1
|
Zhou W, Wang N, Dong S, Huan Z, Sui L, Ge X. PRG4 mitigates hemorrhagic shock-induced cardiac injury by inhibiting mitochondrial dysregulation, oxidative stress and NLRP3-mediated pyroptosis. Int Immunopharmacol 2024; 137:112507. [PMID: 38897120 DOI: 10.1016/j.intimp.2024.112507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Hemorrhagic shock (HS) is one of the main causes of morbidity and death in patients with trauma or major surgery. Cardiac dysfunction is a well-known complication of HS. PRG4, also known as lubricin, is a mucin-like glycoprotein that plays anti-inflammatory and anti-apoptotic roles in a variety of diseases. In this study, we aimed to explore the cardioprotective efficacy of PRG4 in HS-induced cardiac injury. Employing the HS model and RNA-seq, we found that PRG4 was increased in the myocardial tissue of rats after HS. In vivo studies suggested that HS led to abnormal hemodynamic parameters and increased cTnI levels, and PRG4 overexpression effectively reversed these changes. PRG4 also suppressed HS-induced mitochondrial disorders, as reflected by increased mitochondrial membrane potential (MMP), ATP and mitochondria cytochrome c, COXIV and TOM20, as well as decreased BNIP3L and cytoplasmic cytochrome c. Furthermore, HS led to enhanced oxidative stress, as evidenced by upregulated ROS and MDA contents, and downregulated SOD and CAT activities, and these alterations were negated by PRG4 overexpression. Notably, PRG4 repressed the NLRP3-mediated pyroptosis pathway, as illustrated by decreased NLRP3 levels, caspase-1 activity and GSDMD-NT levels. In summary, these observations indicate that PRG4 overexpression protects against HS-induced cardiac dysfunction by inhibiting mitochondrial dysregulation, oxidative stress and NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Wuming Zhou
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Nan Wang
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Sheng Dong
- Department of Emergency, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Zhirong Huan
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Lijun Sui
- Department of Cardiology, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China.
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China; Department of Emergency, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China; Orthopedic Institution of Wuxi City, Wuxi, Jiangsu 214000, People's Republic of China.
| |
Collapse
|
2
|
Zhang H, Xiang L, Yuan H, Yu H. PTPRO inhibition ameliorates spinal cord injury through shifting microglial M1/M2 polarization via the NF-κB/STAT6 signaling pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167141. [PMID: 38565385 DOI: 10.1016/j.bbadis.2024.167141] [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: 11/07/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Spinal cord injury (SCI) induces severe neuroinflammation, and subsequently neurological dysfunction. Activated microglia are critical for modulation of neuroinflammation. Protein tyrosine phosphatase receptor type O (PTPRO), a member of protein tyrosine phosphatases (PTPs), exerts a pro-inflammatory role in multiple human diseases; however, its role in SCI remains unclarified. Here, a T7 spinal cord compression injury model was established in Sprague-Dawley (SD) rats, and PTPRO expression was upregulated in injured spinal cord and microglia after SCI. Microglia M1 and M2 polarization in vitro were induced using LPS/IFN-γ and IL-4, respectively. PTPRO expression was elevated in M1-polarized microglia, and PTPRO downregulation mediated by PTPRO shRNA (shPTPRO) decreased CD86+ cell proportion, iNOS, TNF-α, IL-1β, and IL-6 levels, and p65 phosphorylation. PTPRO was downregulated in M2 microglia, and PTPRO upregulation by PTPRO overexpression plasmid (OE-PTPRO) reduced CD206+ cell percentage, Arg-1, IL-10, and TGF-β1 levels and STAT6 phosphorylation. Mechanistically, the transcription factor SOX4 elevated PTPRO expression and its promoter activity. SOX4 overexpression enhanced M1 polarization and p65 phosphorylation, while its knockdown promoted M2 polarization and STAT6 phosphorylation. PTPRO might mediate the function of SOX4 in BV2 microglia polarization. Furthermore, lentivirus-mediated downregulation of PTPRO following SCI improved locomotor functional recovery, demonstrated by elevated BBB scores, incline angle, consistent hindlimb coordination, and reduced lesion area and neuronal apoptosis. PTPRO downregulation promoted microglia M2 polarization, NF-κB inactivation and STAT6 activation after injury. In conclusion, PTPRO inhibition improves spinal cord injury through facilitating M2 microglia polarization via the NF-κB/STAT6 signaling pathway, which is probably controlled by SOX4.
Collapse
Affiliation(s)
- Haocong Zhang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Liangbi Xiang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Hong Yuan
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Hailong Yu
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
3
|
Dai Y, Shi S, Liu H, Zhou H, Ding W, Liu C, Jin L, Xie W, Kong H, Zhang Q. Protein tyrosine phosphatase PTPRO represses lung adenocarcinoma progression by inducing mitochondria-dependent apoptosis and restraining tumor metastasis. Cell Death Dis 2024; 15:11. [PMID: 38182570 PMCID: PMC10770368 DOI: 10.1038/s41419-023-06375-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: 01/12/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024]
Abstract
Emerging evidence indicates that protein activities regulated by receptor protein tyrosine phosphatases (RPTPs) are crucial for a variety of cellular processes, such as proliferation, apoptosis, and immunological response. Protein tyrosine phosphatase receptor type O (PTPRO), an RPTP, has been revealed as a putative suppressor in the development of particular tumors. However, the function and the underlying mechanisms of PTPRO in regulating of lung adenocarcinoma (LUAD) are not well understood. In this view, the present work investigated the role of PTPRO in LUAD. Analysis of 90 pairs of clinical LUAD specimens revealed significantly lower PTPRO levels in LUAD compared with adjacent non-tumor tissue, as well as a negative correlation of PTPRO expression with tumor size and TNM stage. Survival analyses demonstrated that PTPRO level can help stratify the prognosis of LUAD patients. Furthermore, PTPRO overexpression was found to suppress the progression of LUAD both in vitro and in vivo by inducing cell death via mitochondria-dependent apoptosis, downregulating protein expression of molecules (Bcl-2, Bax, caspase 3, cleaved-caspase 3/9, cleaved-PARP and Bid) essential in cell survival. Additionally, PTPRO decreased LUAD migration and invasion by regulating proteins involved in the epithelial-to-mesenchymal transition (E-cadherin, N-cadherin, and Snail). Moreover, PTPRO was shown to restrain JAK2/STAT3 signaling pathways. Expression of PTPRO was negatively correlated with p-JAK2, p-STAT3, Bcl-2, and Snail levels in LUAD tumor samples. Furthermore, the anti-tumor effect of PTPRO in LUAD was significant but compromised in STAT3-deficient cells. These data support the remarkable suppressive role of PTPRO in LUAD, which may represent a viable therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Yuan Dai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
- Department of Respiratory Medicine, Jiangsu Province Official Hospital, Nanjing, China
| | - Shuangshuang Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hongda Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hong Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wenqiu Ding
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chenyang Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Linling Jin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Weiping Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Hui Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Qun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
4
|
Jia D, Han J, Cai J, Huan Z, Wang Y, Ge X. Mesenchymal stem cells overexpressing PBX1 alleviates haemorrhagic shock-induced kidney damage by inhibiting NF-κB activation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119571. [PMID: 37673222 DOI: 10.1016/j.bbamcr.2023.119571] [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: 04/28/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Mesenchymal stem cells (MSCs) have favourable outcomes in the treatment of kidney diseases. Pre-B-cell leukaemia transcription factor 1 (PBX1) has been reported to be a regulator of self-renewal of stem cells. Whether PBX1 is beneficial to MSCs in the treatment of haemorrhagic shock (HS)-induced kidney damage is unknown. We overexpressed PBX1 in rat bone marrow-derived mesenchymal stem cells (rBMSCs) and human bone marrow-derived mesenchymal stem cells (hBMSCs) to treat rats with HS and hypoxia-treated human proximal tubule epithelial cells (HK-2), respectively. The results indicated that PBX1 enhanced the homing capacity of rBMSCs to kidney tissues and that treatment with rBMSCs overexpressing PBX1 improved the indicators of kidney function, alleviated structural damage to kidney tissues. Furthermore, administration with rBMSCs overexpressing PBX1 inhibited HS-induced NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and the release of proinflammatory cytokines, and further attenuated apoptosis. We then determined whether NF-κB, an important factor in NLRP3 activation and the regulation of inflammation, participates in HS-induced kidney damage, and we found that rBMSCs overexpressing PBX1 inhibited NF-κB activation by decreasing the p-IκBα/IκBα and p-p65/p65 ratios and inhibiting the nuclear translocation and decreasing the DNA-binding capacity of NF-κB. hBMSCs overexpressing PBX1 also exhibited protective effects on HK-2 cells exposed to hypoxia, as shown by the increase in cell viability, the mitigation of apoptosis, the decrease in inflammation, and the inhibition of NF-κB and NLRP3 inflammasome activation. Our study demonstrates that MSCs overexpressing PBX1 ameliorates HS-induced kidney damage by inhibiting NF-κB pathway-mediated NLRP3 inflammasome activation and the inflammatory response.
Collapse
Affiliation(s)
- Di Jia
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Jiahui Han
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Jimin Cai
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Zhirong Huan
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Yan Wang
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214000, People's Republic of China; Orthopedic Institution of Wuxi City, Wuxi, Jiangsu 214000, People's Republic of China.
| |
Collapse
|
5
|
Liu Z, Zhou W, Liu Q, Huan Z, Wang Q, Ge X. Pachymic Acid Prevents Hemorrhagic Shock-Induced Cardiac Injury by Suppressing M1 Macrophage Polarization and NF-[Formula: see text]B Signaling Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:2157-2173. [PMID: 37865871 DOI: 10.1142/s0192415x23500921] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Hemorrhagic shock (HS) is the leading cause of death in trauma patients. Inflammation following HS can lead to cardiac damage. Pachymic acid (PA), a triterpenoid extracted from Poria cocos, has been found to possess various biological activities, including anti-inflammatory and anti-apoptotic properties. Our research aims to investigate the protective effects of PA against HS-induced heart damage and the underlying mechanisms involved. Male Sprague-Dawley rats were intraperitoneally injected with PA (7.5 or 15[Formula: see text]mg/kg) daily for three days. Subsequently, we created a rat model of HS by drawing blood through a catheter inserted into the femoral artery followed by resuscitation. The results revealed that HS led to abnormalities in hemodynamics, serum cardiac enzyme levels, and cardiac structure, as well as induced cardiac apoptosis. However, pretreatment with PA effectively alleviated these effects. PA-pretreatment also suppressed mRNA and protein levels of interleukin (IL)-1[Formula: see text], IL-6, and tumor necrosis factor [Formula: see text] (TNF-[Formula: see text]) in the heart tissues of HS rats. Additionally, PA-pretreatment reduced inflammatory cell infiltration and M1 macrophage polarization while exaggerating M2 polarization in HS rat hearts. The study observed a decreased proportion of the expression of of M1 macrophages (CD86[Formula: see text]) and their marker (iNOS), along with an increased proportion of the expression of M2 macrophages (CD206[Formula: see text]) and their marker (Arg-1). Notably, PA-pretreatment suppressed NF-[Formula: see text]B pathway activation via inhibiting NF-[Formula: see text]B p65 phosphorylation and its nuclear translocation. In conclusion, PA-pretreatment ameliorates HS-induced cardiac injury, potentially through its inhibition of the NF-[Formula: see text]B pathway. Therefore, PA treatment holds promise as a strategy for mitigating cardiac damage in HS.
Collapse
Affiliation(s)
- Zhenfeng Liu
- Department of Traditional Chinese Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
| | - Wuming Zhou
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
| | - Qingyang Liu
- Department of Clinical Laboratory, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
| | - Zhirong Huan
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
| | - Xin Ge
- Department of Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University Wuxi, Jiangsu 214000, P. R. China
- Orthopedic Institution of Wuxi City, Wuxi, Jiangsu 214000, P. R. China
| |
Collapse
|
6
|
Kelestemur T, Németh ZH, Pacher P, Beesley J, Robson SC, Eltzschig HK, Haskó G. Adenosine metabolized from extracellular ATP ameliorates organ injury by triggering A 2BR signaling. Respir Res 2023; 24:186. [PMID: 37438813 PMCID: PMC10339538 DOI: 10.1186/s12931-023-02486-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Trauma and a subsequent hemorrhagic shock (T/HS) result in insufficient oxygen delivery to tissues and multiple organ failure. Extracellular adenosine, which is a product of the extracellular degradation of adenosine 5' triphosphate (ATP) by the membrane-embedded enzymes CD39 and CD73, is organ protective, as it participates in signaling pathways, which promote cell survival and suppress inflammation through adenosine receptors including the A2BR. The aim of this study was to evaluate the role of CD39 and CD73 delivering adenosine to A2BRs in regulating the host's response to T/HS. METHODS T/HS shock was induced by blood withdrawal from the femoral artery in wild-type, global knockout (CD39, CD73, A2BR) and conditional knockout (intestinal epithelial cell-specific deficient VillinCre-A2BRfl/fl) mice. At 3 three hours after resuscitation, blood and tissue samples were collected to analyze organ injury. RESULTS T/HS upregulated the expression of CD39, CD73, and the A2BR in organs. ATP and adenosine levels increased after T/HS in bronchoalveolar lavage fluid. CD39, CD73, and A2BR mimics/agonists alleviated lung and liver injury. Antagonists or the CD39, CD73, and A2BR knockout (KO) exacerbated lung injury, inflammatory cytokines, and chemokines as well as macrophage and neutrophil infiltration and accumulation in the lung. Agonists reduced the levels of the liver enzymes aspartate transferase and alanine transaminase in the blood, whereas antagonist administration or CD39, CD73, and A2BR KO enhanced enzyme levels. In addition, intestinal epithelial cell-specific deficient VillinCre-A2BRfl/fl mice showed increased intestinal injury compared to their wild-type VillinCre controls. CONCLUSION In conclusion, the CD39-CD73-A2BR axis protects against T/HS-induced multiple organ failure.
Collapse
Affiliation(s)
- Taha Kelestemur
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA
- Department of Physiology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zoltán H Németh
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA
- Department of Surgery, Morristown Medical Center, Morristown, NJ, 07960, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Jennet Beesley
- Daresbury Proteins Ltd, Sci-Tech Daresbury, Warrington, UK
| | - Simon C Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA.
| |
Collapse
|