1
|
Chen Y, Li M, Wu Y. Heat shock protein 22: A new direction for cardiovascular disease (Review). Mol Med Rep 2025; 31:82. [PMID: 39886946 PMCID: PMC11800183 DOI: 10.3892/mmr.2025.13447] [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: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Small heat shock proteins (sHSPs) are common molecular chaperone proteins that function in various biological processes, and serve indispensable roles in maintaining cellular protein homeostasis and regulating the hydrolysis of unfolded proteins. HSP22 is a member of the sHSP family that is primarily expressed in the heart and skeletal muscle, as well as in various types of cancer. There have been important findings concerning the role of HSP22 in cardiovascular diseases. The aim of the present study was to provide insights into the various molecular mechanisms by which HSP22 functions in the heart, including oxidative stress, autophagy, apoptosis, the subcellular distribution of proteins and the promoting effect of proteasomes. In addition, drugs and cytokines, including geranylgeranylacetone, can exert protective effects on the heart by regulating the expression of HSP22. Based on increasingly abundant research, HSP22 may be considered a potential therapeutic target in cardiovascular diseases.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Wu R, Wu T, Wang Q, Shi Y, Dong Q, Rong X, Chen M, He Z, Fu Y, Liu L, Shao S, Guan X, Zhang C. The ischemia-enhanced myocardial infarction protection-related lncRNA protects against acute myocardial infarction. MedComm (Beijing) 2024; 5:e632. [PMID: 38988491 PMCID: PMC11234438 DOI: 10.1002/mco2.632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 04/10/2024] [Accepted: 05/14/2024] [Indexed: 07/12/2024] Open
Abstract
Long non-coding RNA RP11-64B16.4 (myocardial infarction protection-related lncRNA [MIPRL]) is among the most abundant and the most upregulated lncRNAs in ischemic human hearts. However, its role in ischemic heart disease is unknown. We found MIPRL was conserved between human and mouse and its expression was increased in mouse hearts after acute myocardial infarction (AMI) and in cultured human and mouse cardiomyocytes after hypoxia. The infarcted size, cardiac cell apoptosis, cardiac dysfunction, and cardiac fibrosis were aggravated in MIPRL knockout mice after AMI. The above adverse results could be reversed by re-expression of MIPRL via adenovirus expressing MIPRL. Both in vitro and in vivo, we identified that heat shock protein beta-8 (HSPB8) was a target gene of MIPRL, which was involved in MIPRL-mediated anti-apoptotic effects on cardiomyocytes. We further discovered that MIPRL could combine with the messenger RNA (mRNA) of HSPB8 and increase its expression in cardiomyocytes by enhancing the stability of HSPB8 mRNA. In summary, we have found for the first time that the ischemia-enhanced lncRNA MIPRL protects against AMI via its target gene HSPB8. MIPRL might be a novel promising therapeutic target for ischemic heart diseases such as AMI.
Collapse
Affiliation(s)
- Rongzhou Wu
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Tingting Wu
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Qiaoyu Wang
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Youyang Shi
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Qianqian Dong
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Xing Rong
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Meiting Chen
- Department of Biomedical EngineeringThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Zhiyu He
- Department of Biomedical EngineeringThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yu Fu
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Lei Liu
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Shuai Shao
- Department of CardiologyKey Laboratory of Medical ElectrophysiologyMinistry of EducationInstitute of Cardiovascular ResearchInstitute of Metabolic Diseasesthe Affiliated Hospital of Southwest Medical UniversitySouthwest Medical UniversityLuzhouChina
| | - Xueqiang Guan
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Chunxiang Zhang
- Children's Heart CenterThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityInstitute of Cardiovascular Development and Translational MedicineThe Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Department of CardiologyKey Laboratory of Medical ElectrophysiologyMinistry of EducationInstitute of Cardiovascular ResearchInstitute of Metabolic Diseasesthe Affiliated Hospital of Southwest Medical UniversitySouthwest Medical UniversityLuzhouChina
| |
Collapse
|
3
|
Cheng J, Ji M, Jing H, Lin H. DUSP12 ameliorates myocardial ischemia-reperfusion injury through HSPB8-induced mitophagy. J Biochem Mol Toxicol 2023; 37:e23310. [PMID: 36644958 DOI: 10.1002/jbt.23310] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
This study aimed to explore the role of dual specificity phosphatase 12 (DUSP12) in regulating myocardial ischemia-reperfusion (I/R) injury and the underlying mechanism. The expression of DUSP12 in myocardial tissues and heat-shock protein beta-8 (HSPB8) and mitophagy-related proteins in myocardial tissues and H9c2 cells were detected by western blot analysis. The serum creatine kinase isoenzymes (CK-MB) and lactate dehydrogenase (LDH), levels of reactive oxygen species and malondialdehyde, superoxide dismutase activity in myocardial tissues and H9c2 cells, and caspase-3 activity in H9c2 cells were analyzed by corresponding assay kits. The infarct area in the rat's heart was observed by triphenyl tetrazolium chloride staining. The apoptosis of myocardial cells in myocardial tissues and H9c2 cells was detected by terminal-deoxynucleotidyl transferase dUTP-biotin nick-end labeling assay. The interaction between DUSP12 and HSPB8 was clarified by the coimmunoprecipitation assay. The transfection efficacy of si-HSPB8#1 and si-HSPB8#2 in H9c2 cells was confirmed by real-time quantitative-polymerase chain reaction and western blot analysis. As a result, DUSP12 expression was downregulated in I/R rats, which was promoted by lentivirus-expressing DUSP12. DUSP12 overexpression reduced the serum creatine kinase isoenzymes (CK-MB) and LDH, decreased the infarct area in the rat's heart, and suppressed the apoptosis and oxidative stress in myocardial tissues. DUSP12 overexpression also upregulated the expression of HSPB8 to promote mitophagy. The coimmunoprecipitation assay indicated that DUSP12 could be combined with HSPB8. In addition, DUSP12 overexpression could inhibit hypoxia/reoxygenation-elicited apoptosis as well as oxidative stress in H9c2 cells by upregulating HSPB8 expression to promote mitophagy, which was countervailed by HSPB8 deficiency. In conclusion, DUSP12 overexpression decreased the apoptosis and oxidative stress in myocardial I/R injury through HSPB8-induced mitophagy.
Collapse
Affiliation(s)
- Jing Cheng
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meihua Ji
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haijuan Jing
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongqi Lin
- Department of Anesthesiology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Sun X, Siri S, Hurst A, Qiu H. Heat Shock Protein 22 in Physiological and Pathological Hearts: Small Molecule, Large Potentials. Cells 2021; 11:cells11010114. [PMID: 35011676 PMCID: PMC8750610 DOI: 10.3390/cells11010114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022] Open
Abstract
Small heat shock protein 22 (HSP22) belongs to the superfamily of heat shock proteins and is predominantly expressed in the heart, brain, skeletal muscle, and different types of cancers. It has been found that HSP22 is involved in variant cellular functions in cardiomyocytes and plays a vital role in cardiac protection against cardiomyocyte injury under diverse stress. This review summarizes the multiple functions of HSP22 in the heart and the underlying molecular mechanisms through modulating gene transcription, post-translational modification, subcellular translocation of its interacting proteins, and protein degradation, facilitating mitochondrial function, cardiac metabolism, autophagy, and ROS production and antiapoptotic effect. We also discuss the association of HSP22 in cardiac pathologies, including human dilated cardiomyopathy, pressure overload-induced heart failure, ischemic heart diseases, and aging-related cardiac metabolism disorder. The collected information would provide insights into the understanding of the HSP22 in heart diseases and lead to discovering the therapeutic targets.
Collapse
|
5
|
Ye Y, Wang X, Yang Z, Xu Q, Zhang B. Hsp22 Inhibits Oxidative Stress-Induced Endplate Chondrocyte Apoptosis by Regulating Mitochondrial Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: Facet joint degeneration (FJD), which is also called facet joint syndrome (FJS), has become one of the most commonly seen etiological factors for lumbago. Cartilage lesion triggered by lumbar facet joint (LFJ) degeneration might be related to mitochondrial impairment,
but the its underlying mechanism remains unclear. Materials and methods: The endplate chondrocytes were induced by hydrogen peroxide (H2O2) to mimic the pathological conditions of oxidative stress. Enzyme linked immunosorbent assay (ELISA) were used for the evaluation
of reactive oxygen species (ROS). Adenosine-triphosphate (ATP) level was assessed using ATP detection, along with the detection the expression of cytochrome C in mitochondria (mito-cyt c) and in cell cytoplasm (cyto-cyt c) and cleaved caspase 3 by Western blot analysis. TUNEL assay was conducted
for the measurement of cell apoptosis in endplate chondrocytes. Reverse transcription-polymerase chain reaction (RT-qPCR) was used to verify the expression of heat shock protein 22 (HSP22) and the transfection efficiency of HSP22 interference plasmid. Results: It was found that H2O2
promoted the mitochondrial dysfunction, ROS generation and cell apoptosis in endplate chondrocytes. Moreover, HSP22 was down-regulated in H2O2-induced endplate chondrocytes, and interference of HSP22 decreased the ROS production, increased the ATP level and promoted the
cell apoptosis, resulting in the enhanced impairment of endplate chondrocytes. Additionally, mitochondrial ROS inhibitor (Mito-TEMPO) ameliorated the injury effects of HSP22 silencing in the H2O2-induced endplate chondrocytes. Conclusion: In conclusion, HSP22 inhibits
oxidative stress-induced endplate chondrocyte apoptosis by regulating mitochondrial pathway, possibly providing novel guidance direction for the treatment of LFJ degeneration.
Collapse
Affiliation(s)
- Yi Ye
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Xucan Wang
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Zhenqing Yang
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Qian Xu
- Department of Orthopedics, Ningbo Fourth Hospital, Xiang Shan County, Zhejiang 315700, P. R. China
| | - Bo Zhang
- Department of Orthopedics, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo Institute of Life and Health Industry, Ningbo, Zhejiang 315010, P. R.China
| |
Collapse
|
6
|
Wu W, Lai L, Xie M, Qiu H. Insights of heat shock protein 22 in the cardiac protection against ischemic oxidative stress. Redox Biol 2020; 34:101555. [PMID: 32388268 PMCID: PMC7215242 DOI: 10.1016/j.redox.2020.101555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
the acute and chronic myocardial ischemia results in oxidative stress that impairs myocardial contractility and eventually leads to heart failure. However, the underlying regulatory molecular mechanisms are not fully understood. The heat shock protein 22 (Hsp22), a small-molecular-weight protein preferentially expressed in the heart, was found to be dramatically increased in the cardiac oxidative stress conditions in both human and animal models after the acute and chronic ischemia. Overexpression of Hsp22 largely protects the heart against ischemic damage. Mechanistically, overexpression of Hsp22 attenuates hypoxia-induced oxidative phosphorylation in mitochondrial and the high rate of superoxide production. Short term gene delivery of Hsp22 reduces the infarct size caused by the ischemia/reperfusion, providing a clinical therapeutic potential. This review discusses the new progress of the studies on Hsp22 by focusing on its protective effect against the excessive cardiac oxidative stress, including its adaptive induction in myocardium upon the oxidative stress, its protective role in myocardial ischemia/reperfusion, its regulation in mitochondrial oxidative phosphorylation and the underlying molecular signaling pathways promoting cell survival. This information will increase our understanding of the molecular regulation of cardiac adaption under the oxidative stress and the potential therapeutic relevance.
Collapse
Affiliation(s)
- Wenqian Wu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA; Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lo Lai
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA
| | - Mingxing Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongyu Qiu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
7
|
Wan GX, Cheng L, Qin HL, Zhang YZ, Wang LY, Zhang YG. MiR-15b-5p is Involved in Doxorubicin-Induced Cardiotoxicity via Inhibiting Bmpr1a Signal in H9c2 Cardiomyocyte. Cardiovasc Toxicol 2019; 19:264-275. [PMID: 30535663 DOI: 10.1007/s12012-018-9495-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The wide use of anthracyclines represented by doxorubicin (DOX) has benefited cancer patients, yet the clinical application is limited due to its cardiotoxicity. Although numerous evidences have supported a role of microRNAs (miRNAs) in DOX-induced myocardial damage, the exact etiology and pathogenesis remain largely obscure. In this study, we focused on the role of miR-15b-5p in DOX-induced cardiotoxicity. We employed a public miRNA and gene microarray to screen differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) in rat cardiomyocytes, and 33 DEMs including miR-15b-5p and 237 DEGs including Bmpr1a and Gata4 were identified. The Gene ontology (GO) and pathway enrichment analysis of 237 DEGs indicated that the DEGs were mainly enriched in heart development and ALK pathway in cardiomyocyte which included the main receptor Bmpr1a and transcription factor Gata4. The up-regulated miR-15b-5p and down-regulated Bmpr1a and Gata4 mRNA expressions were further validated in H9c2 cardiomyocytes exposed to DOX. Moreover, the results showed overexpression of miR-15b-5p or inhibition of Bmpr1a may enhance the DOX-induced apoptosis, oxidative stress and mitochondria damage in H9c2 cardiomyocytes. The Bmpr1a was suggested as a potential target of miR-15b-5p by bioinformatics prediction. We further verified the negatively regulatory effect of miR-15b-5p on Bmpr1a signaling. Moreover, we also confirmed that overexpression of miR-15b-5p may exacerbate the DOX-induced apoptosis of H9c2 cardiomyocytes by affecting the protein expression ratio of Bcl-2/Bax and Akt activation, while this pro-apoptotic effect was able to be suppressed by Bmpr1a agonist. Collectively, the results suggest that miR-15b-5p is likely involved in doxorubicin-induced cardiotoxicity via inhibiting Bmpr1a signaling in H9c2 cardiomyocytes. Our study provides a novel insight for investigating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Guo-Xing Wan
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Lan Cheng
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Hai-Lun Qin
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Yun-Zhang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Ling-Yu Wang
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China
| | - Yong-Gang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Dongxia North Road, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
8
|
Yang S, Tian J, Zhang F, Liu A, Xie B, Chen Q. The protective effects of heat shock protein 22 in lung ischemia-reperfusion injury mice. Biochem Biophys Res Commun 2019; 512:698-704. [PMID: 30922561 DOI: 10.1016/j.bbrc.2019.03.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
Lung ischemia-reperfusion injury (LIRI) often results in respiratory insufficiency after pulmonary embolism, lung transplantation, etc. To investigate the role of HSP22 in LIRI mice, ischemia-reperfusion injury was established in the left lung of an HSP22 overexpression transgenic mouse. Twelve HSP22 transgenic (TG) mice and twelve wild-type (WT) mice were randomly divided into 2 groups: the sham-operated group (SO: TG-SO, WT-SO) and the ischemia-reperfusion group (I/R: TG-I/R, WT-I/R), respectively. We tested the PaO2, W/D ratio, and MDA level; observed morphology changes; and calculated the index of alveolar damage. HSP22 expression was examined in lung tissues of TG and WT C57BL mice by immunohistochemistry. TUNEL assay was performed to measure apoptosis. We found that HSP22 was significantly overexpressed in TG mice. There was no difference in PaO2 among the four groups. In the I/R group, the W/D ratio, MDA and index of alveolar damage were higher than those in the SO group. Moreover, compared with WT-I/R group, the W/D ratio, MDA and index of alveolar damage in the TG-I/R group were significantly decreased. Apoptosis in the I/R groups was increased compared to that in the SO groups, while apoptosis in the TG-I/R groups was decreased compared to that in the WT-I/R groups. Our results showed that HSP22 TG mice and the LIRI model were successfully established. In addition, HSP22 overexpression has protective effects on LIRI by inhibiting lipid peroxidation and apoptosis.
Collapse
Affiliation(s)
- Shasha Yang
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Tian
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Fufeng Zhang
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Aibin Liu
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Xie
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Qiong Chen
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Fang X, Bogomolovas J, Trexler C, Chen J. The BAG3-dependent and -independent roles of cardiac small heat shock proteins. JCI Insight 2019; 4:126464. [PMID: 30830872 DOI: 10.1172/jci.insight.126464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Small heat shock proteins (sHSPs) comprise an important protein family that is ubiquitously expressed, is highly conserved among species, and has emerged as a critical regulator of protein folding. While these proteins are functionally important for a variety of tissues, an emerging field of cardiovascular research reveals sHSPs are also extremely important for maintaining normal cardiac function and regulating the cardiac stress response. Notably, numerous mutations in genes encoding sHSPs have been associated with multiple cardiac diseases. sHSPs (HSPB5, HSPB6, and HSPB8) have been described as mediating chaperone functions within the heart by interacting with the cochaperone protein BCL-2-associated anthanogene 3 (BAG3); however, recent reports indicate that sHSPs (HSPB7) can perform other BAG3-independent functions. Here, we summarize the cardiac functions of sHSPs and present the notion that cardiac sHSPs function via BAG3-dependent or -independent pathways.
Collapse
|
10
|
Yu L, Liang Q, Zhang W, Liao M, Wen M, Zhan B, Bao H, Cheng X. HSP22 suppresses diabetes-induced endothelial injury by inhibiting mitochondrial reactive oxygen species formation. Redox Biol 2019; 21:101095. [PMID: 30640127 PMCID: PMC6327915 DOI: 10.1016/j.redox.2018.101095] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/20/2022] Open
Abstract
The induction of mitochondrial reactive oxygen species (mtROS) by hyperglycemia is a key event responsible for endothelial activation and injury. Heat shock protein 22 (HSP22) is a stress-inducible protein associated with cytoprotection and apoptosis inhibition. However, whether HSP22 prevents hyperglycemia-induced vascular endothelial injury remains unclear. Here, we investigated whether HSP22 protects the vascular endothelium from hyperglycemia-induced injury by reducing mtROS production. We used a high-fat diet and streptozotocin injection model to induce type 2 diabetes mellitus (T2DM, metabolic syndrome) and exposed human umbilical vein endothelial cells (HUVECs) to high glucose following overexpression or silencing of HSP22 to explore the role of HSP22. We found that HSP22 markedly inhibited endothelial cell activation and vascular lesions by inhibiting endothelial adhesion and decreasing cytokine secretion. We performed confocal microscopy and flow cytometry assays using HUVECs and showed that HSP22 attenuated mtROS and mitochondrial dysfunction in hyperglycemia-stimulated endothelial cells. Mechanistically, using the mtROS inhibitor MitoTEMPO, we demonstrated that HSP22 suppressed endothelial activation and injury by eliminating hyperglycemia-mediated increases in mtROS. Furthermore, we found that HSP22 maintained the balance of mitochondrial fusion and fission by mitigating mtROS in vitro. HSP22 attenuated the development of vascular lesions by suppressing mtROS-mediated endothelial activation in a T2DM mouse model. This study provides evidence that HSP22 may be a promising therapeutic target for vascular complications in T2DM. HSP22 reduces endothelial inflammation under diabetic conditions. HSP22 restrains hyperglycemia-induced oxidative stress in the vascular endothelium. HSP22 reduces hyperglycemia-induced mtROS and endothelial mitochondrial dysfunction. HSP22 maintains the balance of mitochondrial fusion and fission by mitigating mtROS.
Collapse
Affiliation(s)
- Lingling Yu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China
| | - Qian Liang
- Key Laboratory of Molecular Biology in Jiangxi Province, The Second Affiliated Hospital of Nanchang University, PR China
| | - Weifang Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, PR China
| | - Minqi Liao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China
| | - Minghua Wen
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China
| | - Biming Zhan
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China
| | - Huihui Bao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China.
| | - Xiaoshu Cheng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, PR China.
| |
Collapse
|
11
|
Sung JH, Song A, Park T, Kim E, Lee S. The Different Expression Patterns of HSP22, a Late Embryogenesis Abundant-like Protein, in Hypertrophic H9C2 Cells Induced by NaCl and Angiotensin II. Electrolyte Blood Press 2018; 16:1-10. [PMID: 30046328 PMCID: PMC6051945 DOI: 10.5049/ebp.2018.16.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/25/2018] [Indexed: 11/05/2022] Open
Abstract
Background High-NaCl diet is a contributing factor for cardiac hypertrophy. The role of HSP22 as a protective protein during cardiac hypertrophy due to hypernatremia is unclear. Accordingly, this study aimed to establish a cellular hypernatremic H9C2 model and to compare the expression of HSP22 in Ca2+ homeostasis between a high-NaCl and angiotensin II-induced hypertrophic cellular H9C2 model. Methods Real-time PCR was performed to compare the mRNA expression. Flow cytometry and confocal microscopy were used to analyze the cells. Results The addition of 30 mM NaCl for 48 h was the most effective condition for the induction of hypertrophic H9C2 cells (termed the in vitro hypernatremic model). Cardiac cellular hypertrophy was induced with 30 mM NaCl and 1 µM angiotensin II for 48 h, without causing abnormal morphological changes or cytotoxicity of the culture conditions. HSP22 contains a similar domain to that found in the consensus sequences of the late embryogenesis abundant protein group 3 from Artemia. The expression of HSP22 gradually decreased in the in vitro hypernatremic model. In contrast to the in vitro hypernatremic model, HSP22 increased after exposure to angiotensin II for 48 h. Intracellular Ca2+ decreased in the angiotensin II model and further decreased in the in vitro hypernatremic model. Impaired intracellular Ca2+ homeostasis was more evident in the in vitro hypernatremic model. Conclusion The results showed that NaCl significantly decreased HSP22. Decreased HSP22, due to the hypernatremic condition, affected the Ca2+ homeostasis in the H9C2 cells. Therefore, hypernatremia induces cellular hypertrophy via impaired Ca2+ homeostasis. The additional mechanisms of HSP22 need to be explored further.
Collapse
Affiliation(s)
- Jae Hwi Sung
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Ahran Song
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Taegun Park
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Eunyoung Kim
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Seunggwan Lee
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| |
Collapse
|
12
|
Penna C, Sorge M, Femminò S, Pagliaro P, Brancaccio M. Redox Aspects of Chaperones in Cardiac Function. Front Physiol 2018; 9:216. [PMID: 29615920 PMCID: PMC5864891 DOI: 10.3389/fphys.2018.00216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/26/2018] [Indexed: 12/14/2022] Open
Abstract
Molecular chaperones are stress proteins that allow the correct folding or unfolding as well as the assembly or disassembly of macromolecular cellular components. Changes in expression and post-translational modifications of chaperones have been linked to a number of age- and stress-related diseases including cancer, neurodegeneration, and cardiovascular diseases. Redox sensible post-translational modifications, such as S-nitrosylation, glutathionylation and phosphorylation of chaperone proteins have been reported. Redox-dependent regulation of chaperones is likely to be a phenomenon involved in metabolic processes and may represent an adaptive response to several stress conditions, especially within mitochondria, where it impacts cellular bioenergetics. These post-translational modifications might underlie the mechanisms leading to cardioprotection by conditioning maneuvers as well as to ischemia/reperfusion injury. In this review, we discuss this topic and focus on two important aspects of redox-regulated chaperones, namely redox regulation of mitochondrial chaperone function and cardiac protection against ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Matteo Sorge
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
13
|
Exploring the multifaceted roles of heat shock protein B8 (HSPB8) in diseases. Eur J Cell Biol 2018; 97:216-229. [PMID: 29555102 DOI: 10.1016/j.ejcb.2018.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
HSPB8 is a member of ubiquitous small heat shock protein (sHSP) family, whose expression is induced in response to a wide variety of unfavorable physiological and environmental conditions. Investigation of HSPB8 structure indicated that HSPB8 belongs to the group of so-called intrinsically disordered proteins and possesses a highly flexible structure. Unlike most other sHSPs, HSPB8 tends to form small-molecular-mass oligomers and exhibits substrate-dependent chaperone activity. In cooperation with BAG3, the chaperone activity of HSPB8 was reported to be involved in the delivery of misfolded proteins to the autophagy machinery. Through this way, HSPB8 interferes with pathological processes leading to neurodegenerative diseases. Accordingly, published studies have identified genetic links between mutations of HSPB8 and some kind of neuromuscular diseases, further supporting its important role in neurodegenerative disorders. In addition to their anti-aggregation properties, HSPB8 is indicated to interact with a wide range of client proteins, modulating their maturations and activities, and therefore, regulates a large repertoire of cellular functions, including apoptosis, proliferation, inflammation and etc. As a result, HSPB8 has key roles in cancer biology, autoimmune diseases, cardiac diseases and cerebral vascular diseases.
Collapse
|
14
|
Flanagan M, Li C, Dietrich MA, Richard M, Yao S. Downregulation of heat shock protein B8 decreases osteogenic differentiation potential of dental pulp stem cells during in vitro proliferation. Cell Prolif 2017; 51:e12420. [PMID: 29266518 DOI: 10.1111/cpr.12420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Tissue-derived stem cells, such as dental pulp stem cells (DPSCs), reduce differentiation capability during in vitro culture. We found that cultured DPSCs reduce expression of heat shock protein B8 (HspB8) and GIPC PDZ domain containing family member 2 (Gipc2). Our objectives were to evaluate the changes in DPSC composition during in vitro proliferation and to determine whether HspB8 and Gipc2 have function in differentiation potential of DPSCs. MATERIALS AND METHODS Different passages of rat DPSCs were evaluated for changes in CD90+ and/or CD271+ stem cells and changes in osteogenic potential. Real-time RT-PCR and immunostaining were conducted to determine expression of HspB8 and Gipc2. Expression of the genes in DPSCs was knocked down by siRNA, followed by osteogenic induction to evaluate the function of the genes. RESULTS About 90% of cells in the DPSC cultures were CD90+ and/or CD271+ cells without dramatic change during in vitro proliferation. The DPSCs at passages 3 to 5 (P3 to P5) possess strong osteogenic potential, but such potential was greatly reduced at later passages. Expression of HspB8 and Gipc2 was significantly reduced at P11 versus P3. Knock-down of HspB8 expression abolished osteogenic potential of the DPSCs, but knock-down of Gipc2 had no effect. CONCLUSIONS CD90+ and CD271+ cells are the major components of DPSCs in in vitro culture. High-level expression of HspB8 was critical for maintaining differentiation potential of DPSCs.
Collapse
Affiliation(s)
- M Flanagan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - C Li
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - M A Dietrich
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - M Richard
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - S Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
15
|
Jo HS, Kim DW, Shin MJ, Cho SB, Park JH, Lee CH, Yeo EJ, Choi YJ, Yeo HJ, Sohn EJ, Son O, Cho SW, Kim DS, Yu YH, Lee KW, Park J, Eum WS, Choi SY. Tat-HSP22 inhibits oxidative stress-induced hippocampal neuronal cell death by regulation of the mitochondrial pathway. Mol Brain 2017; 10:1. [PMID: 28052764 PMCID: PMC5210279 DOI: 10.1186/s13041-016-0281-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/12/2016] [Indexed: 11/24/2022] Open
Abstract
Oxidative stress plays an important role in the progression of various neuronal diseases including ischemia. Heat shock protein 22 (HSP22) is known to protect cells against oxidative stress. However, the protective effects and mechanisms of HSP22 in hippocampal neuronal cells under oxidative stress remain unknown. In this study, we determined whether HSP22 protects against hydrogen peroxide (H2O2)-induced oxidative stress in HT-22 using Tat-HSP22 fusion protein. We found that Tat-HSP22 transduced into HT-22 cells and that H2O2-induced cell death, oxidative stress, and DNA damage were significantly reduced by Tat-HSP22. In addition, Tat-HSP22 markedly inhibited H2O2-induced mitochondrial membrane potential, cytochrome c release, cleaved caspase-3, and Bax expression levels, while Bcl-2 expression levels were increased in HT-22 cells. Further, we showed that Tat-HSP22 transduced into animal brain and inhibited cleaved-caspase-3 expression levels as well as significantly inhibited hippocampal neuronal cell death in the CA1 region of animals in the ischemic animal model. In the present study, we demonstrated that transduced Tat-HSP22 attenuates oxidative stress-induced hippocampal neuronal cell death through the mitochondrial signaling pathway and plays a crucial role in inhibiting neuronal cell death, suggesting that Tat-HSP22 protein may be used to prevent oxidative stress-related brain diseases including ischemia.
Collapse
Affiliation(s)
- Hyo Sang Jo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Chi Hern Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Ora Son
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-Si, 31538, Korea
| | - Yeon Hee Yu
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-Si, 31538, Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea.
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Korea.
| |
Collapse
|
16
|
Duan Y, Zhu W, Liu M, Ashraf M, Xu M. The expression of Smad signaling pathway in myocardium and potential therapeutic effects. Histol Histopathol 2016; 32:651-659. [PMID: 27844469 DOI: 10.14670/hh-11-845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Myocardial infarction (MI) is a life-threatening disease. The expression of Smad proteins in the ischemic myocardium changes significantly following myocardial infarction, suggesting a close relationship between Smad proteins and heart remodeling. Moreover, it is known that the expression of Smads is regulated by transforming growth factor-β (TGF-β) and bone morphogenetic proteins (BMP). Based on these findings, regulating the expression of Smad proteins by targeting TGF-β and BMP in the ischemic myocardium may be considered to be a possible therapeutic strategy for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Yuping Duan
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, P.R. China.
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Muhammad Ashraf
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
17
|
Yamamoto N, Tokuda H, Kuroyanagi G, Kainuma S, Matsushima-Nishiwaki R, Fujita K, Kozawa O, Otsuka T. Heat shock protein 22 (HSPB8) limits TGF-β-stimulated migration of osteoblasts. Mol Cell Endocrinol 2016; 436:1-9. [PMID: 27396899 DOI: 10.1016/j.mce.2016.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/22/2016] [Accepted: 07/06/2016] [Indexed: 11/22/2022]
Abstract
Heat shock proteins (HSPs) are induced in response to various physiological and environmental conditions such as chemical and heat stress, and recognized to function as molecular chaperones. HSP22 (HSPB8), a low-molecular weight HSP, is ubiquitously expressed in many cell types. However, the precise role of HSP22 in bone metabolism remains to be clarified. In the present study, we investigated whether HSP22 is implicated in the transforming growth factor-β (TGF-β)-stimulated migration of osteoblast-like MC3T3-E1 cells. Although protein levels of HSP22 were clearly detected in unstimulated MC3T3-E1 cells, TGF-β failed to induce the protein levels. The TGF-β-stimulated migration was significantly up-regulated by knockdown of HSP22 expression. The cell migration stimulated by platelet-derived growth factor-BB was also enhanced by HSP22 knockdown. SB203580, an inhibitor of p38 mitogen-activated protein kinase, PD98059, an inhibitor of MEK1/2, or SP600125, an inhibitor of stress-activated protein kinase/c-Jun N-terminal kinase had no effects on the TGF-β-induced migration. SIS3, a specific inhibitor of TGF-β-dependent Smad3 phosphorylation, significantly reduced the migration with or without TGF-β stimulation. Smad2, Smad3, Smad4 or Smad7 was not coimmunoprecipitated with HSP22. On the other hand, the TGF-β-induced Smad2 phosphorylation was enhanced by HSP22 down-regulation. The protein levels of TGF-β type II receptor (TGF-β RII) but not TGF-β type I receptor (TGF-β RI) was significantly up-regulated in HSP22 knockdown cells compared with those in the control cells. However, the levels of TGF-β RII mRNA in HSP22 knockdown cells were little different from those of the control cells. Neither TGF-β RI nor TGF-β RII was coimmunoprecipitated with HSP22. SIS3 reduced the amplification by HSP22 knockdown of the TGF-β-stimulated cell migration almost to the basal level. Our results strongly suggest that HSP22 functions as a negative regulator in the TGF-β-stimulated migration of osteoblasts via suppression of the Smad-dependent pathway, resulting from modulating the protein levels of TGF-β RII.
Collapse
Affiliation(s)
- Naohiro Yamamoto
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| | - Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Shingo Kainuma
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | - Kazuhiko Fujita
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| |
Collapse
|
18
|
Hu Z, Yang B, Mo X, Zhou F. HspB8 mediates neuroprotection against OGD/R in N2A cells through the phosphoinositide 3-kinase/Akt pathway. Brain Res 2016; 1644:15-21. [DOI: 10.1016/j.brainres.2016.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/21/2016] [Accepted: 05/07/2016] [Indexed: 01/25/2023]
|
19
|
Elkenani M, Nyamsuren G, Raju P, Liakath-Ali K, Hamdaoui A, Kata A, Dressel R, Klonisch T, Watt FM, Engel W, Thliveris JA, Krishna Pantakani DV, Adham IM. Pelota Regulates Epidermal Differentiation by Modulating BMP and PI3K/AKT Signaling Pathways. J Invest Dermatol 2016; 136:1664-1671. [PMID: 27164299 DOI: 10.1016/j.jid.2016.04.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 04/04/2016] [Accepted: 04/11/2016] [Indexed: 12/25/2022]
Abstract
The depletion of evolutionarily conserved pelota protein causes impaired differentiation of embryonic and spermatogonial stem cells. In this study, we show that temporal deletion of pelota protein before epidermal barrier acquisition leads to neonatal lethality due to perturbations in permeability barrier formation. Further analysis indicated that this phenotype is a result of failed processing of profilaggrin into filaggrin monomers, which promotes the formation of a protective epidermal layer. Molecular analyses showed that pelota protein negatively regulates the activities of bone morphogenetic protein and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways in the epidermis. To address whether elevated activities of bone morphogenetic protein and PI3K/AKT signaling pathways were the cause for the perturbed epidermal barrier in Pelo-deficient mice, we made use of organotypic cultures of skin explants from control and mutant embryos at embryonic day 15.5. Inhibition of PI3K/AKT signaling did not significantly affect the bone morphogenetic protein activity. However, inhibition of bone morphogenetic protein signaling caused a significant attenuation of PI3K/AKT activity in mutant skin and, more interestingly, the restoration of profilaggrin processing and normal epidermal barrier function. Therefore, increased activity of the PI3K/AKT signaling pathway in Pelo-deficient skin might conflict with the dephosphorylation of profilaggrin and thereby affect its proper processing into filaggrin monomers and ultimately the epidermal differentiation.
Collapse
Affiliation(s)
- Manar Elkenani
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany; Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Gunsmaa Nyamsuren
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany
| | - Priyadharsini Raju
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany
| | - Kifayathullah Liakath-Ali
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London, UK; Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Old Addenbrooke's Site, Cambridge, UK
| | - Aicha Hamdaoui
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany
| | - Aleksandra Kata
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, D-37073 Göttingen, Germany
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London, UK
| | - Wolfgang Engel
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D V Krishna Pantakani
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany; Institute of Clinical Chemistry/UMG-Laboratories, University Medical Center Göttingen, D-37075, Göttingen, Germany
| | - Ibrahim M Adham
- Institute of Human Genetics, University of Göttingen, D-37073 Göttingen, Germany.
| |
Collapse
|
20
|
Sehgal SA, Mannan S, Ali S. Pharmacoinformatic and molecular docking studies reveal potential novel antidepressants against neurodegenerative disorders by targeting HSPB8. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1605-18. [PMID: 27226709 PMCID: PMC4866741 DOI: 10.2147/dddt.s101929] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Charcot-Marie-Tooth (CMT) disease is an inherited peripheral neuromuscular disorder characterized by length-dependent and progressive degeneration of peripheral nerves, leading to muscular weakness. Research has shown that mutated HSPB8 may be responsible for depression, neurodegenerative disorders, and improper functioning of peripheral nerves, resulting in neuromuscular disorders like CMT. In the current work, a hybrid approach of virtual screening and molecular docking studies was followed by homology modeling and pharmacophore identification. Detailed screening analyses were carried out by 2-D similarity search against prescribed antidepressant drugs with physicochemical properties. LigandScout was employed to ascertain novel molecules and pharmacophore properties. In this study, we report three novel compounds that showed maximum binding affinity with HSPB8. Docking analysis elucidated that Met37, Ser57, Ser58, Trp60, Thr63, Thr114, Lys115, Asp116, Gly117, Val152, Val154, Leu186, Asp189, Ser190, Gln191, and Glu192 are critical residues for ligand-receptor interactions. Our analyses suggested paroxetine as a potent compound for targeting HSPB8. Selected compounds have more effective energy scores than the selected drug analogs. Additionally, site-directed mutagenesis could be significant for further analysis of the binding pocket. The novel findings based on an in silico approach may be momentous for potent drug design against depression and CMT.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Department of Bioscience, COMSATS Institute of Information Technology, Sahiwal, Pakistan; State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China; University of Chinese Academy of Sciences, Beijing, People's Republic of China; Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Shazia Mannan
- Department of Bioscience, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Sannia Ali
- Department of Bioscience, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| |
Collapse
|
21
|
Rashed E, Lizano P, Dai H, Thomas A, Suzuki CK, Depre C, Qiu H. Heat shock protein 22 (Hsp22) regulates oxidative phosphorylation upon its mitochondrial translocation with the inducible nitric oxide synthase in mammalian heart. PLoS One 2015; 10:e0119537. [PMID: 25746286 PMCID: PMC4352051 DOI: 10.1371/journal.pone.0119537] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/18/2015] [Indexed: 11/21/2022] Open
Abstract
Objectives Stress-inducible heat shock protein 22 (Hsp22) confers protection against ischemia through induction of the inducible isoform of nitric oxide synthase (iNOS). Hsp22 overexpression in vivo stimulates cardiac mitochondrial respiration, whereas Hsp22 deletion in vivo significantly reduces respiration. We hypothesized that Hsp22-mediated regulation of mitochondrial function is dependent upon its mitochondrial translocation together with iNOS. Methods and Results Adenoviruses harboring either the full coding sequence of Hsp22 (Ad-WT-Hsp22) or a mutant lacking a N-terminal 20 amino acid putative mitochondrial localization sequence (Ad-N20-Hsp22) were generated, and infected in rat neonatal cardiomyocytes. Compared to β-Gal control, WT-Hsp22 accumulated in mitochondria by 2.5 fold (P<0.05) and increased oxygen consumption rates by 2-fold (P<0.01). This latter effect was abolished upon addition of the selective iNOS inhibitor, 1400W. Ad-WT-Hsp22 significantly increased global iNOS expression by about 2.5-fold (P<0.01), and also increased iNOS mitochondrial localization by 4.5 fold vs β-gal (P<0.05). Upon comparable overexpression, the N20-Hsp22 mutant did not show significant mitochondrial translocation or stimulation of mitochondrial respiration. Moreover, although N20-Hsp22 did increase global iNOS expression by 4.6-fold, it did not promote iNOS mitochondrial translocation. Conclusion Translocation of both Hsp22 and iNOS to the mitochondria is necessary for Hsp22-mediated stimulation of oxidative phosphorylation.
Collapse
Affiliation(s)
- Eman Rashed
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Paulo Lizano
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Huacheng Dai
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Andrew Thomas
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Carolyn K. Suzuki
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Christophe Depre
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
- Department of Basic Science, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
- * E-mail:
| |
Collapse
|
22
|
Tao X, Lu W, Deng J, Hu Z, Lei Q, Zhang J, Song T, Liu J, Zheng L, He J. HspB8 expression in brain tissue after cerebral ischemic reperfusion and atorvastatin intervention in Sprague-Dawley rats. Neurol Res 2015; 37:229-237. [PMID: 25082545 DOI: 10.1179/1743132814y.0000000427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate the expression of HspB8 in the brain cortex of Sprague-Dawley rats after cerebral ischemic reperfusion (I/R) and atorvastatin intervention. It also aimed to determine the possible mechanism of atorvastatin intervention. A model of cerebral I/R in Sprague-Dawley rats was used. METHODS Fifty-two male Sprague-Dawley rats were randomly divided into four groups. The normal and sham-operated groups received no treatment. The I/R group was intragastrically administered with physiological saline, and the intervention group received atorvastatin (10 mg/kg) prepared with physiological saline according to reperfusion time (time of palinesthesia, 24 and 48 hours). All rats were killed after 72 hours of reperfusion. RESULTS 2,3,5-Triphenyltetrazolium chloride staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling showed that atorvastatin significantly reduced the volume of cerebral infarction (%) in the brain tissue and attenuated neuronal apoptosis in the cortex of rats, respectively. HspB8 was expressed in the pallium of Sprague-Dawley rats in the normal and sham-operated groups. The expression level of HspB8 was significantly upregulated after cerebral I/R. However, the level of HspB8 expressed was lower in the intervention group than in the I/R group. CONCLUSION The expression of HspB8 was upregulated after cerebral I/R injury. Atorvastatin affected the precursors of the heat shock stress response, relieved stress injury due to cerebral ischemia, downregulated HspB8 expression, and inhibited nerve cell apoptosis in the brain cortex. However, HspB8 might not contribute to atorvastatin-mediated cytoprotective effects.
Collapse
|
23
|
Tarone G, Brancaccio M. Keep your heart in shape: molecular chaperone networks for treating heart disease. Cardiovasc Res 2014; 102:346-61. [PMID: 24585203 DOI: 10.1093/cvr/cvu049] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Despite major advances in the treatment of cardiac diseases, there is still a great need for drugs capable of counteracting the deterioration of cardiac muscle function in congestive heart failure. The role of misfolded protein accumulation as a causal event in the physiopathology of common cardiac diseases is an important emerging concept. Indeed, diverse stress conditions, including mechanical stretching and oxidative stress, can induce misfolded protein accumulation, causing cardiomyocyte death. Cells react to these stress conditions by activating molecular chaperones, a class of proteins that represents an endogenous salvage machinery, essential for rescuing physiological cell functions and sustaining cell survival. Chaperones, also known as heat shock proteins (Hsps), prevent accumulation of damaged proteins by promoting either their refolding or degradation via the proteasome or the autophagosome systems. In addition, molecular chaperones play a key role in intracellular signalling by controlling conformational changes required for activation/deactivation of signalling proteins, and their assembly in specific signalosome complexes. The key role of molecular chaperones in heart function is highlighted by the fact that a number of genetic mutations in chaperone proteins result in different forms of cardiomyopathies. Moreover, a considerable amount of experimental evidence indicates that increasing expression of chaperone proteins leads to an important cardio-protective role in ischaemia/reperfusion injury, heart failure, and arrhythmia, implicating these molecules as potential innovative therapeutic agents.
Collapse
Affiliation(s)
- Guido Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Italy, Via Nizza 52, Torino 10126, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Italy, Via Nizza 52, Torino 10126, Italy
| |
Collapse
|
24
|
Dubińska-Magiera M, Jabłońska J, Saczko J, Kulbacka J, Jagla T, Daczewska M. Contribution of small heat shock proteins to muscle development and function. FEBS Lett 2014; 588:517-30. [PMID: 24440355 DOI: 10.1016/j.febslet.2014.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/17/2013] [Accepted: 01/02/2014] [Indexed: 12/17/2022]
Abstract
Investigations undertaken over the past years have led scientists to introduce the concept of protein quality control (PQC) systems, which are responsible for polypeptide processing. The PQC system monitors proteostasis and involves activity of different chaperones such as small heat shock proteins (sHSPs). These proteins act during normal conditions as housekeeping proteins regulating cellular processes, and during stress conditions. They also mediate the removal of toxic misfolded polypeptides and thereby prevent development of pathogenic states. It is postulated that sHSPs are involved in muscle development. They could act via modulation of myogenesis or by maintenance of the structural integrity of signaling complexes. Moreover, mutations in genes coding for sHSPs lead to pathological states affecting muscular tissue functioning. This review focuses on the question how sHSPs, still relatively poorly understood proteins, contribute to the development and function of three types of muscle tissue: skeletal, cardiac and smooth.
Collapse
Affiliation(s)
- Magda Dubińska-Magiera
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland
| | - Jadwiga Jabłońska
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland
| | - Jolanta Saczko
- Department of Medical Biochemistry, Medical University, Chalubinskiego 10, 50-368 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Medical Biochemistry, Medical University, Chalubinskiego 10, 50-368 Wroclaw, Poland
| | - Teresa Jagla
- Institut National de la Santé et de la Recherche Médicale U384, Faculté de Medecine, Clermont-Ferrand, France
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| |
Collapse
|
25
|
Lizano P, Rashed E, Kang H, Dai H, Sui X, Yan L, Qiu H, Depre C. The valosin-containing protein promotes cardiac survival through the inducible isoform of nitric oxide synthase. Cardiovasc Res 2013; 99:685-93. [PMID: 23737493 DOI: 10.1093/cvr/cvt136] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS Expression of the heat shock protein 22 (Hsp22) in the heart stimulates cardiac cell survival through activation of the Akt pathway and expression of the inducible nitric oxide (NO) synthase (iNOS), the mediator of ischaemic preconditioning and the most powerful prophylaxis against cardiac cell death. The goal of the present study was to elucidate the downstream effector by which Hsp22 and Akt increase iNOS expression. We tested both in vivo and in vitro the hypothesis that such an effector is the valosin-containing protein (VCP), an Akt substrate, which activates the transcription factor NF-κB, using a transgenic mouse with cardiac-specific over-expression of Hsp22, as well as isolated rat cardiac myocytes. METHODS AND RESULTS Using two-dimensional gel electrophoresis and mass spectrometry combined with immunoprecipitation, we found that Hsp22 and Akt co-localize and interact together with VCP. Adeno-mediated over-expression of VCP in isolated cardiac myocytes activated NF-κB and dose-dependently increased the expression of iNOS, which was abolished upon NF-κB inhibition. Over-expression of a dominant-negative (DN) mutant of VCP did not increase iNOS expression. VCP, but not its DN mutant, protected against chelerythrine-induced apoptosis, which was suppressed by inhibition of either NF-κB or iNOS. VCP-mediated activation of the NF-κB/iNOS pathway was also prevented upon inhibition of Akt. CONCLUSION We conclude that the Akt substrate, VCP, mediates the increased expression of iNOS downstream from Hsp22 through an NF-κB-dependent mechanism.
Collapse
Affiliation(s)
- Paulo Lizano
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, 07103, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
The α crystallin domain of small heat shock protein b8 (Hspb8) acts as survival and differentiation factor in adult hippocampal neurogenesis. J Neurosci 2013; 33:5785-96. [PMID: 23536091 DOI: 10.1523/jneurosci.6452-11.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adult hippocampal neurogenesis is to a large degree controlled at the level of cell survival, and a number of potential mediators of this effect have been postulated. Here, we investigated the small heat shock protein Hspb8, which, because of its pleiotropic prosurvival effects in other systems, was considered a particularly promising candidate factor. Hspb8 is, for example, found in plaques of Alzheimer disease but exerts neuroprotective effects. We found that expression of Hspb8 increased during differentiation in vitro and was particularly associated with later stages (48-96 h) of differentiation. Gain-of-function and loss-of-function experiments supported the hypothesis that Hspb8 regulates cell survival of new neurons in vitro. In the dentate gyrus of adult mice in vivo, lentiviral overexpression of Hspb8 doubled the surviving cells and concomitantly promoted differentiation and net neurogenesis without affecting precursor cell proliferation. We also discovered that the truncated form of the crystallin domain of Hspb8 was sufficient to affect cell survival and neuronal differentiation in vitro and in vivo. Precursor cell experiments in vitro revealed that Hspb8 increases the phosphorylation of Akt and suggested that the prosurvival effect can be produced by a cell-autonomous mechanism. Analysis of hippocampal Hspb8 expression in mice of 69 strains of the recombinant inbred set BXD revealed that Hspb8 is a cis-acting gene whose expression was associated with clusters of transcript enriched in genes linked to growth factor signaling and apoptosis. Our results strongly suggest that Hspb8 and its α-crystallin domain might act as pleiotropic prosurvival factor in the adult hippocampus.
Collapse
|
27
|
H11/HspB8 and Its Herpes Simplex Virus Type 2 Homologue ICP10PK Share Functions That Regulate Cell Life/Death Decisions and Human Disease. Autoimmune Dis 2012; 2012:395329. [PMID: 23056924 PMCID: PMC3463903 DOI: 10.1155/2012/395329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 12/24/2022] Open
Abstract
Small heat shock proteins (sHsp) also known as HspB are a large family of widely expressed proteins that contain a 90 residues domain known as α-crystallin. Here, we focus on the family member H11/HspB8 and its herpes simplex virus type 2 (HSV-2) homologue ICP10PK, and discuss the possible impact of this relationship on human disease. H11/HspB8 and ICP10PK are atypical protein kinases. They share multi-functional activity that encompasses signaling, unfolded protein response (UPR) and the regulation of life cycle potential. In melanocytes H11/HspB8 causes growth arrest. It is silenced in a high proportion of melanoma prostate cancer, Ewing's sarcoma and hematologic malignancies through aberrant DNA methylation. Its restored expression induces cell death and inhibits tumor growth in xenograft models, identifying H11/HspB8 as a tumor suppressor. This function involves the activation of multiple and distinct death pathways, all of which initiate with H11/HspB8-mediated phosphorylation of transforming growth factor β-activated kinase 1 (TAK1). Both ICP10PK and H11/HspB8 were implicated in inflammatory processes that involve dendritic cells activation through Toll-like receptor-dependent pathways and may contribute to the onset of autoimmunity. The potential evolutionary relationship of H11/HspB8 to ICP10PK, its impact on human disorders and the development of therapeutic strategies are discussed.
Collapse
|
28
|
Restored expression of the atypical heat shock protein H11/HspB8 inhibits the growth of genetically diverse melanoma tumors through activation of novel TAK1-dependent death pathways. Cell Death Dis 2012; 3:e371. [PMID: 22898869 PMCID: PMC3434666 DOI: 10.1038/cddis.2012.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Melanoma is an aggressive and drug-resistant cancer in need of improved therapeutic strategies. Restored expression of transcriptionally silenced genes is a potential approach, but it is limited by the genetic diversity of the melanoma tumors. The atypical heat shock protein H11/HspB8 has kinase activity and is silenced in melanoma through aberrant DNA methylation. We report that its restored expression induces the death of genetically diverse melanoma lines and inhibits tumor growth through the activation of novel TAK1-dependent death pathways. These include (i) caspase-1 activation independent of the inflammasome through upregulation of apoptosis-associated speck-like protein containing a CARD (ASC), (ii) Beclin-1 upregulation through phosphorylation of mammalian target of rapamycin (mTOR) at S2481 and (iii) apoptosis caused by caspase-1-mediated Beclin-1 cleavage. These data extend current understanding of cell death-associated functions, underscore the strong therapeutic promise of H11/HspB8 and identify TAK1 as a potential intervention target in melanoma.
Collapse
|
29
|
Laure L, Long R, Lizano P, Zini R, Berdeaux A, Depre C, Morin D. Cardiac H11 kinase/Hsp22 stimulates oxidative phosphorylation and modulates mitochondrial reactive oxygen species production: Involvement of a nitric oxide-dependent mechanism. Free Radic Biol Med 2012; 52:2168-76. [PMID: 22542467 DOI: 10.1016/j.freeradbiomed.2012.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 02/23/2012] [Accepted: 03/09/2012] [Indexed: 11/23/2022]
Abstract
H11 kinase/Hsp22 (Hsp22), a small heat shock protein upregulated by ischemia/reperfusion, provides cardioprotection equal to ischemic preconditioning (IPC) through a nitric oxide (NO)-dependent mechanism. A main target of NO-mediated preconditioning is the mitochondria, where NO reduces O₂ consumption and reactive oxygen species (ROS) production during ischemia. Therefore, we tested the hypothesis that Hsp22 overexpression modulates mitochondrial function through an NO-sensitive mechanism. In cardiac mitochondria isolated from transgenic (TG) mice with cardiac-specific overexpression of Hsp22, mitochondrial basal, ADP-dependent, and uncoupled O₂ consumption was increased in the presence of either glucidic or lipidic substrates. This was associated with a decrease in the maximal capabilities of complexes I and III to generate superoxide anion in combination with an inhibition of superoxide anion production by the reverse electron flow. NO synthase expression and NO production were increased in mitochondria from TG mice. Hsp22-induced increase in O₂ consumption was abolished either by pretreatment of TG mice with the NO synthase inhibitor L-N(G)-nitroarginine methyl ester (L-NAME) or in isolated mitochondria by the NO scavenger phenyltetramethylimidazoline-1-oxyl-3-oxide. L-NAME pretreatment also restored the reverse electron flow. After anoxia, mitochondria from TG mice showed a reduction in both oxidative phosphorylation and H₂O₂ production, an effect partially reversed by L-NAME. Taken together, these results demonstrate that Hsp22 overexpression increases the capacity of mitochondria to produce NO, which stimulates oxidative phosphorylation in normoxia and decreases oxidative phosphorylation and reactive oxygen species production after anoxia. Such characteristics replicate those conferred by IPC, thereby placing Hsp22 as a potential tool for prophylactic protection of mitochondrial function during ischemia.
Collapse
Affiliation(s)
- Lydie Laure
- INSERM U955, Equipe 03, Université Paris-Est, Faculté de Médecine, F-94010 Créteil, France
| | | | | | | | | | | | | |
Collapse
|
30
|
Acunzo J, Katsogiannou M, Rocchi P. Small heat shock proteins HSP27 (HspB1), αB-crystallin (HspB5) and HSP22 (HspB8) as regulators of cell death. Int J Biochem Cell Biol 2012; 44:1622-31. [PMID: 22521623 DOI: 10.1016/j.biocel.2012.04.002] [Citation(s) in RCA: 221] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/02/2012] [Accepted: 04/04/2012] [Indexed: 01/17/2023]
Abstract
Hsp27, αB-crystallin and HSP22 are ubiquitous small heat shock proteins (sHsp) whose expression is induced in response to a wide variety of unfavorable physiological and environmental conditions. These sHsp protect cells from otherwise lethal conditions mainly by their involvement in cell death pathways such as necrosis, apoptosis or autophagy. At a molecular level, the mechanisms accounting for sHsp functions in cell death are (1) prevention of denatured proteins aggregation, (2) regulation of caspase activity, (3) regulation of the intracellular redox state, (4) function in actin polymerization and cytoskeleton integrity and (5) proteasome-mediated degradation of selected proteins. In cancer cells, these sHsp are often overexpressed and associated with increased tumorigenicity, cancer cells metastatic potential and resistance to chemotherapy. Altogether, these properties suggest that Hsp27, αB-crystallin and Hsp22 are appropriate targets for modulating cell death pathways. In the present, we briefly review recent reports showing molecular evidence of cell death regulation by these sHsp and co-chaperones. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
Affiliation(s)
- Julie Acunzo
- Centre de Recherche en Cancérologie de Marseille, UMR1068 Inserm, Institut Paoli-Calmette, Aix-Marseille Univ, Marseille, France
| | | | | |
Collapse
|
31
|
Nasim MT, Ogo T, Chowdhury HM, Zhao L, Chen CN, Rhodes C, Trembath RC. BMPR-II deficiency elicits pro-proliferative and anti-apoptotic responses through the activation of TGFβ-TAK1-MAPK pathways in PAH. Hum Mol Genet 2012; 21:2548-58. [PMID: 22388934 DOI: 10.1093/hmg/dds073] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a cardiovascular disorder associated with enhanced proliferation and suppressed apoptosis of pulmonary arterial smooth muscle cells (PASMCs). Heterozygous mutations in the type II receptor for bone morphogenetic protein (BMPR2) underlie the majority of the inherited and familial forms of PAH. The transforming growth factor β (TGFβ) pathway is activated in both human and experimental models of PAH. However, how these factors exert pro-proliferative and anti-apoptotic responses in PAH remains unclear. Using mouse primary PASMCs derived from knock-in mice, we demonstrated that BMPR-II dysfunction promotes the activation of small mothers against decapentaplegia-independent mitogen-activated protein kinase (MAPK) pathways via TGFβ-associated kinase 1 (TAK1), resulting in a pro-proliferative and anti-apoptotic response. Inhibition of the TAK1-MAPK axis rescues abnormal proliferation and apoptosis in these cells. In both hypoxia and monocrotaline-induced PAH rat models, which display reduced levels of bmpr2 transcripts, this study further indicates that the TGFβ-MAPK axis is activated in lungs following elevation of both expression and phosphorylation of the TAK1 protein. In ex vivo cell-based assays, TAK1 inhibits BMP-responsive reporter activity and interacts with BMPR-II receptor. In the presence of pathogenic BMPR2 mutations observed in PAH patients, this interaction is greatly reduced. Taken together, these data suggest dysfunctional BMPR-II responsiveness intensifies TGFβ-TAK1-MAPK signalling and thus alters the ratio of apoptosis to proliferation. This axis may be a potential therapeutic target in PAH.
Collapse
Affiliation(s)
- Md Talat Nasim
- Department of Medical and Molecular Genetics, King’s College London, London, UK.
| | | | | | | | | | | | | |
Collapse
|
32
|
Mymrikov EV, Seit-Nebi AS, Gusev NB. Large potentials of small heat shock proteins. Physiol Rev 2011; 91:1123-59. [PMID: 22013208 DOI: 10.1152/physrev.00023.2010] [Citation(s) in RCA: 316] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Modern classification of the family of human small heat shock proteins (the so-called HSPB) is presented, and the structure and properties of three members of this family are analyzed in detail. Ubiquitously expressed HSPB1 (HSP27) is involved in the control of protein folding and, when mutated, plays a significant role in the development of certain neurodegenerative disorders. HSPB1 directly or indirectly participates in the regulation of apoptosis, protects the cell against oxidative stress, and is involved in the regulation of the cytoskeleton. HSPB6 (HSP20) also possesses chaperone-like activity, is involved in regulation of smooth muscle contraction, has pronounced cardioprotective activity, and seems to participate in insulin-dependent regulation of muscle metabolism. HSPB8 (HSP22) prevents accumulation of aggregated proteins in the cell and participates in the regulation of proteolysis of unfolded proteins. HSPB8 also seems to be directly or indirectly involved in regulation of apoptosis and carcinogenesis, contributes to cardiac cell hypertrophy and survival and, when mutated, might be involved in development of neurodegenerative diseases. All small heat shock proteins play important "housekeeping" roles and regulate many vital processes; therefore, they are considered as attractive therapeutic targets.
Collapse
Affiliation(s)
- Evgeny V Mymrikov
- Department of Biochemistry, School of Biology, Moscow State University, Moscow, Russian Federation
| | | | | |
Collapse
|
33
|
Modem S, Chinnakannu K, Bai U, Reddy GPV, Reddy TR. Hsp22 (HspB8/H11) knockdown induces Sam68 expression and stimulates proliferation of glioblastoma cells. J Cell Physiol 2011; 226:2747-51. [PMID: 21678403 DOI: 10.1002/jcp.22868] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sam68 (Src-associated protein in mitosis 68 kDa) is a multifunctional protein, known to govern cellular signal transduction, transcription, RNA metabolism, proliferation, apoptosis, and HIV-1 replication. Although intrinsic mechanisms that modulate Sam68 function are beginning to emerge, the regulatory events contributing to its expression remain elusive. We previously reported that heat shock protein-22 (Hsp22) antagonizes Sam68 function in rev-response element (RRE)-mediated gene expression. We now demonstrate that Sam68 levels correlate inversely with Hsp22 in a variety of cells, including U87, Jurkat, 293T, and U-937. In U87 glioblastoma cells, which contained high levels of Hsp22 than other cell lines tested, Hsp22 knockdown dramatically increased both Sam68 mRNA and protein, altered cellular morphology, and enhanced cell proliferation. This heightened proliferation was associated with a sharp decrease in G(0) /G(1) and a corresponding increase in S and G(2) /M phases in exponentially growing cultures. The increased S phase population in turn correlated with enhanced expression of cell cycle regulatory proteins such as cyclin E, cyclin A, ribonucleotide reductase (RNR), and proliferating cell nuclear antigen (PCNA), which are required for the transition of cells from G(1) to S phase. Collectively, our results demonstrate for the first time that Hsp22 regulates Sam68 expression and the ratio of Sam68 to Hsp22 may determine the proliferative potential of glioblastoma cells.
Collapse
Affiliation(s)
- Suhasini Modem
- Department of Immunology and Microbiology, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | |
Collapse
|
34
|
Drake KM, Zygmunt D, Mavrakis L, Harbor P, Wang L, Comhair SA, Erzurum SC, Aldred MA. Altered MicroRNA processing in heritable pulmonary arterial hypertension: an important role for Smad-8. Am J Respir Crit Care Med 2011; 184:1400-8. [PMID: 21920918 DOI: 10.1164/rccm.201106-1130oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Heritable pulmonary arterial hypertension (HPAH) is primarily caused by mutations of the bone morphogenetic protein (BMP) type-II receptor (BMPR2). Recent identification of mutations in the downstream mediator Smad-8 (gene, SMAD9) was surprising, because loss of Smad-8 function in canonical BMP signaling is largely compensated by Smad-1 and -5. We therefore hypothesized that noncanonical pathways may play an important role in PAH. OBJECTIVES To determine whether HPAH mutations disrupt noncanonical Smad-mediated microRNA (miR) processing. METHODS Expression of miR-21, miR-27a, and miR-100 was studied in pulmonary artery endothelial (PAEC) and pulmonary artery smooth muscle cells (PASMC) from explant lungs of patients with PAH. MEASUREMENTS AND MAIN RESULTS SMAD9 mutation completely abrogated miR induction, whereas canonical signaling was only reduced by one-third. miR-21 levels actually decreased, suggesting that residual canonical signaling uses up or degrades existing miR-21. BMPR2 mutations also led to loss of miR induction in two of three cases. HPAH cells proliferated faster than other PAH or controls. miR-21 and miR-27a each showed antiproliferative effects in PAEC and PASMC, and PAEC growth rate after BMP treatment correlated strongly with miR-21 fold-change. Overexpression of SMAD9 corrected miR processing and reversed the hyperproliferative phenotype. CONCLUSIONS HPAH-associated mutations engender a primary defect in noncanonical miR processing, whereas canonical BMP signaling is partially maintained. Smad-8 is essential for this miR pathway and its loss was not complemented by Smad-1 and -5; this may represent the first nonredundant role for Smad-8. Induction of miR-21 and miR-27a may be a critical component of BMP-induced growth suppression, loss of which likely contributes to vascular cell proliferation in HPAH.
Collapse
Affiliation(s)
- Kylie M Drake
- Genomic Medicine Institute, Cleveland Clinic, OH 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Qiu H, Lizano P, Laure L, Sui X, Rashed E, Park JY, Hong C, Gao S, Holle E, Morin D, Dhar SK, Wagner T, Berdeaux A, Tian B, Vatner SF, Depre C. H11 kinase/heat shock protein 22 deletion impairs both nuclear and mitochondrial functions of STAT3 and accelerates the transition into heart failure on cardiac overload. Circulation 2011; 124:406-15. [PMID: 21747053 DOI: 10.1161/circulationaha.110.013847] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cardiac overload, a major cause of heart failure, induces the expression of the heat shock protein H11 kinase/Hsp22 (Hsp22). METHODS AND RESULTS To determine the specific function of Hsp22 in that context, a knockout mouse model of Hsp22 deletion was generated. Although comparable to wild-type mice in basal conditions, knockout mice exposed to pressure overload developed less hypertrophy and showed ventricular dilation, impaired contractile function, increased myocyte length and accumulation of interstitial collagen, faster transition into heart failure, and increased mortality. Microarrays revealed that hearts from knockout mice failed to transactivate genes regulated by the transcription factor STAT3. Accordingly, nuclear STAT3 tyrosine phosphorylation was decreased in knockout mice. Silencing and overexpression experiments in isolated neonatal rat cardiomyocytes showed that Hsp22 activates STAT3 via production of interleukin-6 by the transcription factor nuclear factor-κB. In addition to its transcriptional function, STAT3 translocates to the mitochondria where it increases oxidative phosphorylation. Both mitochondrial STAT3 translocation and respiration were also significantly decreased in knockout mice. CONCLUSIONS This study found that Hsp22 represents a previously undescribed activator of both nuclear and mitochondrial functions of STAT3, and its deletion in the context of pressure overload in vivo accelerates the transition into heart failure and increases mortality.
Collapse
Affiliation(s)
- Hongyu Qiu
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, 185 S Orange Street, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Smith CC, Li B, Liu J, Lee KS, Aurelian L. The Levels of H11/HspB8 DNA methylation in human melanoma tissues and xenografts are a critical molecular marker for 5-Aza-2'-deoxycytidine therapy. Cancer Invest 2011; 29:383-95. [PMID: 21649464 PMCID: PMC3111925 DOI: 10.3109/07357907.2011.584588] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
H11/HspB8 is a functionally distinct small heat shock protein. It causes growth arrest in melanocytes, associated with the inhibition of Cyclin E/Cdk2 and β-catenin phosphorylation at the transcriptional activity site Ser(552) and is silenced through DNA methylation in 27/35 (77%) melanoma tissues/early cultures. 5-Aza-2'-deoxycytidine (Aza-C) induces melanoma cell death correlated with the levels of H11/HspB8 DNA methylation (p < .001). In line with low/moderate H11/HspB8 methylation, PI3-K inhibition increases Aza-C-induced cell death. Aza-C inhibits the growth of melanoma xenografts related to the levels of H11/HspB8 methylation, and a nonmethylated/non-TAK1 binding H11/HspB8 mutant confers Aza-C resistance. H11/HspB8 is a potential molecular marker for demethylation therapies.
Collapse
Affiliation(s)
- Cynthia C. Smith
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Baiquan Li
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Juan Liu
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Kie-Sok Lee
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Laure Aurelian
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, 655 West Baltimore Street, Baltimore, Maryland 21201, USA
| |
Collapse
|
37
|
Ke L, Meijering RAM, Hoogstra-Berends F, Mackovicova K, Vos MJ, Van Gelder IC, Henning RH, Kampinga HH, Brundel BJJM. HSPB1, HSPB6, HSPB7 and HSPB8 protect against RhoA GTPase-induced remodeling in tachypaced atrial myocytes. PLoS One 2011; 6:e20395. [PMID: 21731611 PMCID: PMC3123278 DOI: 10.1371/journal.pone.0020395] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 04/28/2011] [Indexed: 11/29/2022] Open
Abstract
Background We previously demonstrated the small heat shock protein, HSPB1, to prevent tachycardia remodeling in in vitro and in vivo models for Atrial Fibrillation (AF). To gain insight into its mechanism of action, we examined the protective effect of all 10 members of the HSPB family on tachycardia remodeling. Furthermore, modulating effects of HSPB on RhoA GTPase activity and F-actin stress fiber formation were examined, as this pathway was found of prime importance in tachycardia remodeling events and the initiation of AF. Methods and Results Tachypacing (4 Hz) of HL-1 atrial myocytes significantly and progressively reduced the amplitude of Ca2+ transients (CaT). In addition to HSPB1, also overexpression of HSPB6, HSPB7 and HSPB8 protected against tachypacing-induced CaT reduction. The protective effect was independent of HSPB1. Moreover, tachypacing induced RhoA GTPase activity and caused F-actin stress fiber formation. The ROCK inhibitor Y27632 significantly prevented tachypacing-induced F-actin formation and CaT reductions, showing that RhoA activation is required for remodeling. Although all protective HSPB members prevented the formation of F-actin stress fibers, their mode of action differs. Whilst HSPB1, HSPB6 and HSPB7 acted via direct prevention of F-actin formation, HSPB8-protection was mediated via inhibition of RhoA GTPase activity. Conclusion Overexpression of HSPB1, as well as HSPB6, HSPB7 and HSPB8 independently protect against tachycardia remodeling by attenuation of the RhoA GTPase pathway at different levels. The cardioprotective role for multiple HSPB members indicate a possible therapeutic benefit of compounds able to boost the expression of single or multiple members of the HSPB family.
Collapse
Affiliation(s)
- Lei Ke
- Department of Radiation and Stress Cell Biology, University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roelien A. M. Meijering
- Department of Clinical Pharmacology, GUIDE, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Femke Hoogstra-Berends
- Department of Clinical Pharmacology, GUIDE, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Nyken BV, Groningen, The Netherlands
| | - Katarina Mackovicova
- Department of Clinical Pharmacology, GUIDE, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michel J. Vos
- Department of Radiation and Stress Cell Biology, University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Isabelle C. Van Gelder
- Department of Cardiology, GUIDE, University of Groningen, University Medical Center Groningen, and the Interuniversity Cardiology Institute Netherlands, Utrecht, The Netherlands
| | - Robert H. Henning
- Department of Clinical Pharmacology, GUIDE, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harm H. Kampinga
- Department of Radiation and Stress Cell Biology, University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bianca J. J. M. Brundel
- Department of Clinical Pharmacology, GUIDE, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Fortin CF, Cloutier A, Ear T, Sylvain-Prévost S, Mayer TZ, Bouchelaghem R, McDonald PP. A class IA PI3K controls inflammatory cytokine production in human neutrophils. Eur J Immunol 2011; 41:1709-19. [DOI: 10.1002/eji.201040945] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 02/15/2011] [Accepted: 03/11/2011] [Indexed: 12/30/2022]
|
39
|
Chen L, Lizano P, Zhao X, Sui X, Dhar SK, Shen YT, Vatner DE, Vatner SF, Depre C. Preemptive conditioning of the swine heart by H11 kinase/Hsp22 provides cardiac protection through inducible nitric oxide synthase. Am J Physiol Heart Circ Physiol 2011; 300:H1303-10. [PMID: 21317305 DOI: 10.1152/ajpheart.00979.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The second window of ischemic preconditioning (SWOP) provides maximal protection against ischemia through regulation of the inducible nitric oxide synthase (iNOS), yet its application is limited by the inconvenience of the preliminary ischemic stimulus required for prophylaxis. Overexpression of H11 kinase/Hsp22 (Hsp22) in a transgenic mouse model provides cardioprotection against ischemia that is equivalent to that conferred by SWOP. We hypothesized that short-term, prophylactic overexpression of Hsp22 would offer an alternative to SWOP in reducing ischemic damage through a nitric oxide (NO)-dependent mechanism. Adeno-mediated overexpression of Hsp22 was achieved in the area at risk of the left circumflex (Cx) coronary artery in chronically instrumented swine and compared with LacZ controls (n = 5/group). Hsp22-injected myocardium showed an average fourfold increase in Hsp22 protein expression compared with controls and a doubling in iNOS expression (both P < 0.05). Four days after ischemia-reperfusion, regional wall thickening was reduced by 58 ± 2% in the Hsp22 group vs. 82 ± 7% in the LacZ group, and Hsp22 reduced infarct size by 40% (both P < 0.05 vs. LacZ). Treatment with the NOS inhibitor N(G)-nitro-L-arginine (L-NNA) before ischemia suppressed the protection induced by Hsp22. In isolated cardiomyocytes, Hsp22 increased iNOS expression through the transcription factors NF-κB and STAT, the same effectors activated by SWOP, and reduced by 60% H(2)O(2)-mediated apoptosis, which was also abolished by NOS inhibitors. Therefore, short-term, prophylactic conditioning by Hsp22 provides NO-dependent cardioprotection that reproduces the signaling of SWOP, placing Hsp22 as a potential alternative for preemptive treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Li Chen
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, 07103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Willis MS, Patterson C. Hold me tight: Role of the heat shock protein family of chaperones in cardiac disease. Circulation 2010; 122:1740-51. [PMID: 20975010 DOI: 10.1161/circulationaha.110.942250] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Monte S Willis
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7126, USA
| | | |
Collapse
|
41
|
Abstract
Activation of the ubiquitin-proteasome system has been described in different models of cardiac hypertrophy. Cardiac cell growth in response to pressure or volume overload, as well as physiological adaptive hypertrophy, is accompanied by an increase in protein ubiquitination, proteasome subunit expression, and proteasome activity. Importantly, an inhibition of proteasome activity prevents and reverses cardiac hypertrophy and remodelling in vivo. The focus of this review is to provide an update about the mechanisms by which proteasome inhibitors affect cardiac cell growth in adaptive and maladaptive models of cardiac hypertrophy. In the first part, we summarize how the proteasome affects both proteolysis and protein synthesis in a context of cardiac cell growth. In the second part, we show how proteasome inhibition can prevent and reverse cardiac hypertrophy and remodelling in response to different conditions of overload.
Collapse
Affiliation(s)
- Nadia Hedhli
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ 07103, USA
| | | |
Collapse
|