathway. After RAW264.7 cells were passaged to expanded culture and transfected with EGFR-interfering plasmid, macrophages were induced to polarize towards M1 with LPS and then treated with 80 µM emodin. CKD modeling was performed to test how emodin is regulated during CKD. RESULTS There are 15 common targets between emodin and kidney injury, of which the EGFR/MAPK pathway is the pathway through which emodin affects macrophage function. Emodin significantly reduced the levels of IL-6, IL-1β and TNF-α (p < 0.05) and the ratio of M1 macrophage surface markers F4/80 + CD86+ (p < 0.01) in the supernatant of RAW264.7 cells in a dose-dependent manner. Furthermore, the inhibitory effect of emodin on RAW264.7 cells was achieved by interfering with the EGFR/MAPK pathway. Moreover, emodin also affected the mRNA and protein expression of EGFR and Ras, leading to a decrease in the rate of M1 macrophages, thus inhibiting the pro-inflammatory effect of M1 macrophages. The addition of emodin reduced the rate of M1 macrophages in CKD and inhibited the further polarization of M1 macrophages, thus maintaining the pro-inflammatory and anti-inflammatory homeostasis in CKD, and these effects were achieved by emodin through the control of the EGRF/ERK pathway. CONCLUSION Emodin attenuates M1 macrophage polarization and pro-inflammatory responses via the EGFR/MAPK signalling pathway. And the addition of emodin maintains pro- and anti-inflammatory homeostasis, which is important for maintaining organ function and tissue repair.
Collapse
Affiliation(s)
- Weijian Xiong
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Jing Tang
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Hangxing Yu
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Yan Luo
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Minghuan Yu
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Ying Li
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China.
| |
Collapse
|
2
|
EGFR-TNFR1 pathway in endothelial cell facilitates acute lung injury by NF-κB/MAPK-mediated inflammation and RIP3-dependent necroptosis. Int Immunopharmacol 2023; 117:109902. [PMID: 36827922 DOI: 10.1016/j.intimp.2023.109902] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
Tumor necrosis factor-α (TNFα) has emerged as a pivotal effector critically correlated with disease severity in acute lung injury (ALI). Because both the excessive activation of epidermal growth factor receptor (EGFR) and tumor necrosis factor receptor 1 (TNFR1) in sepsis-induced vasculitis are markedly diminished through EGFR tyrosine kinase inhibitor, a specific mechanism must exist to modulate TNFR1 cellular fates regulated by EGFR. Here, we demonstrated that EGFR, a specific binding partner of TNFR1, exhibited an increased NF-κB/MAPK-mediated inflammation that was governed by enhanced recruitment of TNFR-associated factor 2 (TRAF2) to TNFR1 complex I in endothelial cell (EC). Moreover, EGFR activation triggered a remarkable increase in the phosphorylation of receptor-interacting protein 1 (RIP1) and its binding with receptor-interacting protein 3 (RIP3) which led to enhanced frequency of necroptosis in complex IIb. Inhibiting the kinase of EGFR disrupted the formation of complex I and complex IIb and prevents EC from NF-κB/MAPK-mediated inflammation and RIP3-dependent necroptosis. Consistently, pharmacological inhibition of EGFR can limit the destructive effects of neutrophils activation and the hyperpermeability of lung vascular in hyperinflammation period. Collectively, we have identified EC-EGFR as a modulator of TNFR1-mediated inflammation and RIP3-dependent necroptosis, providing a possible explanation for the immunological basis of anti-EGFR therapy in sepsis-induced ALI.
Collapse
|
3
|
Zhang X, Chen C, Ling C, Luo S, Xiong Z, Liu X, Liao C, Xie P, Liu Y, Zhang L, Chen Z, Liu Z, Tang J. EGFR tyrosine kinase activity and Rab GTPases coordinate EGFR trafficking to regulate macrophage activation in sepsis. Cell Death Dis 2022; 13:934. [PMID: 36344490 PMCID: PMC9640671 DOI: 10.1038/s41419-022-05370-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
EGFR phosphorylation is required for TLR4-mediated macrophage activation during sepsis. However, whether and how intracellular EGFR is transported during endotoxemia have largely been unknown. Here, we show that LPS promotes high levels cell surface expression of EGFR in macrophages through two different transport mechanisms. On one hand, Rab10 is required for EEA1-mediated the membrane translocation of EGFR from the Golgi. On the other hand, EGFR phosphorylation prevents its endocytosis in a kinase activity-dependent manner. Erlotinib, an EGFR tyrosine kinase inhibitor, significantly reduced membrane EGFR expression in LPS-activated macrophage. Mechanistically, upon LPS induced TLR4/EGFR phosphorylation, MAPK14 phosphorylated Rab7a at S72 impaired membrane receptor late endocytosis, which maintains EGFR membrane localization though blocking its lysosomal degradation. Meanwhile, Rab5a is also involved in the early endocytosis of EGFR. Subsequently, inhibition of EGFR phosphorylation switches M1 phenotype to M2 phenotype and alleviates sepsis-induced acute lung injury. Mechanistic study demonstrated that Erlotinib suppressed glycolysis-dependent M1 polarization via PKM2/HIF-1ɑ pathway and promoted M2 polarization through up-regulating PPARγ induced glutamine metabolism. Collectively, our data elucidated a more in-depth mechanism of macrophages activation, and provided stronger evidence supporting EGFR as a potential therapeutic target for the treatment of sepsis.
Collapse
Affiliation(s)
- Xuedi Zhang
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Cuiping Chen
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Chunxiu Ling
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Shuhua Luo
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Ziying Xiong
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Xiaolei Liu
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Chaoxiong Liao
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China ,grid.410560.60000 0004 1760 3078Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Pengyun Xie
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Youtan Liu
- grid.284723.80000 0000 8877 7471The Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000 Guangdong China
| | - Liangqing Zhang
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| | - Zhanghui Chen
- Department of Hematology, Zhanjiang Institute of Clinical Medicine, Zhanjiang Central Hospital, 524000 Zhanjiang, China
| | - Zhifeng Liu
- The Department of Critical Care Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou, 510010 Guangdong China
| | - Jing Tang
- grid.410560.60000 0004 1760 3078The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000 Guangdong China
| |
Collapse
|
4
|
Huang L, Zhang X, Fan J, Liu X, Luo S, Cao D, Liu Y, Xia Z, Zhong H, Chen C, Zhang L, Liu Z, Tang J. EGFR promotes the apoptosis of CD4 + T lymphocytes through TBK1/Glut1 induced Warburg effect in sepsis. J Adv Res 2022; 44:39-51. [PMID: 35618635 PMCID: PMC9936423 DOI: 10.1016/j.jare.2022.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/17/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Sepsis-induced apoptosis leads to lymphopenia including the decrease of CD4+ T cells thus favoring immunosuppression. OBJECTIVES Although epidermal growth factor receptor (EGFR) inhibitors significantly improve the survival rate of septic mice, the effect of EGFR on the function and metabolism of CD4+ T cells in sepsis remained unknown. METHODS CD4+ T cells from septic mice and patients were assessed for apoptosis, activation, Warburg metabolism and glucose transporter 1 (Glut1) expression with or without the interference of EGFR activation. RESULTS EGFR facilitates CD4+ T cell activation and apoptosis through Glut1, which is a key enzyme that controls glycolysis in T cells. EGFR, TANK binding kinase 1 (TBK1) and Glut1 form a complex to facilitate Glut1 transportation from cytoplasm to cell surface. Both the levels of membrane expression of EGFR and Glut1 and the activation levels of CD4+ T cells were significantly higher in patients with sepsis as compared with healthy subjects. CONCLUSION Our data demonstrated that through its downstream TBK1/Exo84/RalA protein system, EGFR regulates Glut1 transporting to the cell surface, which is a key step for inducing the Warburg effect and the subsequent cellular activation and apoptosis of CD4+ T lymphocytes and may eventually affect the immune functional status, causing immune cell exhaustion in sepsis.
Collapse
Affiliation(s)
- Li Huang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xuedi Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junyu Fan
- The Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shuhua Luo
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Dianqing Cao
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Youtan Liu
- The Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhengyuan Xia
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Hanhui Zhong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cuiping Chen
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Liangqing Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhifeng Liu
- The Department of Critical Care Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China.
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
5
|
Liu ZN, Su QQ, Wang YH, Wu X, Lv XW. Blockade of the P2Y2 Receptor Attenuates Alcoholic Liver Inflammation by Targeting the EGFR-ERK1/2 Signaling Pathway. Drug Des Devel Ther 2022; 16:1107-1120. [PMID: 35444406 PMCID: PMC9013714 DOI: 10.2147/dddt.s346376] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/01/2022] [Indexed: 01/12/2023] Open
Affiliation(s)
- Zhen-Ni Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People’s Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, People’s Republic of China
| | - Qian-Qian Su
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Yu-Hui Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People’s Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, People’s Republic of China
| | - Xue Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People’s Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, People’s Republic of China
| | - Xiong-Wen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, People’s Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, People’s Republic of China
- Correspondence: Xiong-Wen Lv, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province, 230032, People’s Republic of China, Email
| |
Collapse
|
6
|
Xue Q, Liu X, Chen C, Zhang X, Xie P, Liu Y, Zhou S, Tang J. Erlotinib protests against LPS-induced parthanatos through inhibiting macrophage surface TLR4 expression. Cell Death Discov 2021; 7:181. [PMID: 34282120 PMCID: PMC8290014 DOI: 10.1038/s41420-021-00571-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/05/2021] [Accepted: 07/03/2021] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a life-threatening cascading systemic inflammatory response syndrome on account of serve infection. In inflamed tissues, activated macrophages generate large amounts of inflammatory cytokines reactive species, and are exposed to the damaging effects of reactive species. However, comparing with necroptosis and pyroptosis, so far, there are few studies focusing on the overproduction-related cell death, such as parthanatos in macrophage during sepsis. In LPS-treated macrophage, we observed PARP-1 activation, PAR formation and AIF translocation. All these phenomena could be inhibited by both erlotinib and 3-AB, indicating the presence of parthanatos in endotoxemia. We further found that LPS induced the increase of cell surface TLR4 expression responsible for the production of ROS and subsequent parthanatos in endotoxemia. All these results shed a new light on how TLR4 regulating the activation of PARP-1 by LPS in macrophage.
Collapse
Affiliation(s)
- Qiong Xue
- The Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cuiping Chen
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xuedi Zhang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Pengyun Xie
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yupin Liu
- Department of Medical Iconography, The Second Affiliated Hospital University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Shuangnan Zhou
- Department of Liver Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
7
|
Massip-Copiz MM, Valdivieso ÁG, Clauzure M, Mori C, Asensio CJA, Aguilar MÁ, Santa-Coloma TA. Epidermal growth factor receptor activity upregulates lactate dehydrogenase A expression, lactate dehydrogenase activity, and lactate secretion in cultured IB3-1 cystic fibrosis lung epithelial cells. Biochem Cell Biol 2021; 99:476-487. [PMID: 33481676 DOI: 10.1139/bcb-2020-0522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. It has been postulated that reduced HCO3- transport through CFTR may lead to a decreased airway surface liquid pH. In contrast, others have reported no changes in the extracellular pH (pHe). We have recently reported that in carcinoma Caco-2/pRS26 cells (transfected with short hairpin RNA for CFTR) or CF lung epithelial IB3-1 cells, the mutation in CFTR decreased mitochondrial complex I activity and increased lactic acid production, owing to an autocrine IL-1β loop. The secreted lactate accounted for the reduced pHe, because oxamate fully restored the pHe. These effects were attributed to the IL-1β autocrine loop and the downstream signaling kinases c-Src and JNK. Here we show that the pHe of IB3-1 cells can be restored to normal values (∼7.4) by incubation with the epidermal growth factor receptor (EGFR, HER1, ErbB1) inhibitors AG1478 and PD168393. PD168393 fully restored the pHe values of IB3-1 cells, suggesting that the reduced pHe is mainly due to increased EGFR activity and lactate. Also, in IB3-1 cells, lactate dehydrogenase A mRNA, protein expression, and activity are downregulated when EGFR is inhibited. Thus, a constitutive EGFR activation seems to be responsible for the reduced pHe in IB3-1 cells.
Collapse
Affiliation(s)
- María Macarena Massip-Copiz
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Ángel G Valdivieso
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Mariángeles Clauzure
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Consuelo Mori
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Cristian J A Asensio
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - María Á Aguilar
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Tomás A Santa-Coloma
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina.,Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research, School of Medical Sciences, Pontifical Catholic University of Argentina, and the Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| |
Collapse
|
8
|
Denning NL, Aziz M, Diao L, Prince JM, Wang P. Targeting the eCIRP/TREM-1 interaction with a small molecule inhibitor improves cardiac dysfunction in neonatal sepsis. Mol Med 2020; 26:121. [PMID: 33276725 PMCID: PMC7716442 DOI: 10.1186/s10020-020-00243-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal sepsis and the associated myocardial dysfunction remain a leading cause of infant mortality. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a ligand of triggering receptor expressed on myeloid cells-1 (TREM-1). M3 is a small CIRP-derived peptide that inhibits the eCIRP/TREM-1 interaction. We hypothesize that the eCIRP/TREM-1 interaction in cardiomyocytes contributes to sepsis-induced cardiac dysfunction in neonatal sepsis, while M3 is cardioprotective. Methods Serum was collected from neonates in the Neonatal Intensive Care Unit (NICU). 5–7-day old C57BL/6 mouse pups were used in this study. Primary murine neonatal cardiomyocytes were stimulated with recombinant murine (rm) CIRP with M3. TREM-1 mRNA and supernatant cytokine levels were assayed. Mitochondrial oxidative stress, ROS, and membrane potential were assayed. Neonatal mice were injected with rmCIRP and speckle-tracking echocardiography was conducted to measure cardiac strain. Sepsis was induced by i.p. cecal slurry. Mouse pups were treated with M3 or vehicle. After 16 h, echocardiography was performed followed by euthanasia for tissue analysis. A 7-day survival study was conducted. Results Serum eCIRP levels were elevated in septic human neonates. rmCIRP stimulation of cardiomyocytes increased TREM-1 gene expression. Stimulation of cardiomyocytes with rmCIRP upregulated TNF-α and IL-6 in the supernatants, while this upregulation was inhibited by M3. Stimulation of cardiomyocytes with rmCIRP resulted in a reduction in mitochondrial membrane potential (MMP) while M3 treatment returned MMP to near baseline. rmCIRP caused mitochondrial calcium overload; this was inhibited by M3. rmCIRP injection impaired longitudinal and radial cardiac strain. Sepsis resulted in cardiac dysfunction with a reduction in cardiac output and left ventricular end diastolic diameter. Both were improved by M3 treatment. Treatment with M3 attenuated serum, cardiac, and pulmonary levels of pro-inflammatory cytokines compared to vehicle-treated septic neonates. M3 dramatically increased sepsis survival. Conclusions Inhibition of eCIRP/TREM-1 interaction with M3 is cardioprotective, decreases inflammation, and improves survival in neonatal sepsis. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Li Diao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.,Division of Pediatric Surgery, Cohen Children's Medical Center At Hofstra/Northwell, New Hyde Park, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA. .,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA. .,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
9
|
Elmassry MM, Mudaliar NS, Colmer-Hamood JA, San Francisco MJ, Griswold JA, Dissanaike S, Hamood AN. New markers for sepsis caused by Pseudomonas aeruginosa during burn infection. Metabolomics 2020; 16:40. [PMID: 32170472 PMCID: PMC7223005 DOI: 10.1007/s11306-020-01658-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/05/2020] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Sepsis is a leading cause of mortality in burn patients. One of the major causes of sepsis in burn patients is Pseudomonas aeruginosa. We hypothesized that during dissemination from infected burn wounds and subsequent sepsis, P. aeruginosa affects the metabolome of the blood resulting in changes to specific metabolites that would serve as biomarkers for early diagnosis of sepsis caused by P. aeruginosa. OBJECTIVES To identify specific biomarkers in the blood after sepsis caused by P. aeruginosa infection of burns. METHODS Gas chromatography with time-of-flight mass spectrometry was used to compare the serum metabolome of mice that were thermally injured and infected with P. aeruginosa (B-I) to that of mice that were neither injured nor infected, mice that were injured but not infected, and mice that were infected but not injured. RESULTS Serum levels of 19 metabolites were significantly increased in the B-I group compared to controls while levels of eight metabolites were significantly decreased. Thymidine, thymine, uridine, and uracil (related to pyrimidine metabolism), malate and succinate (a possible sign of imbalance in the tricarboxylic acid cycle), 5-oxoproline (related to glutamine and glutathione metabolism), and trans-4-hydroxyproline (a major component of the protein collagen) were increased. Products of amino acid metabolism were significantly decreased in the B-I group, including methionine, tyrosine, indole-3-acetate, and indole-3-propionate. CONCLUSION In all, 26 metabolites were identified, including a unique combination of five metabolites (trans-4-hydroxyproline, 5-oxoproline, glycerol-3-galactoside, indole-3-acetate, and indole-3-propionate) that could serve as a set of biomarkers for early diagnosis of sepsis caused by P. aeruginosa in burn patients.
Collapse
Affiliation(s)
- Moamen M Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Nithya S Mudaliar
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Caris Life Sciences, Phoenix, AZ, USA
| | - Jane A Colmer-Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Michael J San Francisco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
- Honors College, Texas Tech University, Lubbock, TX, USA
| | - John A Griswold
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sharmila Dissanaike
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Abdul N Hamood
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.
| |
Collapse
|
10
|
Kumar S, Tripathy S, Jyoti A, Singh SG. Recent advances in biosensors for diagnosis and detection of sepsis: A comprehensive review. Biosens Bioelectron 2018; 124-125:205-215. [PMID: 30388563 DOI: 10.1016/j.bios.2018.10.034] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/28/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022]
Abstract
Sepsis is one of the leading causes of mortality among critically ill patients globally. According to WHO report 2018, it is estimated to affect beyond 30 million people worldwide every year. It causes loss of human lives, which arise from infection and inflammation and long term stay in intensive care unit (ICU) in hospitals. Despite the availability of satisfactory prognostic markers contributing to the diagnosis of sepsis, millions of people die even after admission to the hospitals. Correct and early diagnosis of sepsis leads to rapid administration of appropriate antibiotics can thus potentially avert the attainment to critical stages of sepsis, thereby saving human lives. Conventional diagnostic practices are costly, time consuming and they lack adequate sensitivity and selectivity, provoking an urgent need for developing alternate sepsis diagnosis systems. Nevertheless, biosensors have the much-treasured scope for reasonable sepsis diagnosis. Advancement in nano-biotechnology has provided new paradigm for biosensor platforms with upgraded features. Here, we provide an overview of the recent advances in biosensors with a brief introduction to sepsis, followed by the conventional methods of diagnosis and bio-sensing. To conclude, a proactive role and an outlook on technologically advanced biosensor platforms are discoursed with possible biomedical applications.
Collapse
Affiliation(s)
- Sanni Kumar
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan 303002, India.
| | | | - Anupam Jyoti
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan 303002, India.
| | | |
Collapse
|
11
|
Chen W, Zhong H, Wang X, Pang Q, Zhuang J, Hu J, Chen Y, Hu J, Liu J, Tang J. Mig6 reduces inflammatory mediators production by regulating the activation of EGFR in LPS‐induced endotoxemia. J Cell Physiol 2018; 233:6975-6983. [DOI: 10.1002/jcp.26488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/11/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Wenting Chen
- Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Hanhui Zhong
- The Department of Anesthesia, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaofei Wang
- The Department of Anesthesia, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Qiongni Pang
- The Department of Anesthesia, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jinling Zhuang
- The Department of Anesthesia, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jian Hu
- Heart, Lung, Blood, and Vascular Medicine InstituteUniversity of PittsburghPittsburgh, Pennsylvania
| | - Yeming Chen
- The Department of AnesthesiaThe Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Jijie Hu
- The Department of Orthopaedics and Traumatology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jing Tang
- The Department of Anesthesia, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- The Department of AnesthesiaAffiliated hospital of Guangdong Medical UniversityGuangdongChina
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologySouthern Medical SciencesGuangzhouChina
| |
Collapse
|
12
|
Autophagic flux is essential for the downregulation of D-dopachrome tautomerase by atractylenolide I to ameliorate intestinal adenoma formation. J Cell Commun Signal 2018; 12:689-698. [PMID: 29368299 DOI: 10.1007/s12079-018-0454-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/18/2018] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer is generally believed to progress through an adenoma - carcinoma sequence. Adenomatous polyposis coli (APC) mutations serve as the initiating event in adenoma formation. The ApcMin/+ mouse harbors a mutation in the APC gene, which is similar or identical to the mutation found in individuals with familial adenomatous polyposis and 70% of all sporadic CRC cases. Autophagy is a constitutive process required for proper cellular homeostasis. However, its role in intestinal adenoma formation is still controversial. Atractylenolide I (AT1) is a sesquiterpenoid that possesses various clinically relevant properties such as anti-tumor and anti-inflammatory activities. The role of AT1 on adenoma formation was tested in ApcMin/+ mice and its underlying mechanism in regulating autophagy was documented. D-dopachrome tautomerase (D-DT) was identified as a potential target of AT1 by an proteomics-based approach. The effects of p53 modification on autophgic flux was monitored in p53-/- and p53+/+ HCT116 cells. Small interfering RNA was used to investigate the function of Atg7 and D-DT on autophagy programme induce by AT1. AT1 effectively reduced the formation of adenoma and downregulated the tumorigenic proteins in ApcMin/+ mice. Importantly, AT1 stimulated autophagic flux through downregulating acetylation of p53. Activation of Sirt1 by AT1 was essential for the deacetylation of p53 and downregulation of D-DT. The lowered expression of COX-2 and β-catenin by AT1 were partly recovered by Atg7 knockdown. AT1 activates autophagy machinery to downregulate D-DT and reduce intestinal adenoma formation. This discovery provides evidence in vivo and in vitro that inducing autophagy by natural products maybe a potential therapy to ameliorate colorectal adenoma formation.
Collapse
|
13
|
Exogenous carbon monoxide inhibits neutrophil infiltration in LPS-induced sepsis by interfering with FPR1 via p38 MAPK but not GRK2. Oncotarget 2018; 7:34250-65. [PMID: 27144520 PMCID: PMC5085153 DOI: 10.18632/oncotarget.9084] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/11/2016] [Indexed: 12/29/2022] Open
Abstract
Excessive neutrophil infiltration in vital organs is life-threatening to patients who suffer from sepsis. We identified a critical role of exogenous carbon monoxide (CO) in the inhibition of neutrophil infiltration during lipopolysaccharide (LPS)-induced sepsis. CO delivered from carbon monoxide-releasing molecule 2 (CORM-2) dramatically increased the survival rate of C57BL/6 mice subjected to LPS in vivo. CORM-2 significantly suppressed neutrophil infiltration in liver and lung as well as markers of inflammatory responses. Affymetrix GeneChip array analysis revealed that the increased expression of chemoattractant receptor formyl peptide receptor 1 (FPR1) may contribute to the excessive neutrophil infiltration. The under agarose migration assay demonstrated that LPS stimulation promoted migration to the ligand of FPR1, N-Formyl-Met-Leu-Phe (fMLP) but that CORM-2 treatment inhibited this promotion. Further studies demonstrated that CORM-2 internalized FPR1 by inhibiting p38 mitogen-activated protein kinase (MAPK) but not G protein-coupled receptor kinase 2 (GRK2), which may explain the inhibitory effect of CORM-2 on LPS-stimulated neutrophils. In summary, our study demonstrates that exogenous CO inhibits sepsis-induced neutrophil infiltration by interfering with FPR1 via p38 MAPK but not GRK2.
Collapse
|
14
|
Kersh AE, Ng S, Chang YM, Sasaki M, Thomas SN, Kissick HT, Lesinski GB, Kudchadkar RR, Waller EK, Pollack BP. Targeted Therapies: Immunologic Effects and Potential Applications Outside of Cancer. J Clin Pharmacol 2018; 58:7-24. [PMID: 29136276 PMCID: PMC5972536 DOI: 10.1002/jcph.1028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
Two pharmacologic approaches that are currently at the forefront of treating advanced cancer are those that center on disrupting critical growth/survival signaling pathways within tumor cells (commonly referred to as "targeted therapies") and those that center on enhancing the capacity of a patient's immune system to mount an antitumor response (immunotherapy). Maximizing responses to both of these approaches requires an understanding of the oncogenic events present in a given patient's tumor and the nature of the tumor-immune microenvironment. Although these 2 modalities were developed and initially used independently, combination regimens are now being tested in clinical trials, underscoring the need to understand how targeted therapies influence immunologic events. Translational studies and preclinical models have demonstrated that targeted therapies can influence immune cell trafficking, the production of and response to chemokines and cytokines, antigen presentation, and other processes relevant to antitumor immunity and immune homeostasis. Moreover, because these and other effects of targeted therapies occur in nonmalignant cells, targeted therapies are being evaluated for use in applications outside of oncology.
Collapse
Affiliation(s)
- Anna E. Kersh
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Spencer Ng
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yun Min Chang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Atlanta, GA
| | | | - Susan N. Thomas
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haydn T. Kissick
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gregory B. Lesinski
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ragini R. Kudchadkar
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Edmund K. Waller
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian P. Pollack
- Atlanta VA Medical Center, Atlanta, GA, USA
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA
- Emory University Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
15
|
Lai Y, Tao L, Zhao Y, Zhang X, Sun X, Wang Q, Xu C. Cx32 inhibits TNFα-induced extrinsic apoptosis with and without EGFR suppression. Oncol Rep 2017; 38:2885-2892. [PMID: 28901517 DOI: 10.3892/or.2017.5950] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor α (TNFα) and TNF-related apoptosis-inducing ligand (TRAIL) can trigger the extrinsic apoptosis pathway. Our previous study indicated that connexin32 (Cx32) inhibited streptonigrin-induced intrinsic apoptosis via the epidermal growth factor receptor (EGFR) pathway. However, whether Cx32 can exert effects on the extrinsic apoptosis pathway through EGFR signaling remains unclear. In the present study, we investigated the role of Cx32 in extrinsic apoptosis induced by treatment with TNFα + cycloheximide (CHX) or afatinib in human cervical cancer (CaCx) cells. In stable inducible Cx32-transfected HeLa cells (HeLa-Cx32), Cx32 expression was induced by treatment with doxycycline (Dox). Furthermore, C-33A cells, which natively express high levels of Cx32, were used as a cell model for knockdown of Cx32 with siRNA. To determine the non-junctional function of Cx32 in apoptosis, 18α-glycyrrhetinic acid (18α-GA), a gap junction intracellular communication (GJIC) inhibitor, was used. Our results showed that Cx32 could inhibit apoptosis induced by TNFα + afatinib with or without the GJIC inhibitor. In clinical cervical tissue samples, we found that the expression of survivin was markedly higher in CaCx than in normal cervix tissue, which was in accordance with the expression of Cx32 in our previous study. In HeLa-Cx32 cells, we also found that Cx32 upregulated the expression of Cox-2. In addition, Cx32 upregulated EGFR expression in low-density culture (lacking GJ formation). Cx32 could also promote the expression of EGFR, phospho-STAT3 and phospho-ERK in HeLa-Cx32 cells following TNFα treatment. After knocking down Cx32 in C-33A cells, the expression levels of survivin and TNFα were downregulated. The present study verifies that Cx32 exerts an inhibitory effect on extrinsic apoptosis in CaCx cells, and suggests that Cx32 may regulate the progression and micro-environment of CaCx cells.
Collapse
Affiliation(s)
- Yongchang Lai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yifan Zhao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaomin Zhang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xingjuan Sun
- Traditional Chinese Medicine Hospital of Guangdong, Guangzhou, Guangdong 510120, P.R. China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chengfang Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
16
|
Transactivation of the epidermal growth factor receptor in responses to myocardial stress and cardioprotection. Int J Biochem Cell Biol 2017; 83:97-110. [PMID: 28049018 DOI: 10.1016/j.biocel.2016.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/25/2016] [Accepted: 12/26/2016] [Indexed: 12/20/2022]
|
17
|
Xu J, Mukerjee S, Silva-Alves CRA, Carvalho-Galvão A, Cruz JC, Balarini CM, Braga VA, Lazartigues E, França-Silva MS. A Disintegrin and Metalloprotease 17 in the Cardiovascular and Central Nervous Systems. Front Physiol 2016; 7:469. [PMID: 27803674 PMCID: PMC5067531 DOI: 10.3389/fphys.2016.00469] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/30/2016] [Indexed: 01/19/2023] Open
Abstract
ADAM17 is a metalloprotease and disintegrin that lodges in the plasmatic membrane of several cell types and is able to cleave a wide variety of cell surface proteins. It is somatically expressed in mammalian organisms and its proteolytic action influences several physiological and pathological processes. This review focuses on the structure of ADAM17, its signaling in the cardiovascular system and its participation in certain disorders involving the heart, blood vessels, and neural regulation of autonomic and cardiovascular modulation.
Collapse
Affiliation(s)
- Jiaxi Xu
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Snigdha Mukerjee
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | | | | - Josiane C Cruz
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Camille M Balarini
- Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Valdir A Braga
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | |
Collapse
|