1
|
Liu Z, Liu Y, Xing T, Li J, Zhang L, Zhao L, Jiang Y, Gao F. Unraveling the role of long non-coding RNAs in chronic heat stress-induced muscle injury in broilers. J Anim Sci Biotechnol 2024; 15:135. [PMID: 39375773 PMCID: PMC11459952 DOI: 10.1186/s40104-024-01093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Chronic heat stress (CHS) is a detrimental environmental stressor with a negative impact on the meat quality of broilers. However, the underlying mechanisms are not fully understood. This study investigates the effects of CHS on long non-coding RNA (lncRNA) expression and muscle injury in broilers, with a focus on its implications for meat quality. RESULTS The results showed that CHS diminished breast muscle yield, elevated abdominal fat deposition, induced cellular apoptosis (P < 0.05), and caused myofibrosis. Transcriptomic analysis revealed 151 differentially expressed (DE) lncRNAs when comparing the normal control (NC) and HS groups, 214 DE lncRNAs when comparing the HS and PF groups, and 79 DE lncRNAs when comparing the NC and pair-fed (PF) groups. After eliminating the confounding effect of feed intake, 68 lncRNAs were identified, primarily associated with cellular growth and death, signal transduction, and metabolic regulation. Notably, the apoptosis-related pathway P53, lysosomes, and the fibrosis-related gene TGF-β2 were significantly upregulated by lncRNAs. CONCLUSIONS These findings indicate that chronic heat stress induces cellular apoptosis and muscle injury through lncRNA, leading to connective tissue accumulation, which likely contributes to reduced breast muscle yield and meat quality in broilers.
Collapse
Affiliation(s)
- Zhen Liu
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
- Anhui Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Yingsen Liu
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Jiaolong Li
- Institute of Agro-Products Processing, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Liang Zhao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Yun Jiang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
2
|
Zhang Z, Wu X, Zou Z, Shen M, Liu Q, Zhangsun Z, Zhao H, Lei W, Wang Z, Dong Y, Yang Y. Heat stroke: Pathogenesis, diagnosis, and current treatment. Ageing Res Rev 2024; 100:102409. [PMID: 38986844 DOI: 10.1016/j.arr.2024.102409] [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: 02/05/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Recently, the incidence of heat-related illnesses has exhibited a steadily upward trend, which is closely associated with several environmental factors such as climate change and air pollution. The progression of heat-related illnesses is a continuous process and can progress to the terminal period when it transforms into heat stroke, the most severe form. Heat stroke is markedly by a core body temperature above 40°C and central nervous system dysfunction. Current knowledge suggests that the pathogenesis of heat stroke is complex and varied, including inflammatory response, oxidative stress, cell death, and coagulation dysfunction. This review consolidated recent research progress on the pathophysiology and pathogenesis of heat stroke, with a focus on the related molecular mechanisms. In addition, we reviewed common strategies and sorted out the drugs in various preclinical stages for heat stroke, aiming to offer a comprehensive research roadmap for more in-depth researches into the mechanisms of heat stroke and the reduction in the mortality of heat stroke in the future.
Collapse
Affiliation(s)
- Zhe Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Xiaopeng Wu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Zheng Zou
- Department of Neurosurgery, The General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of Chinese PLA General Hospital, 80 Jianglin Road, Hainan, 572013, China
| | - Qiong Liu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ziyin Zhangsun
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Yushu Dong
- Department of Neurosurgery, The General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China.
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
3
|
Wang Z, Zhu J, Zhang D, Lv J, Wu L, Liu Z. The significant mechanism and treatments of cell death in heatstroke. Apoptosis 2024; 29:967-980. [PMID: 38886312 DOI: 10.1007/s10495-024-01979-w] [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] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
With global warming, extreme environmental heat is becoming a social issue of concern, which can cause adverse health results including heatstroke (HS). Severe heat stress is characterized by cell death of direct heat damage, excessive inflammatory responses, and coagulation disorders that can lead to multiple organ dysfunction (MODS) and even death. However, the significant pathophysiological mechanism and treatment of HS are still not fully clear. Various modes of cell death, including apoptosis, pyroptosis, ferroptosis, necroptosis and PANoptosis are involved in MODS induced by heatstroke. In this review, we summarized molecular mechanism, key transcriptional regulation as for HSF1, NRF2, NF-κB and PARP-1, and potential therapies of cell death resulting in CNS, liver, intestine, reproductive system and kidney injury induced by heat stress. Understanding the mechanism of cell death provides new targets to protect multi-organ function in HS.
Collapse
Affiliation(s)
- Zixin Wang
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510010, China
| | - Jie Zhu
- Department of Pediatric, General Hospital of Southern Theater Command of PLA, Guangzhou, 510010, China
| | - Dingshun Zhang
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou, 510010, China
| | - Jinke Lv
- Department of Thoracic Surgery, Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liangping Wu
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510010, China.
| | - Zhifeng Liu
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou, 510010, China.
| |
Collapse
|
4
|
Liang M, Lyu ZS, Zhang YY, Tang SQ, Xing T, Chen YH, Wang Y, Jiang Q, Xu LP, Zhang XH, Huang XJ, Kong Y. Activation of PPARδ in bone marrow endothelial progenitor cells improves their hematopoiesis-supporting ability after myelosuppressive injury. Cancer Lett 2024; 592:216937. [PMID: 38704134 DOI: 10.1016/j.canlet.2024.216937] [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: 03/07/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Dysfunctional bone marrow (BM) endothelial progenitor cells (EPCs) with high levels of reactive oxygen species (ROS) are responsible for defective hematopoiesis in poor graft function (PGF) patients with acute leukemia or myelodysplastic neoplasms post-allotransplant. However, the underlying mechanism by which BM EPCs regulate their intracellular ROS levels and the capacity to support hematopoiesis have not been well clarified. Herein, we demonstrated decreased levels of peroxisome proliferator-activated receptor delta (PPARδ), a lipid-activated nuclear receptor, in BM EPCs of PGF patients compared with those with good graft function (GGF). In vitro assays further identified that PPARδ knockdown contributed to reduced and dysfunctional BM EPCs, characterized by the impaired ability to support hematopoiesis, which were restored by PPARδ overexpression. Moreover, GW501516, an agonist of PPARδ, repaired the damaged BM EPCs triggered by 5-fluorouracil (5FU) in vitro and in vivo. Clinically, activation of PPARδ by GW501516 benefited the damaged BM EPCs from PGF patients or acute leukemia patients in complete remission (CR) post-chemotherapy. Mechanistically, we found that increased expression of NADPH oxidases (NOXs), the main ROS-generating enzymes, may lead to elevated ROS level in BM EPCs, and insufficient PPARδ may trigger BM EPC damage via ROS/p53 pathway. Collectively, we found that defective PPARδ contributes to BM EPC dysfunction, whereas activation of PPARδ in BM EPCs improves their hematopoiesis-supporting ability after myelosuppressive therapy, which may provide a potential therapeutic target not only for patients with leukemia but also for those with other cancers.
Collapse
Affiliation(s)
- Mi Liang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhong-Shi Lyu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| | - Shu-Qian Tang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Tong Xing
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yu-Hong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, China.
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| |
Collapse
|
5
|
She L, Zhang X, Shen R, He S, Miao X. Expression and role of FKBPL in lung adenocarcinoma. J Cancer 2024; 15:166-175. [PMID: 38164287 PMCID: PMC10751668 DOI: 10.7150/jca.87758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/29/2023] [Indexed: 01/03/2024] Open
Abstract
Dysregulated expression of FK506-binding protein like (FKBPL) has been demonstrated to play crucial roles in tumour development. However, the role of FKBPL in lung adenocarcinoma (ADC) remains unclear. Using immunohistochemical staining, we showed that FKBPL expression was significantly lower in lung ADC than the normal tissues (P < 0.0001). Patients with well or moderately differentiated tumours have higher FKBPL expression compared with patients with poor differentiated tumours (P = 0.037). However, no significant associations were found between FKBPL expression and other clinicopathological variables (P > 0.05 for all). Cox univariate analysis showed that high FKBPL expression was correlated with prolonged overall survival (OS) (P = 0.010). Kaplan-Meier analysis further confirmed that the FKBPL-low group showed a significantly shorter OS than the FKBPL-high group (P = 0.0081). FKBPL expression was not shown as an independent prognostic factor for OS in the multivariate analysis (P = 0.063). Moreover, our study demonstrated that FKBPL could suppress the proliferation of lung ADC cells by delaying cell cycle G1/S phase transition. In addition, FKBPL resulted in increased apoptosis in lung ADC cells. Using the Human Apoptosis Array Kit, we observed that overexpression of FKBPL in lung ADC A549 cells significantly decreased the anti-apoptotic proteins, including heat shock protein 32 (HSP32), heat shock protein 27 (HSP27), and paraoxonase-2 (PON2). FKBPL depletion significantly attenuated the pro-apoptotic protein, phospho-p53 (S46), in lung ADC H1975 cells. These new findings provide an experimental basis for further theoretical investigation of lung ADC.
Collapse
Affiliation(s)
- Lili She
- Department of Pathology, Affiliated Tumour Hospital of Nantong University, Nantong, China
- Department of Pathology, Nantong Sixth People's Hospital, Nantong, China
| | - Xingsong Zhang
- Department of Pathology, Affiliated Tumour Hospital of Nantong University, Nantong, China
| | - Rong Shen
- Department of Pathology, Affiliated Tumour Hospital of Nantong University, Nantong, China
| | - Song He
- Department of Pathology, Affiliated Tumour Hospital of Nantong University, Nantong, China
| | - Xiaobing Miao
- Department of Pathology, Affiliated Tumour Hospital of Nantong University, Nantong, China
| |
Collapse
|
6
|
Zhou Y, Nakajima R, Shirasawa M, Fikriyanti M, Zhao L, Iwanaga R, Bradford AP, Kurayoshi K, Araki K, Ohtani K. Expanding Roles of the E2F-RB-p53 Pathway in Tumor Suppression. BIOLOGY 2023; 12:1511. [PMID: 38132337 PMCID: PMC10740672 DOI: 10.3390/biology12121511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
The transcription factor E2F links the RB pathway to the p53 pathway upon loss of function of pRB, thereby playing a pivotal role in the suppression of tumorigenesis. E2F fulfills a major role in cell proliferation by controlling a variety of growth-associated genes. The activity of E2F is controlled by the tumor suppressor pRB, which binds to E2F and actively suppresses target gene expression, thereby restraining cell proliferation. Signaling pathways originating from growth stimulative and growth suppressive signals converge on pRB (the RB pathway) to regulate E2F activity. In most cancers, the function of pRB is compromised by oncogenic mutations, and E2F activity is enhanced, thereby facilitating cell proliferation to promote tumorigenesis. Upon such events, E2F activates the Arf tumor suppressor gene, leading to activation of the tumor suppressor p53 to protect cells from tumorigenesis. ARF inactivates MDM2, which facilitates degradation of p53 through proteasome by ubiquitination (the p53 pathway). P53 suppresses tumorigenesis by inducing cellular senescence or apoptosis. Hence, in almost all cancers, the p53 pathway is also disabled. Here we will introduce the canonical functions of the RB-E2F-p53 pathway first and then the non-classical functions of each component, which may be relevant to cancer biology.
Collapse
Affiliation(s)
- Yaxuan Zhou
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Rinka Nakajima
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mashiro Shirasawa
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Mariana Fikriyanti
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Lin Zhao
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| | - Ritsuko Iwanaga
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Andrew P. Bradford
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; (R.I.); (A.P.B.)
| | - Kenta Kurayoshi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Keigo Araki
- Department of Morphological Biology, Ohu University School of Dentistry, 31-1 Misumido Tomitamachi, Koriyama, Fukushima 963-8611, Japan;
| | - Kiyoshi Ohtani
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan; (Y.Z.); (R.N.); (M.S.); (M.F.); (L.Z.)
| |
Collapse
|
7
|
Wang H, Yu W, Wang Y, Wu R, Dai Y, Deng Y, Wang S, Yuan J, Tan R. p53 contributes to cardiovascular diseases via mitochondria dysfunction: A new paradigm. Free Radic Biol Med 2023; 208:846-858. [PMID: 37776918 DOI: 10.1016/j.freeradbiomed.2023.09.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
Cardiovascular diseases (CVDs) are leading causes of global mortality; however, their underlying mechanisms remain unclear. The tumor suppressor factor p53 has been extensively studied for its role in cancer and is also known to play an important role in regulating CVDs. Abnormal p53 expression levels and modifications contribute to the occurrence and development of CVDs. Additionally, mounting evidence underscores the critical involvement of mitochondrial dysfunction in CVDs. Notably, studies indicate that p53 abnormalities directly correlate with mitochondrial dysfunction and may even interact with each other. Encouragingly, small molecule inhibitors targeting p53 have exhibited remarkable effects in animal models of CVDs. Moreover, therapeutic strategies aimed at mitochondrial-related molecules and mitochondrial replacement therapy have demonstrated their advantageous potential. Therefore, targeting p53 or mitochondria holds immense promise as a pioneering therapeutic approach for combating CVDs. In this comprehensive review, we delve into the mechanisms how p53 influences mitochondrial dysfunction, including energy metabolism, mitochondrial oxidative stress, mitochondria-induced apoptosis, mitochondrial autophagy, and mitochondrial dynamics, in various CVDs. Furthermore, we summarize and discuss the potential significance of targeting p53 or mitochondria in the treatment of CVDs.
Collapse
Affiliation(s)
- Hao Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Wei Yu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yibo Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ruihao Wu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yifei Dai
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ye Deng
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, 272000, China.
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
8
|
Rius-Pérez S. p53 at the crossroad between mitochondrial reactive oxygen species and necroptosis. Free Radic Biol Med 2023; 207:183-193. [PMID: 37481144 DOI: 10.1016/j.freeradbiomed.2023.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
p53 is a redox-sensitive transcription factor that can regulate multiple cell death programs through different signaling pathways. In this review, we assess the role of p53 in the regulation of necroptosis, a programmed form of lytic cell death highly involved in the pathophysiology of multiple diseases. In particular, we focus on the role of mitochondrial reactive oxygen species (mtROS) as essential contributors to modulate necroptosis execution through p53. The enhanced generation of mtROS during necroptosis is critical for the correct interaction between receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and 3 (RIPK3), two key components of the functional necrosome. p53 controls the occurrence of necroptosis by modulating the levels of mitochondrial H2O2 via peroxiredoxin 3 and sulfiredoxin. Furthermore, in response to increased levels of H2O2, p53 upregulates the long non-coding RNA necrosis-related factor, favoring the translation of RIPK1 and RIPK3. In parallel, a fraction of cytosolic p53 migrates into mitochondria, a process notably involved in necroptosis execution via its interaction with the mitochondrial permeability transition pore. In conclusion, p53 is located at the intersection between mtROS and the necroptosis machinery, making it a key protein to orchestrate redox signaling during necroptosis.
Collapse
Affiliation(s)
- Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100, Valencia, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
| |
Collapse
|
9
|
Chen J, Ding C, Cao J, Tong H, Chen Y. Heat stress combined with lipopolysaccharide induces pulmonary microvascular endothelial cell glycocalyx inflammatory damage in vitro. Immun Inflamm Dis 2023; 11:e1034. [PMID: 37904703 PMCID: PMC10552074 DOI: 10.1002/iid3.1034] [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: 01/05/2023] [Revised: 06/29/2023] [Accepted: 09/15/2023] [Indexed: 11/01/2023] Open
Abstract
Heat stroke is a life-threatening disease with high mortality and complications. Endothelial glycocalyx (EGCX) is essential for maintaining endothelial cell structure and function as well as preventing the adhesion of inflammatory cells. Potential relationship that underlies the imbalance in inflammation and coagulation remains elusive. Moreover, the role of EGCX in heat stroke-induced organ injury remained unclear. Therefore, the current study aimed to illustrate if EGCX aggravates apoptosis, inflammation, and oxidative damage in human pulmonary microvascular endothelial cells (HPMEC). Heat stress and lipopolysaccharide (LPS) were employed to construct in vitro models to study the changes of glycocalyx structure and function, as well as levels of heparansulfate proteoglycan (HSPG), syndecan-1 (SDC-1), heparansulfate (HS), tumor necrosis factor-α (TNF-α), interleukin (IL)-6, Von Willebrand factor (vWF), endothelin-1 (ET-1), occludin, E-selectin, vascular cell adhesion molecule-1 (VCAM-1), and reactive oxygen species (ROS). Here, we showed that heat stress and LPS devastated EGCX structure, activated EGCX degradation, and triggered oxidative damage and apoptosis in HPMEC. Stimulation of heat stress and LPS decreased expression of HSPG, increased levels of SDC-1 and HS in culture supernatant, promoted the production and release of proinflammation cytokines (TNF-α and IL-6,) and coagulative factors (vWF and ET-1) in HPMEC. Furthermore, Expressions of E-selection, VCAM-1, and ROS were upregulated, while that of occludin was downregulated. These changes could be deteriorated by heparanase, whereas they meliorated by unfractionated heparin. This study indicated that EGCX may contribute to apoptosis and heat stroke-induced coagulopathy, and these effects may have been due to the decrease in the shedding of EGCX.
Collapse
Affiliation(s)
- Jiadi Chen
- Department of Intensive Care Medicine First WardThe First Affiliated Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Chengjia Ding
- Department of Critical Care Medicine, Binhaiwan Central Hospital of DongguanDongguan Hospital Affiliated to Jinan UniversityDongguanGuangdongChina
- The Key Laboratory for Prevention and Treatment of Critical Illness in Dongguan CityDongguanGuangdongChina
| | - Jingjing Cao
- Department of Critical Care Medicine, Binhaiwan Central Hospital of DongguanDongguan Hospital Affiliated to Jinan UniversityDongguanGuangdongChina
- The Key Laboratory for Prevention and Treatment of Critical Illness in Dongguan CityDongguanGuangdongChina
| | - Huasheng Tong
- Department of Emergency MedicineGeneral Hospital of Southern Theatre Command of PLAGuangzhouGuangdongChina
| | - Yi Chen
- Department of Critical Care Medicine, Binhaiwan Central Hospital of DongguanDongguan Hospital Affiliated to Jinan UniversityDongguanGuangdongChina
- The Key Laboratory for Prevention and Treatment of Critical Illness in Dongguan CityDongguanGuangdongChina
| |
Collapse
|
10
|
Niechoda A, Milewska K, Roslan J, Ejsmont K, Holownia A. Cell cycle-specific phosphorylation of p53 protein in A549 cells exposed to cisplatin and standardized air pollutants. Front Physiol 2023; 14:1238150. [PMID: 37645562 PMCID: PMC10460999 DOI: 10.3389/fphys.2023.1238150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
Exposure to particulate matter is associated with DNA damage and the risk of lung cancer. Protein p53 is activated by multi-site phosphorylation in the early stages of DNA damage and affects cell outcome. Our study aimed to assess the effect of (100 µg/mL-1/24 h) standardized air pollutants: carbon black (CB), urban dust (UD), and nanoparticle carbon black (NPCB) on cell cycle, DNA damage and 53 phosphorylation at Ser 9, Ser 20, Ser 46, and Ser 392 in proliferating and quiescent A549 cells and in cells that survived cisplatin (CisPT) exposure. Phosphorylated p53 was quantified in cell subpopulations by flow cytometry using specific fluorochrome-tagged monoclonal antibodies and analysis of bivariate fluorescence distribution scatterplots. CisPT, UD and NPCB increased site-specific p53 phosphorylation producing unique patterns. NPCB activated all sites irrespectively on the cell cycle, while the UD was more selective. p53 Ser 9-P and p53 Ser 20-P positively correlated with the numbers of CisPT-treated cells at G0/G1, and NPCB and NPCB + CisPT produced a similar effect. A positive correlation and integrated response were also found between Ser 20-P and Ser 392-P in resting A549 cells treated with NPCB and CisPT but not UD. Interdependence between the expression of p53 phosphorylated at Ser 20, and Ser 392 and cell cycle arrest show that posttranslational alterations are related to functional activation. Our data suggest that p53 protein phosphorylation in response to specific DNA damage is driven by multiple independent and integrated pathways to produce functional activation critical in cancer prevention and treatment.
Collapse
Affiliation(s)
| | | | | | | | - Adam Holownia
- Department of Pharmacology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
11
|
Abstract
Regulated cell death predominantly involves apoptosis, autophagy, and regulated necrosis. It is vital that we understand how key regulatory signals can control the process of cell death. Pin1 is a cis-trans isomerase that catalyzes the isomerization of phosphorylated serine or threonine-proline motifs of a protein, thereby acting as a crucial molecular switch and regulating the protein functionality and the signaling pathways involved. However, we know very little about how Pin1-associated pathways might play a role in regulated cell death. In this paper, we review the role of Pin1 in regulated cell death and related research progress and summarize Pin1-related pathways in regulated cell death. Aside from the involvement of Pin1 in the apoptosis that accompanies neurodegenerative diseases, accumulating evidence suggests that Pin1 also plays a role in regulated necrosis and autophagy, thereby exhibiting distinct effects, including both neurotoxic and neuroprotective effects. Gaining an enhanced understanding of Pin1 in neuronal death may provide us with new options for the development of therapeutic target for neurodegenerative disorders.
Collapse
|
12
|
Pan PK, Wang KT, Nan FH, Wu TM, Wu YS. Red Algae “Sarcodia suieae” Acetyl-Xylogalactan Downregulate Heat-Induced Macrophage Stress Factors Ddit3 and Hyou1 Compared to the Aquatic Animal Model of Nile Tilapia (Oreochromis niloticus) Brain Arachidonic Acid Expression. Int J Mol Sci 2022; 23:ijms232314662. [PMID: 36498988 PMCID: PMC9737935 DOI: 10.3390/ijms232314662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Anthropogenic climate change is known to be an increased stress that affects aquatic animal behavior and physiological alternations, which can induce the animal's death. In order to known whether the extracted acetyl-xylogalactan function on the regulation of the external high temperature induced death, we first selected the mammalian cell line "RAW 264.7" used in the previous experiment to evaluate the extracted acetyl-xylogalactan function. We aimed to evaluate the effects of the acetyl-xylogalactan on the RAW 264.7 macrophages and Nile Tilapia stress factor expression under the heat environment. In the in vitro cell observation, we assessed the cell survival, phagocytic activity, intracellular Ca2+ level, mitochondria potential exchange, apoptotic assay findings, galactosidase activity, RNA-seq by NGS and real-time polymerase chain reaction (QPCR) expression. In the in vivo Nile Tilapia observation aimed to evaluate the blood biochemical indicator, brain metabolites exchange and the liver morphology. In our evaluation of RAW 264.7 macrophages, the RNA sequencing and real-time polymerase chain reaction (PCR) was shown to upregulate the expression of the anti-apoptosis Cflar gene and downregulate the expression of the apoptosis factors Ddit3 and Hyou1 to protect macrophages under heat stress. We already knew the extracted acetyl-xylogalactan function on the mammalian "RAW 264.7" system. Following, we used the aquatic Nile Tilapia model as the anthropogenic climate change high temperature experiment. After feeding the Nile Tilapia with the acetyl-xylogalactan, it was found to reduce the brain arachidonic acid (AA) production, which is related to the NF-κB-induced apoptosis mechanism. Combined with the in vitro and in vivo findings, the acetyl-xylogalactan was able to reduce the heat induced cell or tissue stress.
Collapse
Affiliation(s)
- Po-Kai Pan
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Kuang-Teng Wang
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Fan-Hua Nan
- Department of Aquaculture, National Taiwan Ocean University, Keelung 202, Taiwan
| | - Tsung-Meng Wu
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Correspondence: (T.-M.W.); (Y.-S.W.); Tel.: +886-8-7703202 (ext. 6223) (Y.-S.W.)
| | - Yu-Sheng Wu
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Correspondence: (T.-M.W.); (Y.-S.W.); Tel.: +886-8-7703202 (ext. 6223) (Y.-S.W.)
| |
Collapse
|
13
|
Zhang JH, Ni SY, Tan YT, Luo J, Wang SC. A bibliometric analysis of PIN1 and cell death. Front Cell Dev Biol 2022; 10:1043725. [DOI: 10.3389/fcell.2022.1043725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Regulation of cell death plays a key role in numerous diseases. As a proline isomerase, prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) is important for the regulation of signaling pathways. An in-depth understanding of how Pin1 participates in the process of cell death, which affects the occurrence and development of diseases, will aid in the discovery of new disease mechanisms and therapeutic methods. Thus, the purpose of our study was to discover the research trends and hotspots of Pin1 and cell death through bibliometric analyses and to provide insights for understanding the future development of basic research and treatment of diseases.Methods: Documents were extracted from the Web of Science Core Collection on 7 May 2022. We selected articles and reviews published in English from 2000 to 2021, and visual and statistical analyses of countries, institutions, authors, references and keywords were performed using VOSviewer 1.6.18 and CiteSpace 5.8.Results: A total of 395 articles and reviews were selected. Since 2001, the number of articles on Pin1 and cell death has increased annually. Publications come from 43 countries, with the US having the most publications and citations. We identified 510 authors, with Giannino Del Sal having the most articles and Paola Zacchi having the most co-citations. The Journal of Biological Chemistry is the most researched journal, and Nature and its subjournals are the most cited journals. Apoptosis, phosphorylation, and breast cancer were the three most common keywords.Conclusion: The number of documents showed an increasing trend from 2001 to 2014. Stagnant growth after 2014 may be related to the absence of new research hotspots. Cooperative links between core institutions need to be strengthened, and the institution with the highest citation count in recent years is Fujian Medical University in China. The role of Pin1 in cell death requires further research to discover new research hotspots. Before breakthroughs in molecular mechanism or signaling pathway research, future research will focus more on the treatment of diseases represented by Pin1 inhibitors.
Collapse
|
14
|
Wang Y, Zhang Z, Mi X, Li M, Huang D, Song T, Qi X, Yang M. Elevation of effective p53 expression sensitizes wild-type p53 breast cancer cells to CDK7 inhibitor THZ1. Cell Commun Signal 2022; 20:96. [PMID: 36058938 PMCID: PMC9442925 DOI: 10.1186/s12964-022-00837-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/03/2022] [Indexed: 12/02/2022] Open
Abstract
Background The cyclin-dependent kinase 7 (CDK7) inhibitor THZ1 represses multiple cancer cells. However, its tumor-repressive efficiency in wild-type p53 breast cancer cells remains controversial. Methods We conducted various assays, including CCK8, colony formation, flow cytometry, western blotting, and lactate dehydrogenase release detection, to clarify whether p53 elevation sensitizes breast cancer cells to THZ1. Results We found that upregulating functional p53 contributes to the increased sensitivity of breast cancer cells to THZ1. Increased THZ1 sensitivity requires active p53 and an intact p53 pathway, which was confirmed by introducing exogenous wild-type p53 and the subsequent elevation of THZ1-mediated tumor suppression in breast cancer cells carrying mutant p53. We confirmed that p53 accumulates in the nucleus and mitochondria during cell death. Furthermore, we identified extensive transcriptional disruption, rather than solely CDK7 inhibition, as the mechanism underlying the nutlin-3 and THZ1-induced death of breast cancer cells. Finally, we observed the combined nutlin-3 and THZ1 treatment amplified gasdermin E cleavage. Conclusion Enhanced sensitivity of breast cancer cells to THZ1 can be achieved by increasing effective p53 expression. Our approach may serve as a potential treatment for patients with breast cancer resistant to regular therapies. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00837-z.
Collapse
Affiliation(s)
- Yueyuan Wang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhihao Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xuguang Mi
- Tumor Biotherapy Center, Jilin Province People's Hospital, Changchun, 130021, Jilin, Republic of China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Dan Huang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Tingting Song
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xiaoyan Qi
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Ming Yang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
15
|
Zhang H, He Z, Deng P, Lu M, Zhou C, Yang L, Yu Z. PIN1-mediated ROS production is involved in antagonism of N-acetyl-L-cysteine against arsenic-induced hepatotoxicity. Toxicol Res (Camb) 2022; 11:628-643. [PMID: 36051664 PMCID: PMC9424717 DOI: 10.1093/toxres/tfac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/09/2022] [Accepted: 06/24/2022] [Indexed: 08/26/2023] Open
Abstract
Arsenic, a widely existing environmental contaminant, is recognized to be toxic to multiple organs. Exposure to arsenic results in liver damage via excessive production of reactive oxidative species (ROS). PIN1 regulates the levels of ROS. N-acetyl-L-cysteine (NAC) is an ROS scavenger that protects the hepatic functions. Whether PIN1 plays a regulatory role in NAC-mediated antagonism against arsenic hepatotoxicity remains largely unknown. In our study, the protective effects of NAC against arsenic (NaAsO2)-induced hepatotoxicity were evaluated in vitro and in vivo. Arsenic exposure induced cytotoxicity by increasing the intracellular ROS production, impairing mitochondrial function and inducing apoptosis in L02 hepatocytes. Overexpression of PIN1 markedly protected against arsenic cytotoxicity, decreased ROS levels, and mitigated mitochondrial dysfunction and apoptosis in L02 cells. However, loss of PIN1 further aggravated arsenic-induced cytotoxicity and abolished the protective effects of NAC in L02 cells. An in vivo study showed that pretreatment with NAC rescued arsenic-induced liver injury by restoring liver function and suppressing hepatic oxidative stress. Overexpression of PIN1 in mice transfected with AAV-Pin1 relieved arsenic-induced liver dysfunction and hepatic oxidative stress. Taken together, our study identified PIN1 as a novel intervention target for antagonizing arsenic-induced hepatotoxicity, highlighting a new pharmacological mechanism of NAC targeting PIN1 in antagonism against arsenic toxicity.
Collapse
Affiliation(s)
- Huijie Zhang
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Zhixin He
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Muxue Lu
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Chao Zhou
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Zhengping Yu
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| |
Collapse
|
16
|
Chen H, Lin X, Yi X, Liu X, Yu R, Fan W, Ling Y, Liu Y, Xie W. SIRT1-mediated p53 deacetylation inhibits ferroptosis and alleviates heat stress-induced lung epithelial cells injury. Int J Hyperthermia 2022; 39:977-986. [PMID: 35853732 DOI: 10.1080/02656736.2022.2094476] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
OBJECTIVE Acute lung injury (ALI) is a common complication of heat stroke (HS) and a direct cause of death. However, the mechanism underlying ALI following HS remains unclear. METHOD To investigate whether ferroptosis is involved in HS-ALI. We established a HS model of mice and mouse lung epithelial-2 cells (MLE-2). The severity of lung injury was measured by H&E staining, the wet-to-dry lung weight ratio, and Transmission electron microscopy. Potential markers of ferroptosis Fe2+, malondialdehyde (MDA), hydroxynonenal (4-HNE) and lipid peroxidation were detected. The percentages of cell death and viability induced by HS were assessed by LDH and CCK8 assays. SLC7A11, ACSL4, GPX4, SIRT1, p53, and p53 K382 acetylation levels were measured by Western blot. RESULTS The administration of ferroptosis inhibitor ferrostatin-1(Fer-1) could significantly ameliorate lung injury, inhibiting levels of MDA and 4-HNE, and ameliorating HS-induced increased ACSL4, decreased SLC7A11 and GPX4, suggesting ferroptosis was involved in HS-induced ALI in vivo and in vitro. Moreover, SIRT1 expression decreased, and p53 K382 acetylation levels increased in MLE-2 cells. Activation of SIRT1 could improve lung epithelial ferroptosis caused by HS in vivo ang in vitro. Besides, the activation of SIRT1 could significantly reduce the p53 K382 acetylation levels, suggesting that activation of SIRT1 could prevent ferroptosis via inhibiting p53 acetylation. CONCLUSION These findings substantiate the vital role of the SIRT1/p53 axis in mediating ferroptosis in HS-ALI, suggesting that targeting SIRT1 may represent a novel therapeutic strategy to ameliorate ALI during HS.
Collapse
Affiliation(s)
- Hui Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiaoping Lin
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiaohong Yi
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, P.R. China
| | - Xiaofeng Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ranghui Yu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Wenhao Fan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yaping Ling
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
17
|
At the Crossroads of Life and Death: The Proteins That Influence Cell Fate Decisions. Cancers (Basel) 2022; 14:cancers14112745. [PMID: 35681725 PMCID: PMC9179324 DOI: 10.3390/cancers14112745] [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: 05/16/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cellular senescence and apoptosis were historically thought of as two distinct cell fate pathways. However, many of the proteins involved are integral to both pathways. In particular, the ability of p53 to regulate both senescence and apoptosis meant it was seen as the decisive factor in these decisions, yet questions remain about its ability to select on its own the most appropriate cell fate according to each situation. Therefore, cell fates are no longer considered fixed endpoints but dynamic states that can be shifted given the right combination of activation and/or inhibitions of cofactors. Abstract When a cell is damaged, it must decide how to respond. As a consequence of a variety of stresses, cells can induce well-regulated programmes such as senescence, a persistent proliferative arrest that limits their replication. Alternatively, regulated programmed cell death can be induced to remove the irreversibly damaged cells in a controlled manner. These programmes are mainly triggered and controlled by the tumour suppressor protein p53 and its complex network of effectors, but how it decides between these wildly different responses is not fully understood. This review focuses on the key proteins involved both in the regulation and induction of apoptosis and senescence to examine the key events that determine cell fate following damage. Furthermore, we examine how the regulation and activity of these proteins are altered during the progression of many chronic diseases, including cancer.
Collapse
|
18
|
Huang W, Xie W, Zhong H, Cai S, Huang Q, Liu Y, Zeng Z, Liu Y. Cytosolic p53 Inhibits Parkin-Mediated Mitophagy and Promotes Acute Liver Injury Induced by Heat Stroke. Front Immunol 2022; 13:859231. [PMID: 35634298 PMCID: PMC9139682 DOI: 10.3389/fimmu.2022.859231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/20/2022] [Indexed: 11/25/2022] Open
Abstract
Heat stroke (HS) is a severe condition characterized by increased morbidity and high mortality. Acute liver injury (ALI) is a well-documented complication of HS. The tumor suppressor p53 plays an important role in regulation of mitochondrial integrity and mitophagy in several forms of ALI. However, the role of p53-regulated mitophagy in HS-ALI remains unclear. In our study, we discovered the dynamic changes of mitophagy in hepatocytes and demonstrated the protective effects of mitophagy activation on HS-ALI. Pretreatment with 3-MA or Mdivi-1 significantly exacerbated ALI by inhibiting mitophagy in HS-ALI mice. Consistent with the animal HS-ALI model results, silencing Parkin aggravated mitochondrial damage and apoptosis by inhibiting mitophagy in HS-treated normal human liver cell line (LO2 cells). Moreover, we described an increase in the translocation of p53 from the nucleus to the cytoplasm, and cytosolic p53 binds to Parkin in LO2 cells following HS. p53 overexpression using a specific adenovirus or Tenovin-6 exacerbated HS-ALI through Parkin-dependent mitophagy both in vivo and in vitro, whereas inhibition of p53 using siRNA or PFT-α effectively reversed this process. Our results demonstrate that cytosolic p53 binds to Parkin and inhibits mitophagy by preventing Parkin's translocation from the cytosol to the mitochondria, which decreases mitophagy activation and leads to hepatocyte apoptosis in HS-ALI. Overall, pharmacologic induction of mitophagy by inhibiting p53 may be a promising therapeutic approach for HS-ALI treatment.
Collapse
Affiliation(s)
- Wei Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanhui Zhong
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Huang W, Mao L, Xie W, Cai S, Huang Q, Liu Y, Chen Z. Impact of UCP2 depletion on heat stroke-induced mitochondrial function in human umbilical vein endothelial cells. Int J Hyperthermia 2022; 39:287-296. [PMID: 35129048 DOI: 10.1080/02656736.2022.2032846] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Wei Huang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, P.R. China
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Liangfeng Mao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, P.R. China
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Sumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, P.R. China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Zhongqing Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, P.R. China
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
20
|
Stanculescu D, Sepúlveda N, Lim CL, Bergquist J. Lessons From Heat Stroke for Understanding Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front Neurol 2021; 12:789784. [PMID: 34966354 PMCID: PMC8710546 DOI: 10.3389/fneur.2021.789784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/11/2021] [Indexed: 01/01/2023] Open
Abstract
We here provide an overview of the pathophysiological mechanisms during heat stroke and describe similar mechanisms found in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Both conditions are characterized by disturbed homeostasis in which inflammatory pathways play a central role. Splanchnic vasoconstriction, increased gut permeability, gut-related endotoxemia, systemic inflammatory response, central nervous system dysfunction, blood coagulation disorder, endothelial-cell injury, and mitochondrial dysfunction underlie heat stroke. These mechanisms have also been documented in ME/CFS. Moreover, initial transcriptomic studies suggest that similar gene expressions are altered in both heat stroke and ME/CFS. Finally, some predisposing factors for heat stroke, such as pre-existing inflammation or infection, overlap with those for ME/CFS. Notwithstanding important differences - and despite heat stroke being an acute condition - the overlaps between heat stroke and ME/CFS suggest common pathways in the physiological responses to very different forms of stressors, which are manifested in different clinical outcomes. The human studies and animal models of heat stroke provide an explanation for the self-perpetuation of homeostatic imbalance centered around intestinal wall injury, which could also inform the understanding of ME/CFS. Moreover, the studies of novel therapeutics for heat stroke might provide new avenues for the treatment of ME/CFS. Future research should be conducted to investigate the similarities between heat stroke and ME/CFS to help identify the potential treatments for ME/CFS.
Collapse
Affiliation(s)
| | - Nuno Sepúlveda
- CEAUL—Centro de Estatística e Aplicações da Universidade de Lisboa, Lisbon, Portugal
- Department of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Chin Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jonas Bergquist
- Analytical Chemistry and Neurochemistry, Department of Chemistry—BMC, Uppsala University, Uppsala, Sweden
- The ME/CFS Collaborative Research Center at Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Fagiani F, Vlachou M, Di Marino D, Canobbio I, Romagnoli A, Racchi M, Govoni S, Lanni C. Pin1 as Molecular Switch in Vascular Endothelium: Notes on Its Putative Role in Age-Associated Vascular Diseases. Cells 2021; 10:cells10123287. [PMID: 34943794 PMCID: PMC8699654 DOI: 10.3390/cells10123287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 01/04/2023] Open
Abstract
By controlling the change of the backbones of several cellular substrates, the peptidyl-prolyl cis-trans isomerase Pin1 acts as key fine-tuner and amplifier of multiple signaling pathways, thereby inducing several biological consequences, both in physiological and pathological conditions. Data from the literature indicate a prominent role of Pin1 in the regulating of vascular homeostasis. In this review, we will critically dissect Pin1’s role as conformational switch regulating the homeostasis of vascular endothelium, by specifically modulating nitric oxide (NO) bioavailability. In this regard, Pin1 has been reported to directly control NO production by interacting with bovine endothelial nitric oxide synthase (eNOS) at Ser116-Pro117 (human equivalent is Ser114-Pro115) in a phosphorylation-dependent manner, regulating its catalytic activity, as well as by regulating other intracellular players, such as VEGF and TGF-β, thereby impinging upon NO release. Furthermore, since Pin1 has been found to act as a critical driver of vascular cell proliferation, apoptosis, and inflammation, with implication in many vascular diseases (e.g., diabetes, atherosclerosis, hypertension, and cardiac hypertrophy), evidence indicating that Pin1 may serve a pivotal role in vascular endothelium will be discussed. Understanding the role of Pin1 in vascular homeostasis is crucial in terms of finding a new possible therapeutic player and target in vascular pathologies, including those affecting the elderly (such as small and large vessel diseases and vascular dementia) or those promoting the full expression of neurodegenerative dementing diseases.
Collapse
Affiliation(s)
- Francesca Fagiani
- Pharmacology Section, Department of Drug Sciences, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (M.V.); (M.R.); (C.L.)
| | - Marieva Vlachou
- Pharmacology Section, Department of Drug Sciences, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (M.V.); (M.R.); (C.L.)
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (D.D.M.); (A.R.)
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy;
| | - Alice Romagnoli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy; (D.D.M.); (A.R.)
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Marco Racchi
- Pharmacology Section, Department of Drug Sciences, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (M.V.); (M.R.); (C.L.)
| | - Stefano Govoni
- Pharmacology Section, Department of Drug Sciences, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (M.V.); (M.R.); (C.L.)
- Correspondence:
| | - Cristina Lanni
- Pharmacology Section, Department of Drug Sciences, University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy; (F.F.); (M.V.); (M.R.); (C.L.)
| |
Collapse
|
22
|
李 莉, 邹 志, 李 琴, 张 堃, 苏 磊, 古 正. [Extranuclear p53 suppresses autophagy through AMPK/mTOR signaling to promote heat stress-induced vascular endothelial cell damage]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1664-1671. [PMID: 34916192 PMCID: PMC8685697 DOI: 10.12122/j.issn.1673-4254.2021.11.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To explore the role of extranuclear p53-mediated autophagy suppression by regulating AMPK/mTOR signaling pathway in heat stress (HS)-induced injury of mouse aortic endothelial cells (MAECs). METHODS Primary cultures of MAECs were pretreated with compound C (an AMPK inhibitor), rapamycin (a mTOR inhibitor) or pifithrin-α (PFT, a selective p53 inhibitor) for 1 h before exposure to HS (43 ℃) for 2 h. The changes in cell viability at different time points after HS were examined using CCK-8 assay, and the protein expressions of P53, LC3-II, Beclin-1, p62 and the AMPK/mTOR signaling proteins were detected using Western blotting. In the animal experiment, C57 mice were pretreated with compound C, rapamycin or PFT and exposed to a high temperature at 40 ℃ to induce HS. The pathological changes in the aorta of the mice were observed with HE staining, and cell apoptosis was detected using TUNEL staining. RESULTS In cultured MAECs, the cell viability was significantly reduced (P < 0.05) and the mitochondrial fraction of p53 increased while its cytoplasmic fraction decreased progressively over time following HS. HS significantly lowered the expressions of LC3-II and Beclin-1, increased p62 level, suppressed AMPK phosphorylation, and increased mTOR phosphorylation and the expressions of its downstream proteins at 6 h after the exposure (P < 0.05). Pretreatment with compound C significantly inhibited LC3-II and Beclin- 1 expression, enhanced p62 expression, and aggravated HS-induced cell injury and apoptosis in MAECs; rapamycin treatment produced the opposite effects (P < 0.05). PFT treatment significantly enhanced the viability of MAECs and alleviated HSinduced injury and apoptosis; PFT also significantly promoted activation of AMPK phosphorylation, inhibited mTOR phosphorylation and its downstream proteins (P < 0.05), enhanced the expressions of LC3-II and Beclin 1, and inhibited p62 expression in the MAECs (P < 0.05). In C57 mice, HS resulted in swelling, shedding and apoptosis of aortic vascular endothelial cells. Pretreatment with compound C obviously aggravated HS-induced vascular injury and endothelial cell apoptosis, while pretreatment with either rapamycin or PFT significantly alleviated these injuries. CONCLUSION Autophagy inhibition mediated by extranuclear p53 via inhibiting AMPK activity and activating mTOR signaling participates in HS-induced injury of MAECs.
Collapse
Affiliation(s)
- 莉 李
- 南方医科大学第三附属医院创伤救治中心,广东 广州 510630Treatment Center for Traumatic Injuries, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- 广东省骨科研究院//广东省骨科医院//广东省骨与关节退行性疾病重点实验室,广东 广州 510630Academy of Orthopedics of Guangdong Province//Orthopedic Hospital of Guangdong Province//Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou 510630, China
| | - 志敏 邹
- 南方医科大学第三附属医院创伤救治中心,广东 广州 510630Treatment Center for Traumatic Injuries, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- 广东省骨科研究院//广东省骨科医院//广东省骨与关节退行性疾病重点实验室,广东 广州 510630Academy of Orthopedics of Guangdong Province//Orthopedic Hospital of Guangdong Province//Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou 510630, China
| | - 琴 李
- 南方医科大学第三附属医院创伤救治中心,广东 广州 510630Treatment Center for Traumatic Injuries, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- 广东省骨科研究院//广东省骨科医院//广东省骨与关节退行性疾病重点实验室,广东 广州 510630Academy of Orthopedics of Guangdong Province//Orthopedic Hospital of Guangdong Province//Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou 510630, China
| | - 堃 张
- 南方医科大学第三附属医院创伤救治中心,广东 广州 510630Treatment Center for Traumatic Injuries, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- 广东省骨科研究院//广东省骨科医院//广东省骨与关节退行性疾病重点实验室,广东 广州 510630Academy of Orthopedics of Guangdong Province//Orthopedic Hospital of Guangdong Province//Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou 510630, China
| | - 磊 苏
- 中国人民解放军南部战区总医院重症医学科,广东 广州 510010Department of Critical Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - 正涛 古
- 南方医科大学第三附属医院创伤救治中心,广东 广州 510630Treatment Center for Traumatic Injuries, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- 广东省骨科研究院//广东省骨科医院//广东省骨与关节退行性疾病重点实验室,广东 广州 510630Academy of Orthopedics of Guangdong Province//Orthopedic Hospital of Guangdong Province//Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou 510630, China
| |
Collapse
|
23
|
Xie W, Huang W, Cai S, Chen H, Fu W, Chen Z, Liu Y. NF‑κB/IκBα signaling pathways are essential for resistance to heat stress‑induced ROS production in pulmonary microvascular endothelial cells. Mol Med Rep 2021; 24:814. [PMID: 34558646 PMCID: PMC8477608 DOI: 10.3892/mmr.2021.12454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022] Open
Abstract
The results of a previous study demonstrated that heat stress (HS) triggered oxidative stress, which in turn induced the apoptosis of epithelial cells. These results uncovered a novel mechanism underlying the activation of NF-κB in primary human umbilical vein endothelial cells. The present study aimed to further investigate the role of NF-κB/IκBα signaling pathways in the inhibition of HS-induced reactive oxygen species (ROS) generation and cytotoxicity in endothelial cells. The results of the present study demonstrated that HS triggered a significant amount of NF-κB and IκBα nuclear translocation without IκBα degradation in a time-dependent manner. Mutant constructs of IκBα phosphorylation sites (Ser32, Ser36) were employed in rat pulmonary microvascular endothelial cells (PMVECs). Cell Counting Kit-8 assays demonstrated that both the small interfering (si)RNA-mediated knockdown of p65 and IκBα mutant constructs significantly decreased cell viability and aggravated ROS accumulation in HS-induced rat PMVECs compared with the control. Additionally, western blot analysis revealed that p65 siRNA attenuated the protein expression of IκBα. However, IκBα mutant constructs failed to attenuate NF-κB activation and nuclear translocation, indicating that IκBα-independent pathways contributed to NF-κB activity and nucleus translocation in a time-dependent manner following HS. Collectively, the results of the present study suggested that the NF-κB/IκBα pathway was essential for resistance to HS-induced ROS production and cytotoxicity in rat PMVECs, and that it could be a potential therapeutic target to reduce the mortality and morbidity of heat stroke.
Collapse
Affiliation(s)
- Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wei Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hui Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Weijun Fu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
24
|
Wan H, Yang Y, Li Z, Cheng L, Ding Z, Wan H, Yang J, Zhou H. Compatibility of ingredients of Danshen (Radix Salviae Miltiorrhizae) and Honghua ( Flos Carthami) and their protective effects on cerebral ischemia-reperfusion injury in rats. Exp Ther Med 2021; 22:849. [PMID: 34149895 PMCID: PMC8210257 DOI: 10.3892/etm.2021.10281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/23/2021] [Indexed: 12/17/2022] Open
Abstract
Danshen (Radix Salviae Miltiorrhizae) and Honghua (Flos Carthami) (Danhong) are two drugs commonly prescribed together, which are often used in the treatment of cerebrovascular diseases in China. Due to the complexity of the ingredients of Danhong, the present study focused on performing the orthogonal compatibility method on the primary effective molecules of this drug: Tanshinol, salvianolic acid A, salvianolic acid B and hydroxysafflor yellow A. These four molecules were studied to determine their protective effects and to screen for the most compatible ingredients to improve cerebral ischemia-reperfusion injury (IR) in rats. Focal middle cerebral artery occlusion was performed to establish the cerebral IR model in rats. Male Sprague-Dawley rats were randomly divided into sham operation group, IR group and nine orthogonal administration groups with different ratios of Danhong effective ingredients and Danhong injection group. Neurological deficit score and cerebral infarction volume were measured postoperatively. Morphological pathological alterations were observed via H&E staining. Bcl-2 and Bax were quantified using ELISA. Immunohistochemistry was conducted to analyze the expression of caspase-3 in the hippocampus. The expression levels of cytochrome c, apoptotic peptidase activating factor 1 (apaf-1), caspase-9, caspase-3 and p53 mRNA in the hippocampus were assessed via reverse transcription-quantitative PCR. The results demonstrated that different compatibility groups significantly reduced the neurological function score and decreased the volume of cerebral infarct compared with the IR group. These groups were also indicated to improve the pathological damage to the brain tissue. In addition, certain compatibility groups significantly decreased the number of caspase-3 positive cells in the hippocampus and the expression levels of cytochrome c, apaf-1, caspase-9, caspase-3 and p53 mRNA in the brain tissue. Orthogonal group 4 (30 mg/kg tanshinol; 2.5 mg/kg salvianolic acid A; 16 mg/kg salvianolic acid B; 8 mg/kg hydroxysafflor yellow A) was indicated to be the most effective. The four effective ingredients of Danhong exhibited a protective effect on rats with cerebral IR injury, potentially through the inhibition of apoptosis via the downregulation of key targets upstream of the caspase-3 pathway. In addition, the present study provided novel insights for the continued study of the drug compatibility rules of TCM.
Collapse
Affiliation(s)
- Haoyu Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yuting Yang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhiwei Li
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Lan Cheng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhishan Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jiehong Yang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Huifen Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
25
|
Du X, Liu H, Liu X, Chen X, Yuan L, Ma Y, Huang H, Wang Y, Wang R, Zhang S, Tian Z, Shi L, Zhang H. Microcystin-LR induces ovarian injury and apoptosis in mice via activating apoptosis signal-regulating kinase 1-mediated P38/JNK pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 213:112066. [PMID: 33610944 DOI: 10.1016/j.ecoenv.2021.112066] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
As an emerging pollutant in the aquatic environment, microcystin-LR (MC-LR) can enter the body through multiple pathways, and then induce apoptosis and gonadal damage, affecting reproductive function. Previous studies focused on male reproductive toxicity induced by MC-LR neglecting its effects on females. The apoptotic signal-regulated kinase 1 (ASK1) is an upstream protein of P38/JNK pathway, closely associated with apoptosis and organ damage. However, the role of ASK1 in MC-LR-induced reproductive toxicity is unclear. Therefore, this study investigated the role of ASK1 in mouse ovarian injury and apoptosis induced by MC-LR. After MC-LR exposure, ASK1 expression in mouse ovarian granulosa cells was increased at the protein and mRNA levels, and decreased following pretreatment by antioxidant N-acetylcysteine, suggesting that MC-LR-induced oxidative stress has a regulatory role in ASK1 expression. Inhibition of ASK1 expression with siASK1 and NQDI-1 could effectively alleviate MC-LR-induced mitochondrial membrane potential damage and apoptosis in ovarian granulosa cells, as well as pathological damage, apoptosis and the decreased gonadal index in ovaries of C57BL/6 mice. Moreover, the P38/JNK pathway and downstream apoptosis-related proteins (P-P38, P-JNK, P-P53, Fas) and genes (MKK4, MKK3, Ddit3, Mef2c) were activated in vivo and vitro, but their activation was restrained after ASK1 inhibition. Data presented herein suggest that the ASK1-mediated P38/JNK pathway is involved in ovarian injury and apoptosis induced by MC-LR in mice. It is confirmed that ASK1 has an important role in MC-LR-induced ovarian injury, which provides new insights for preventing MCs-induced reproductive toxicity in females.
Collapse
Affiliation(s)
- Xingde Du
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Haohao Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaohui Liu
- School of Basic Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xinghai Chen
- Department of Chemistry and Biochemistry, St Mary's University, San Antonio, TX 78228, USA
| | - Le Yuan
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Ya Ma
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Hui Huang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yueqin Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Rui Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shiyu Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhihui Tian
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Linjia Shi
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
26
|
Li L, Tan H, Zou Z, Gong J, Zhou J, Peng N, Su L, Maegele M, Cai D, Gu Z. Preventing necroptosis by scavenging ROS production alleviates heat stress-induced intestinal injury. Int J Hyperthermia 2020; 37:517-530. [PMID: 32423248 DOI: 10.1080/02656736.2020.1763483] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Worldwide heat stroke incidence has increased in recent years and is associated with high morbidity and mortality. Therefore, it is critical to identify mechanisms that mediate heat stroke. Previous studies suggested that damage to the small intestine may be a major factor in heat stroke-related morbidity and mortality. However, the mechanism underlying heat stroke related small intestine injury remains unclear.Methods: To explore how heat stroke promotes intestinal damage, we applied two well established models: mouse and IEC-6 cells heat stress (HS) to mimic heat stroke both in vivo and in vitro. The percentages of viability and cell death were assessed by WST-1 and LDH release assays. Induction of HS-induced cell death was analyzed by flow cytometry with Annexin V-FITC/PI staining. Flow cytometry was used to analyze HS-induced mitochondrial superoxide with MitoSOX staining. Malondialdehyde (MDA) levels and superoxide dismutase (SOD) levels were detected by ELISA. Flow cytometry was used to analyze HS-induced mitochondrial depolarization (low ΔΨm) with JC-1 staining. Histopathology changes in the ileum were detected by H&E staining.The ileum ultrastructure was observed by transmission electron microscopy (TEM). RIPK1, RIPK3, phosphorylated MLKL, and MLKL levels were detected by Western blot. RIPK1-RIPK3 complexes were measured by immunoprecipitation assay.Results: HS increased both necrotic cell rate and RIPK1, RIPK3, and phosphorylated MLKL expression levels in IEC-6 cells. These increased expression levels promoted higher RIPK1-RIPK3 complex formation, leading to necrosome formation both in vivo and in vitro. Moreover, HS caused dyshomeostasis, an oxidative stress response, and mitochondrial damage, along with small intestinal tissue injury and cell death. However, IEC-6 cells or mice pretreated with the RIPK1 activity chemical inhibitor Nec-1 or RIPK3 activity chemical inhibitor GSK'872 significantly reversed these phenomena and promoted balance in oxidative stress response homeostasis. More importantly, the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) pretreatment significantly inhibited HS-induced RIPK1/RIPK3-dependent necroptosis formation both in vivo and in vitro, suggesting that preventing necroptosis via scavenging ROS production might alleviate HS-induced small intestinal tissue injury and cell death.Conclusion: This study provides strong evidence that HS causes damage to both the small intestine and intestinal epithelial cells, scavenging ROS production can significantly alleviate such RIPK1/RIPK3-dependent necroptosis, mediating HS-induced intestinal damage both in vitro and in vivo. These findings provide a clear target for future mechanism-based therapeutic strategies for patients diagnosed with heat stroke.
Collapse
Affiliation(s)
- Li Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China
| | - Hongping Tan
- Department of epilepsy centre, Guangdong Sanjiu Brain Hospital, Guangzhou China
| | - Zhimin Zou
- Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China
| | - Jian Gong
- Department of Intensive Care Unit, Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Junjie Zhou
- Department of Intensive Care Unit, Heyuan People's Hospital, Heyuan, Guangdong, China
| | - Na Peng
- Department of Intensive Care Unit, General Hospital of Southern Theater Command, PLA; Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, Guangzhou, Guangdong, China
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Southern Theater Command, PLA; Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, Guangzhou, Guangdong, China
| | - Marc Maegele
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Treatment Center for Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center (CMMC), University Witten/Herdecke (UW/H), Cologne, Germany
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhengtao Gu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathophysiology, Southern Medical University, Guangdong provincial key laboratory of shock and microcirculation research, Guangzhou, Guangdong, China.,Department of Treatment Center for Traumatic Injuries, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Huang W, Xie W, Gong J, Wang W, Cai S, Huang Q, Chen Z, Liu Y. Heat stress induces RIP1/RIP3-dependent necroptosis through the MAPK, NF-κB, and c-Jun signaling pathways in pulmonary vascular endothelial cells. Biochem Biophys Res Commun 2020; 528:206-212. [PMID: 32471717 DOI: 10.1016/j.bbrc.2020.04.150] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/29/2020] [Indexed: 01/30/2023]
Abstract
Necroptosis represents a newly defined form of regulated necrosis and participates in various human inflammatory diseases. It remains unclear whether necroptosis is presented in heatstroke-induced lung injury. We show that heat stress(HS) triggered an significant upregulation of receptor-interacting protein 1 (RIP1) and mixed lineage kinase domain-like protein (MLKL) expression in a time-dependent manner, without a significant change of receptor-interacting protein 3 (RIP3). Furthermore, co-immunoprecipitation assays showed that RIP1 binds to RIP3 to form the necrosome in heat stress-induced PMVECs. In vitro, necrostatin-1 (Nec-1) pre-treatment reduced heat stress-induced PMVECs necroptosis, which also inhibited HMGB1 translocation from the nucleus into the cytoplasm. Similarly, inhibition for ERK (PD98059), NF-κB (BAY11-7082) and c-Jun (c-Jun peptide), respectively, also suppressed the HMGB1 cytoplasm translocation. Furthermore, siRNA-mediated RIP1/RIP3 knockdown negatively regulated the release of HMGB1 in HS-induced necroptosis through the ERK, NF-κB, and c-Jun signaling pathways. Our study reveals that HS induces RIP1/RIP3-dependent necroptosis through the MAPK, NF-κB, and c-Jun signaling pathways in PMVECs.
Collapse
Affiliation(s)
- Wei Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jian Gong
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Intensive Care Medicine, The Third People's Hospital of Longgang District, Shenzhen, 518115, China
| | - Wenyan Wang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
28
|
Liu Z, Zhao W, Yuan P, Zhu P, Fan K, Xia Z, Xu S. The mechanism of CaMK2α-MCU-mitochondrial oxidative stress in bupivacaine-induced neurotoxicity. Free Radic Biol Med 2020; 152:363-374. [PMID: 32275945 DOI: 10.1016/j.freeradbiomed.2020.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 01/09/2023]
Abstract
Ca2+/calmodulin dependent protein kinase2α (CaMK2α) is a serine/threonine protein kinase in neurons and leads to neuronal injury when it is activated abnormally. Bupivacaine, a local anesthetic commonly used in regional nerve block, could induce neurotoxicity via apoptotic injury. Whether or not CaMK2α is involved in bupivacaine-induced neurotoxicity and it is regulated remains unclear. In this study, bupivacaine was administered for intrathecal injection in C57BL/6 mice for building vivo injury model and was used to culture human neuroblastoma (SH-SY5Y) cells for building vitro injury model. The results showed that bupivacaine induced mitochondrial oxidative stress and neurons apoptotic injury, promoted phosphorylation of CaMK2α and cAMP-response element binding protein (CREB), and elevated mitochondrial Ca2+ uniporter (MCU) expression. Furthermore, it induced CaMK2α phosphorylation at Thr286 which phosphorylated CREB at Ser133 and up-regulated MCU transcriptional expression. Inhibition of CaMK2α-MCU signaling with knock-down of CaMK2α and MCU or with inhibitors (KN93 and Ru360) significantly mitigated bupivacaine-induced neurotoxic injury. Over-expression of CaMK2α significantly enhanced above oxidative injury. Activated MCU with agonist (spermine) reversed protective effect of siCaMK2α on bupivacaine-induced mitochondrial oxidative stress. Our data revealed that CaMK2α-MCU-mitochondrial oxidative stress pathway is a major mechanism whereby bupivacaine induces neurotoxicity and inhibition of above signaling could be a therapeutic strategy in the treatment of bupivacaine-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhongjie Liu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Wei Zhao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Pengfei Yuan
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Pian Zhu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Keke Fan
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China; Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA, 95817, USA.
| | - Shiyuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China.
| |
Collapse
|
29
|
Liebl MC, Hofmann TG. Cell Fate Regulation upon DNA Damage: p53 Serine 46 Kinases Pave the Cell Death Road. Bioessays 2019; 41:e1900127. [PMID: 31621101 DOI: 10.1002/bies.201900127] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Indexed: 12/20/2022]
Abstract
Mild and massive DNA damage are differentially integrated into the cellular signaling networks and, in consequence, provoke different cell fate decisions. After mild damage, the tumor suppressor p53 directs the cellular response to cell cycle arrest, DNA repair, and cell survival, whereas upon severe damage, p53 drives the cell death response. One posttranslational modification of p53, phosphorylation at Serine 46, selectively occurs after severe DNA damage and is envisioned as a marker of the cell death response. However, the molecular mechanism of action of the p53 Ser46 phospho-isomer, the molecular timing of this phosphorylation event, and its activating effects on apoptosis and ferroptosis still await exploration. In this essay, the current body of evidence on the molecular function of this deadly p53 mark, its evolutionary conservation, and the regulation of the key players of this response, the p53 Serine 46 kinases, are reviewed and dissected.
Collapse
Affiliation(s)
- Magdalena C Liebl
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg, University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - Thomas G Hofmann
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg, University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| |
Collapse
|