1
|
Song J, Du J, Tan X, Li Y, Yu Q, Liu W, Zhu X, Cong B. Tissue kallikrein-related peptidase8 accentuates cardiac fibrosis after myocardial ischemia-reperfusion injury via regulation of cardiac fibroblasts. Life Sci 2023; 329:121973. [PMID: 37482211 DOI: 10.1016/j.lfs.2023.121973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
AIMS Tissue kallikrein-related peptidase8 (KLK8) has been found to mitigate acute myocardial ischemia-reperfusion (IR) injury. However, the effect of KLK8 on cardiac remodeling in response to IR injury has not been determined. MATERIALS AND METHODS KLK8 overexpressing transgenic rat (KLK8-TG) was used as the animal model. IR injury was induced by ligating the left anterior descending coronary artery for 1 h and subsequent reperfusion. The functional and morphological changes of the heart were examined 14 days after the injury. Neonatal rat cardiac fibroblasts (CFs) were used to investigate the molecular mechanisms in vitro. KEY FINDINGS KLK8 overexpression enhanced cardiac diastolic dysfunction, fibrosis, and hypertrophy after IR injury, indicating that KLK8 accentuated cardiac remodeling in response to IR injury. Moreover, KLK8 overexpression increased epidermal growth factor (EGF) release and promoted the phosphorylation of EGF receptor (EGFR) and ERK1/2 in the heart after IR injury. It was interesting to find that both EGFR antagonist (AG 1478) and MEK inhibitor (PD98059) attenuated the KLK8-induced proliferation and activation of CFs in vitro, indicating that EGFR signaling might mediate the pro-fibrotic action of KLK8. SIGNIFICANCE KLK8 plays a crucial role in cardiac remodeling after myocardial infarction. KLK8 accentuates cardiac fibrosis after IR injury, possibly mediated by EGFR signaling in CFs.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China; Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yang Li
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Qing Yu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China; Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen Liu
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
2
|
Peng Y, Wang L, Zhao X, Lai S, He X, Fan Q, He H, He M. Puerarin attenuates lipopolysaccharide-induced myocardial injury via the 14-3-3γ/PKCε pathway activating adaptive autophagy. Int Immunopharmacol 2022; 108:108905. [DOI: 10.1016/j.intimp.2022.108905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022]
|
3
|
Rex DAB, Vaid N, Deepak K, Dagamajalu S, Prasad TSK. A comprehensive review on current understanding of bradykinin in COVID-19 and inflammatory diseases. Mol Biol Rep 2022; 49:9915-9927. [PMID: 35596055 PMCID: PMC9122735 DOI: 10.1007/s11033-022-07539-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/28/2022] [Indexed: 12/28/2022]
Abstract
Bradykinin, a member of the kallikrein–kinin system (KKS), is a potent, short-lived vasoactive peptide that acts as a vasodilator and an inflammatory mediator in a number of signaling mechanisms. Bradykinin induced signaling is mediated through kinin B1 (BDKRB1) and B2 (BDKRB2) transmembrane receptors coupled with different subunits of G proteins (Gαi/Gα0, Gαq and Gβ1γ2). The bradykinin-mediated signaling mechanism activates excessive pro-inflammatory cytokines, including IL-6, IL-1β, IL-8 and IL-2. Upregulation of these cytokines has implications in a wide range of clinical conditions such as inflammation leading to fibrosis, cardiovascular diseases, and most recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In SARS-CoV-2 infection, bradykinin is found to be at raised levels and is reported to trigger a diverse array of symptoms. All of this brings bradykinin to the core point as a molecule of immense therapeutic value. Our understanding of its involvement in various pathways has expanded with time. Therefore, there is a need to look at the overall picture that emerges from the developments made by deciphering the bradykinin mediated signaling mechanisms involved in the pathological conditions. It will help devise strategies for developing better treatment modalities in the implicated diseases. This review summarizes the current state of knowledge on bradykinin mediated signaling in the diverse conditions described above, with a marked emphasis on the therapeutic potential of targeting the bradykinin receptor.
Collapse
Affiliation(s)
- Devasahayam Arokiar Balaya Rex
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Neelanchal Vaid
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - K Deepak
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|
4
|
Qian M, Shi Y, Lu W. LINC00707 knockdown inhibits IL-1β-induced apoptosis and extracellular matrix degradation of osteoarthritis chondrocytes by the miR-330-5p/FSHR axis. Immunopharmacol Immunotoxicol 2022; 44:671-681. [PMID: 35536095 DOI: 10.1080/08923973.2022.2076241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is a severe disabling condition that causes major health problems. The roles of lncRNAs in regulating OA progression have been increasingly researched. Based on previously published microarray analysis, LINC00707 is upregulated in OA. This research was done to uncover the function of LINC00707 in IL-1β-induced chondrocyte injury and its possible mechanisms. METHODS LINC00707, miR-330-5p, and FSHR expression in OA cartilage tissues were assessed by RT-qPCR. Primary chondrocytes were isolated from OA tissues and treated with IL-1β to establish an in vitro OA model. Under the indicated treatment, chondrocyte apoptosis, senescence, ECM degradation, and inflammation were determined using flow cytometry, TUNEL, SA-β-Gal staining, and ELISA experiments, respectively. Interactions between gene were evaluated using Ago2 RIP and luciferase reporter assays. RESULTS LINC00707 and FSHR were elevated, and miR-330-5p was reduced in cartilage tissues of OA patients and in IL-1β-treated primary chondrocytes. Silencing LINC00707 hampered chondrocytes apoptosis, senescence, ECM degradation, and inflammation. LINC00707 acted as a ceRNA to regulate FSHR through controlling miR-330-5p availability. Additionally, both miR-330-5p depletion and FSHR overexpression diminished the effects of silencing LINC00707 in OA progression. CONCLUSION Silencing LINC00707 mitigates chondrocyte injury in osteoarthritis via sponging miR-330-5p and inhibiting FSHR.
Collapse
Affiliation(s)
- Minglei Qian
- Department of Traumatology, Changshu No.2 People's Hospital, Changshu 215500, Jiangsu, China
| | - Yuanxin Shi
- Department of Traumatology, Changshu No.2 People's Hospital, Changshu 215500, Jiangsu, China
| | - Wei Lu
- Department of Traumatology, Changshu No.2 People's Hospital, Changshu 215500, Jiangsu, China
| |
Collapse
|
5
|
Petchdee S, Yalong M, Kaewnet M, Ithisariyanont B, Padawong T. Assessment of Right Ventricular Function, Blood Lactate Levels, and Serum Peptidomics Profiles Associated With Mitral Valve Disease in Dogs. Front Vet Sci 2022; 8:789137. [PMID: 35127880 PMCID: PMC8811185 DOI: 10.3389/fvets.2021.789137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 11/18/2022] Open
Abstract
Background Degenerative mitral valve disease is a common heart problem in dogs. The aims are to evaluate the relationships between right and left ventricular function, and blood lactate concentrations, assess prognostic contribution, and investigate whether serum peptidomics profile could reveal markers or determine the stage in dogs with valve degeneration. Materials and Methods Ninety-three dogs were evaluated in this study. Thirty-nine dogs' serum was collected and analyzed using matrix-assisted laser desorption/ionization-time of flight mass spectrometry. The Kaplan–Meier curve was used to predict the outcomes of mitral valve disease. Follow-up was obtained by a questionnaire or telephone to determine a survival time. Results The BUN/creatinine ratio, vertebral heart score, and left atrium/aorta ratio were the independent predictors of cardiac mortality. Right ventricular systolic dysfunction was found in 50% of dogs with mitral valve disease. Dogs with right ventricular dysfunction had a significantly higher incidence of lower fractional shortening and larger right ventricular dimensions. The occurrence of right-sided dysfunction is proportionate to age and the degree of left ventricular dysfunction. High blood lactate concentrations were investigated in dogs with mitral valve disease stage C compared with stage B. The peptides such as mitogen-activated protein kinase, kallikrein, and tenascin-C appeared in the heart disease progression group. Conclusion Right-hearted function assessment, blood lactate levels, and peptidomics analysis may help early detection and prognosis of this disease in dogs. Peptidomics profiles from this study demonstrate the possibility for prognosis indicators of heart valve degeneration.
Collapse
|
6
|
Song J, Du J, Tan X, Wu Z, Yuan J, Cong B. Dexmedetomidine protects the heart against ischemia reperfusion injury via regulation of the bradykinin receptors. Eur J Pharmacol 2021; 911:174493. [PMID: 34506777 DOI: 10.1016/j.ejphar.2021.174493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Dexmedetomidine (DEX) has been reported to protect the heart against ischemia reperfusion (I/R) injury. However, the exact mechanisms are still not fully understood. METHODS AND RESULTS A rat cardiac I/R injury model was induced by ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion for 2 h, and DEX was administered intravenously 30 min before ischemia. We confirmed that DEX treatment mitigated cardiac I/R injury. Interestingly, we found that DEX regulated the expression of bradykinin (BK) receptors (B1R and B2R) in rat hearts during I/R injury and enhanced the protective action of BK administered during reperfusion. Moreover, in vitro hypoxia reoxygenation (H/R) injury was induced in neonatal rat cardiomyocytes (CMs), and DEX was administered 1 h before hypoxia. The in vitro findings were consistent with the in vivo experiments. We found that an α2-adrenoceptor (α2-AR) antagonist (yohimbine) completely aborted DEX-induced B1R and B2R regulation; an adenylyl cyclase (AC) agonist (forskolin) blocked B1R downregulation, while a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) blocked B2R upregulation. The above findings indicated that DEX interacted with α2-AR in cardiomyocytes, inhibited B1R expression via suppression of AC, and stimulated B2R expression via activation of PI3K. CONCLUSIONS DEX regulates BK receptor expression and potentiates the protection of BK in cardiac I/R injury, which suggests that modulating endogenous cardioprotective factors may play an important role in DEX-induced cardioprotection.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China.
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Zhaotang Wu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Jihong Yuan
- Department of Nephropathy, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| |
Collapse
|
7
|
Freitas-Lima LC, Budu A, Estrela GR, da Silva TA, Arruda AC, de Carvalho Araujo R. Metabolic fasting stress is ameliorated in Kinin B1 receptor-deficient mice. Life Sci 2021; 294:120007. [PMID: 34600938 DOI: 10.1016/j.lfs.2021.120007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
The liver has an essential role in responding to metabolic demands under stress conditions. The organ stores, releases, and recycles metabolism-related substrates. However, it is not clear how the Kallikrein-Kinin System modulates metabolic flexibility shift between energetic sources. AIMS To analyze the hepatic metabolism in kinin B1 receptor deficient mice (B1KO mice) under fasting conditions. MAIN METHODS WT and B1KO male mice were allocated in a calorimetric cage for 7 days and 48 h before the euthanasia, half of the animals of both groups were under fasting conditions. Biochemical parameters, ketone bodies (KB), and gene expression involving the liver energetic metabolism genes were evaluated. KEY FINDINGS Kinin B1 receptor (B1R) modulates the metabolic shift under fasting conditions, reducing the VO2 expenditure. A preference for carbohydrates as an energetic source is suggested, as the B1KO group did not display an increase in KB in the serum. Moreover, the B1KO animals displayed higher serum triglycerides concentration compared to WT fasting mice. Interestingly, the lack of B1R induces the increase expression of enzymes from the glycolysis and lipolysis pathways under the fed. However, under fasting, the enzymatic expression of gluconeogenesis, glyceroneogenesis, and ketogenesis of these pathways does not occur, suggesting an absence of the shift metabolism responsivity, and this condition is modulated by PDK4 under FOXO1 control. SIGNIFICANCE B1R has an important role in the hepatic glucose metabolism, which in turn influences the energetic metabolism, and in long-term outcomes, such as in the decrease in hepatic glycogen stores and in the enhancement of hepatic metabolism.
Collapse
Affiliation(s)
| | - Alexandre Budu
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Gabriel Rufino Estrela
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil; Department of Clinical and Experimental Oncology, Discipline of Hematology and Hematotherapy, Federal University of São Paulo, 04037002 São Paulo, Brazil.
| | - Thais Alves da Silva
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Adriano Cleis Arruda
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Ronaldo de Carvalho Araujo
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil; Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
8
|
Shen M, Yu M, Qiu C, Zhang G, Li J, Fang W, Wang Q. Myocardial angiogenesis induced by exercise training involves a regulatory mechanism mediated by kinin receptors. Clin Exp Hypertens 2021; 43:408-415. [PMID: 33687297 DOI: 10.1080/10641963.2021.1896725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To demonstrate that the kallikrein-kinin system (KKS) is upstream of angiogenic signaling pathway, and to determine the role of the kinin B1 and B2 receptors in myocardial angiogenesis induced by exercise training. METHODS Forty Wistar rats were randomly assigned to an exercise control (EC) group, a B1 receptor antagonist (B1Ant) group, a B2 receptor antagonist (B2Ant) group, and a double receptor antagonist ((B1+ B2)Ant) group. A myocardial infarction model was employed. Animals in all groups received 30 min of exercise training for 4 weeks. The expression of VEGF and eNOS, capillary supply, and apoptosis rate were evaluated. RESULTS The mRNA and protein expression of VEGF and eNOS showed similar trends in all groups, and were lowest in the (B1+ B2) Ant group, and highest in the EC group. Levels of VEGF and eNOS mRNA were significantly lower in the B1Ant group than in the B2Ant group (p< .001 and p< .05, respectively). VEGF and eNOS protein in the B1Ant group was also significantly lower (p< .01 and p< .05, respectively) than in the B2Ant group. The capillary numbers in the (B1+ B2) Ant group were significantly lower than in the EC group (395.8 ± 105 vs. 1127.9 ± 192.98, respectively). The apoptosis rate of cardiomyocytes was highest in the (B1+ B2) Ant group. CONCLUSION KKS may act as an upstream signal transduction pathway for angiogenic factors in myocardial angiogenesis. The B1 and B2 receptors exert additive effects, and the B1 receptor has the most prominent role in mediating KKS-induced myocardial angiogenesis.
Collapse
MESH Headings
- Animals
- Capillaries/metabolism
- Kinins/metabolism
- Male
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Neovascularization, Physiologic
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Physical Conditioning, Animal
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Rats
Collapse
Affiliation(s)
- Mei Shen
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Min Yu
- Department of Rehabilitation Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Chengxiu Qiu
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Ge Zhang
- Department of Electrocardiogram, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Jingya Li
- Department of Rehabilitation Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Wei Fang
- Department of Nursing, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| | - Qiwen Wang
- Department of Rehabilitation Medicin, The People's Hospital of Longhua District, Shenzhen, Guangdong Province, China
| |
Collapse
|
9
|
Yang H, Li GP, Liu Q, Zong SB, Li L, Xu ZL, Zhou J, Cao L, Wang ZZ, Zhang QC, Li M, Fan QR, Hu HF, Xiao W. Neuroprotective effects of Ginkgolide B in focal cerebral ischemia through selective activation of prostaglandin E2 receptor EP4 and the downstream transactivation of epidermal growth factor receptor. Phytother Res 2021; 35:2727-2744. [PMID: 33452698 DOI: 10.1002/ptr.7018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 11/07/2022]
Abstract
The present study was undertaken to identify whether prostaglandin E2 receptor is the potential receptor/binding site for Ginkgolide A, Ginkgolide B, Ginkgolide K, and Bilobalide, the four main ingredients of the Ginkgo biloba L., leaves. Using functional assays, we identified EP4, coupled with Gs protein, as a target of Ginkgolide B. In human neuroblastoma SH-SY5Y cells suffered from oxygen-glucose deprivation/reperfusion, Ginkgolide B-activated PKA, Akt, and ERK1/2 as well as Src-mediated transactivation of epidermal growth factor receptor. These resulted in downstream signaling pathways, which enhanced cell survival and inhibited apoptosis. Knockdown of EP4 prevented Ginkgolide B-mediated Src, epidermal growth factor receptor (EGFR), Akt, and ERK1/2 phosphorylation and neuroprotective effects. Moreover, Src inhibitor prevented Ginkgolide B-mediated EGFR transactivation and the downstream Akt and ERK1/2 activation, while the phosphorylation of PKA induced by Ginkgolide B was not affected, indicating Ginkgolide B might transactivate EGFR in a ligand-independent manner. EP4 knockdown in a rat middle cerebral artery occlusion (MCAO) model prevented Ginkgolide B-mediated infarct size reduction and neurological assessment improvement. At the same time, the increased expressions of p-Akt, p-ERK1/2, p-PKA, p-Src, and p-EGFR and the deceased expression of cleaved capases-3 induced by Ginkgolide B in cerebral cortex were blocked due to EP4 knockdown. In conclusion, Ginkgolide B exerts neuroprotective effects in rat MCAO model through the activation of EP4 and the downstream transactivation of EGFR.
Collapse
Affiliation(s)
- Hao Yang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Gui-Ping Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Qiu Liu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Shao-Bo Zong
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Liang Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhi-Liang Xu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Jun Zhou
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Liang Cao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhen-Zhong Wang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Quan-Chang Zhang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Ming Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Qi-Ru Fan
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Han-Fei Hu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| |
Collapse
|
10
|
Yang W, Lai Q, Zhang L, Zhang Y, Zhang Y, Yu B, Li F, Kou J. Mechanisms dissection of the combination GRS derived from ShengMai preparations for the treatment of myocardial ischemia/reperfusion injury. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113381. [PMID: 32946961 DOI: 10.1016/j.jep.2020.113381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 08/12/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Recently, a new drug combination GRS comprising ginsenoside Rb1 (G-Rb1), ruscogenin (R-Rus) and schisandrin (S-SA) was screened based on ShengMai preparations, which exhibited a prominent cardioprotective effects against myocardial ischemia/reperfusion (MI/R) injury. AIM OF THE STUDY To investigate their systemic and individual mechanism of each compound in combination GRS. MATERIALS AND METHODS The mice model of MI/R and hypoxia/reoxygenation (H/R)-induced cardiomyocytes injury were performed to explore the respective characteristics of each compound in GRS against myocardial injury. RESULTS Each component in the combination GRS attenuated MI/R injury as evidenced by decreased myocardial infarct size, ameliorated histological features, and improved biochemical indicators. Meanwhile, ingredient G, R and S in combination also individually performed a significant decrease of apoptotic index in MI/R mice and H/R-induced cardiomyocytes injury. Mechanistically, component G in GRS could markedly increase the ATP content in cardiomyocytes through activation of AMPKα phosphorylation. Interestingly, the anti-apoptotic actions of G were profoundly attenuated by knockdown of AMPKα, while no alteration was observed on composition R and S. Moreover, component R in GRS significantly reduced the IL-6 and TNF-α mRNA expression, as well as the content of IL-6 via the modulation of NF-κB signaling pathway. Further, component S exhibited the most powerful anti-oxidative capacity in GRS and remarkably decreased the production of MDA and ROS, and potential mechanisms might at least in part through activating the Akt-14-3-3 signaling pathway and inhibiting the phosphorylation of Bad and ERK1/2. CONCLUSIONS Our results indicated that the respective mechanism of each compound in combination GRS against MI/R injury might closely associated with energy metabolism modulation, suppression of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Weiwei Yang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Qiong Lai
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Ling Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Yu Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, PR China.
| |
Collapse
|
11
|
Lü G, Li L, Wang B, Kuang L. LINC00623/miR-101/HRAS axis modulates IL-1β-mediated ECM degradation, apoptosis and senescence of osteoarthritis chondrocytes. Aging (Albany NY) 2020; 12:3218-3237. [PMID: 32062610 PMCID: PMC7066905 DOI: 10.18632/aging.102801] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/19/2020] [Indexed: 05/12/2023]
Abstract
Chondrocyte apoptosis and extracellular matrix (ECM) degeneration have been implicated in the pathogenesis of osteoarthritis (OA). Based on previously reported microarray analysis, HRAS (Harvey rat sarcoma viral oncogene homolog), a member of the RAS protein family, was chosen as a potential regulator of OA chondrocyte apoptosis and ECM degradation. HRAS expression was downregulated in OA tissues, particularly in mild-OA tissues. HRAS overexpression partially attenuated IL-1β-induced OA chondrocyte apoptosis and ECM degradation. Similar to HRAS, the long non-coding RNA LINC00623 was downregulated in OA tissues. LINC00623 knockdown enhanced IL-1β-induced OA chondrocyte apoptosis and ECM degradation, which could be partially reversed by HRAS overexpression. It has been reported that lncRNAs act as ceRNAs of miRNAs to exert their function. Herein, miR-101 was predicted to bind to both LINC00623 and HRAS, which was further confirmed by luciferase reporter and RIP assays. LINC00623 competed with HRAS for miR-101 binding, therefore reducing the inhibitory effect of miR-101 on HRAS expression. More importantly, the effect of LINC00623 was partially eliminated by miR-101 inhibition. Overall, the LINC00623/miR-101/HRAS axis modulates OA chondrocyte apoptosis, senescence and ECM degradation through MAPK signaling, which might play a critical role in OA development.
Collapse
Affiliation(s)
- Guohua Lü
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lei Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lei Kuang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
12
|
Huang M, Du J, Wang Y, Ma S, Hu T, Shang J, Yu Q, Zhu X, Zhang G, Cong B. Tissue kallikrein-related peptidase8 protects rat heart against acute ischemia reperfusion injury. Int J Biol Macromol 2019; 140:1126-1133. [PMID: 31449861 DOI: 10.1016/j.ijbiomac.2019.08.195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 01/06/2023]
Abstract
Tissue kallikrein-related peptidases (KLKs) play important roles in acute cardiac injury and cardiac remodeling. However, the exact cardiac actions of KLK8 have not been determined. Transgenic rat overexpressing KLK8 was established to examine the role of KLK8 in the heart. Cardiac injury was induced by ischemia/reperfusion (I/R) and examined by infarct size measurement and TUNEL staining. The molecular mechanisms were investigated in cultured neonatal rat cardiomyocytes (CMs). Western blot analysis was used to determine the protein levels. KLK8 protein level was significantly increased in the cardiac ischemic risk area. KLK8 overexpression mitigated I/R-induced cardiac injury, as evidenced by decreased infarct size and apoptosis in cardiac ischemic risk area in vivo. Via in vitro studies, it was found that KLK8 overexpression attenuated the Hypoxia/Reoxygenation (H/R) injury in CMs; both B2R and PAR2 antagonist significantly attenuated KLK8-induced protective actions under H/R injury. Moreover, KLK8 overexpressed CMs showed significant higher phosphorylation levels of Akt, ERK1/2 and PKA under H/R stimulation; B2R antagonist attenuated the phosphorylation levels of Akt and ERK1/2, while PAR2 antagonist attenuated the phosphorylation levels of PKA and ERK1/2. KLK8 protects the heart against I/R-induced cardiac injury, which may represent a new therapeutic target in cardiac medicine.
Collapse
Affiliation(s)
- Meineng Huang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Jiankui Du
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Yifei Wang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Shiyu Ma
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Ting Hu
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Jing Shang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Qing Yu
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Guanxin Zhang
- Department of Cardiothoracic Surgery, Changhai Hospital, Shanghai 200433, China.
| | - Binhai Cong
- Department of Physiology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
13
|
Micheu MM, Scarlatescu AI, Scafa-Udriste A, Dorobantu M. The Winding Road of Cardiac Regeneration-Stem Cell Omics in the Spotlight. Cells 2018; 7:255. [PMID: 30544622 PMCID: PMC6315576 DOI: 10.3390/cells7120255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/18/2022] Open
Abstract
Despite significant progress in treating ischemic cardiac disease and succeeding heart failure, there is still an unmet need to develop effective therapeutic strategies given the persistent high-mortality rate. Advances in stem cell biology hold great promise for regenerative medicine, particularly for cardiac regeneration. Various cell types have been used both in preclinical and clinical studies to repair the injured heart, either directly or indirectly. Transplanted cells may act in an autocrine and/or paracrine manner to improve the myocyte survival and migration of remote and/or resident stem cells to the site of injury. Still, the molecular mechanisms regulating cardiac protection and repair are poorly understood. Stem cell fate is directed by multifaceted interactions between genetic, epigenetic, transcriptional, and post-transcriptional mechanisms. Decoding stem cells' "panomic" data would provide a comprehensive picture of the underlying mechanisms, resulting in patient-tailored therapy. This review offers a critical analysis of omics data in relation to stem cell survival and differentiation. Additionally, the emerging role of stem cell-derived exosomes as "cell-free" therapy is debated. Last but not least, we discuss the challenges to retrieve and analyze the huge amount of publicly available omics data.
Collapse
Affiliation(s)
- Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alina Ioana Scarlatescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| |
Collapse
|
14
|
Tanshinone IIA Pretreatment Protects H9c2 Cells against Anoxia/Reoxygenation Injury: Involvement of the Translocation of Bcl-2 to Mitochondria Mediated by 14-3-3 η. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3583921. [PMID: 30050654 PMCID: PMC6046124 DOI: 10.1155/2018/3583921] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/09/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022]
Abstract
Tanshinone IIA is an important component that is isolated from danshen (Salvia miltiorrhiza), which is known to be beneficial for cardiovascular health. In this study, we determined the effects of Tanshinone IIA and its underlying mechanisms of action in an anoxia/reoxygenation (A/R) cell line model. Prior to inducing A/R injury, rat cardiomyocyte-derived cell line H9c2 was stimulated with 8 μM of Tanshinone IIA for 48 hours. When compared with the A/R group, the Tanshinone IIA treatment significantly increased cell viability and decreased lactate dehydrogenase activity. Tanshinone IIA upregulated 14-3-3η expression and facilitated Bcl-2 translocation to the mitochondrial outer membrane, which bound with voltage-dependent anion channel 1. In addition, pretreatment with Tanshinone IIA reduced the generation of reactive oxygen species and cytochrome c release, inactivated caspase-3, prevented mitochondrial permeability transition pore opening, and reduced the percentage of apoptotic cells. Moreover, treatment with Tanshinone IIA reduced the level of malondialdehyde, thereby increasing the activity of superoxide dismutase and glutathione peroxidase. Silencing the expression of 14-3-3η by adenovirus blocked the above-mentioned results. These novel findings showed that pretreatment with Tanshinone IIA alleviated H9c2 cell damage against A/R injury and was associated with upregulation of 14-3-3η, thereby facilitating Bcl-2 translocation to the mitochondrial outer membrane and preventing mitochondrial permeability transition pore opening, decreasing cytochrome c release, preventing caspase-3 activation, and restraining apoptosis.
Collapse
|
15
|
Tang Z, Cui K, Luan Y, Ruan Y, Wang T, Yang J, Wang S, Liu J, Wang D. Human tissue kallikrein 1 ameliorates erectile function via modulation of macroautophagy in aged transgenic rats. Andrology 2018; 6:766-774. [PMID: 29939496 DOI: 10.1111/andr.12512] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/03/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022]
Abstract
Previously, we have demonstrated that human tissue kallikrein 1 (hKLK1) improves age-related erectile dysfunction (ED). Autophagy has been implicated in age-related diseases, including ED. However, the molecular mechanisms underlying hKLK1-mediated amelioration of age-related ED via regulation of autophagy remains unknown. To explore the potential mechanism, male wild-type Sprague-Dawley rats (WTR) and transgenic rats harboring human KLK1 (TGR) were bred till 4 or 18 months of age and divided into three groups: young WTR (yWTR) as the control group, aged WTR (aWTR) group, and aged TGR (aTGR) group. The erectile function of each rat was evaluated using cavernous nerve electrostimulation. The ratio of intracavernous pressure/mean arterial pressure (ICP/MAP) and total ICP were also measured. Western blotting, immunohistochemistry, and transmission electron microscopy were performed to detect the levels of autophagy. The expression levels of related signaling pathways were determined by western blotting and immunohistochemistry. We found that hKLK1 improved the impaired erectile function of aged rats. Compared to the yWTR and aTGR groups, the aWTR group showed reduced smooth muscle/collagen ratio, fewer autophagosomes, and lower expression of Beclin 1 and LC3-II, which indicate impaired smooth muscle function and low level of autophagy in the smooth muscle cells. Moreover, the PI3K/Akt/mTOR signaling pathway, which is considered to be a negative regulator of autophagy, was upregulated in the aWTR group. hKLK1 may partially restore erectile function in aged transgenic rats by upregulating protective autophagy via the PI3K/Akt/mTOR pathway. These observations indicate that hKLK1 is a potential gene therapy candidate for age-related ED.
Collapse
Affiliation(s)
- Z Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - K Cui
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Luan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Ruan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - T Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - S Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - D Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Si H, Zhang Y, Song Y, Li L. Overexpression of adrenomedullin protects mesenchymal stem cells against hypoxia and serum deprivation‑induced apoptosis via the Akt/GSK3β and Bcl‑2 signaling pathways. Int J Mol Med 2018; 41:3342-3352. [PMID: 29512737 PMCID: PMC5881801 DOI: 10.3892/ijmm.2018.3533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/02/2018] [Indexed: 01/12/2023] Open
Abstract
The poor survival rate of transplanted mesenchymal stem cells (MSCs) within the ischemic heart limits their therapeutic potential for cardiac repair. Adrenomedullin (ADM) has been identified as a potent apoptotic inhibitor. The present study aimed to investigate the protective effects of ADM on MSCs against hypoxia and serum deprivation (H/SD)‑induced apoptosis, and to determine the potential underlying mechanisms. In the present study, a recombinant adenovirus expressing the ADM gene was established and was infected into MSCs. The infection rate was determined via microscopic detection of green fluorescence and flow cytometric analysis. The mRNA expression levels of ADM were detected by reverse transcription‑polymerase chain reaction. In addition, a model of H/SD was generated. The MSCs were randomly separated into six groups: Control, enhanced green fluorescent protein (EGFP)‑Adv, EGFP‑ADM, H/SD, EGFP‑Adv + H/SD and EGFP‑ADM + H/SD. Cell viability and proliferation were determined using the Cell Counting kit‑8 assay. Apoptosis was assessed by terminal deoxynucleotidyl transferase‑mediated‑dUTP nick‑end labeling assay and flow cytometric analysis using Annexin V‑phycoerythrin/7‑aminoactinomycin D staining. The protein expression levels of total protein kinase B (Akt), phosphorylated (p)‑Akt, total glycogen synthase kinase (GSK)3β, p‑GSK3β, B‑cell lymphoma 2 (Bcl‑2), Bcl‑2‑associated X protein (Bax), caspase‑3 and cleaved caspase‑3 were detected by western blot analysis. The results indicated that ADM overexpression could improve MSC proliferation and viability, and protect MSCs against H/SD‑induced apoptosis. In addition, ADM overexpression increased Akt and GSK3β phosphorylation, and Bcl‑2/Bax ratio, and decreased the activation of caspase‑3. These results suggested that ADM protects MSCs against H/SD‑induced apoptosis, which may be mediated via the Akt/GSK3β and Bcl‑2 signaling pathways.
Collapse
Affiliation(s)
- Hongjin Si
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Yao Zhang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Yuqing Song
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| | - Lili Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
17
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
18
|
Neuroprotection of bradykinin/bradykinin B2 receptor system in cerebral ischemia. Biomed Pharmacother 2017; 94:1057-1063. [DOI: 10.1016/j.biopha.2017.08.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022] Open
|
19
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|
20
|
Eupatilin inhibits the apoptosis in H9c2 cardiomyocytes via the Akt/GSK-3β pathway following hypoxia/reoxygenation injury. Biomed Pharmacother 2016; 82:373-8. [DOI: 10.1016/j.biopha.2016.05.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 11/20/2022] Open
|
21
|
Kallikrein-related peptidase 8 is expressed in myocardium and induces cardiac hypertrophy. Sci Rep 2016; 7:20024. [PMID: 26823023 PMCID: PMC4731818 DOI: 10.1038/srep20024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
The tissue kallikrein-related peptidase family (KLK) is a group of trypsin- and chymotrypsin-like serine proteases that share a similar homology to parent tissue kallikrein (KLK1). KLK1 is identified in heart and has anti-hypertrophic effects. However, whether other KLK family members play a role in regulating cardiac function remains unknown. In the present study, we demonstrated for the first time that KLK8 was expressed in myocardium. KLK8 expression was upregulated in left ventricle of cardiac hypertrophy models. Both intra-cardiac adenovirus-mediated and transgenic-mediated KLK8 overexpression led to cardiac hypertrophy in vivo. In primary neonatal rat cardiomyocytes, KLK8 knockdown inhibited phenylephrine (PE)-induced cardiomyocyte hypertrophy, whereas KLK8 overexpression promoted cardiomyocyte hypertrophy via a serine protease activity-dependent but kinin receptor-independent pathway. KLK8 overexpression increased epidermal growth factor (EGF) production, which was blocked by the inhibitors of serine protease. EGF receptor (EGFR) antagonist and EGFR knockdown reversed the hypertrophy induced by KLK8 overexpression. KLK8-induced cardiomyocyte hypertrophy was also significantly decreased by blocking the protease-activated receptor 1 (PAR1) or PAR2 pathway. Our data suggest that KLK8 may promote cardiomyocyte hypertrophy through EGF signaling- and PARs-dependent but a kinin receptor-independent pathway. It is implied that different KLK family members can subtly regulate cardiac function and remodeling.
Collapse
|
22
|
Tissue Kallikrein Activity, Detected by a Novel Method, May Be a Predictor of Recurrent Stroke: A Case-Control Study. DISEASE MARKERS 2015; 2015:159750. [PMID: 26451066 PMCID: PMC4584216 DOI: 10.1155/2015/159750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/24/2015] [Accepted: 08/30/2015] [Indexed: 11/21/2022]
Abstract
Aim. Tissue kallikrein (TK) protein content in plasma has been shown to be negatively associated with both incident and recurrent strokes. The aims of this study were to develop a novel method for detecting TK activity and to investigate its association with event-free survival over 5 years in Chinese first-ever stroke patients. Methods. We designed a case-control study with 321 stroke patients (174: ischemic stroke, 147: hemorrhagic stroke) and 323 healthy local controls. TK activity was measured by a novel assay utilizing the immunological characteristics of TK and the catalysis of benzoyl arginine ethyl ester hydrochloride (BAEE). Results. TK protein levels above 0.200 mg/L in plasma were not associated with urinary TK activity or the risk of stroke recurrence. TK activity was significantly lower in stroke patients compared with controls (1.583 ± 0.673 Eu/mL versus 1.934 ± 0.284 Eu/mL, P < 0.001). After adjusting for traditional risk factors, TK activity was negatively associated, in a dose-response manner, with the risk of overall stroke recurrence and positively associated with event-free survival during a 5-year follow-up (relative risk (RR), 0.69; 95% CI, 0.57–0.84; P < 0.001). Conclusions. Our findings suggest that urinary TK activity may be a stronger predictor of stroke recurrence than plasma TK levels.
Collapse
|
23
|
Das J, Kang MH, Kim E, Kwon DN, Choi YJ, Kim JH. Hexavalent chromium induces apoptosis in male somatic and spermatogonial stem cells via redox imbalance. Sci Rep 2015; 5:13921. [PMID: 26355036 PMCID: PMC4564811 DOI: 10.1038/srep13921] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023] Open
Abstract
Hexavalent chromium [Cr(VI)], an environmental toxicant, causes severe male reproductive abnormalities. However, the actual mechanisms of toxicity are not clearly understood and have not been studied in detail. The present in vitro study aimed to investigate the mechanism of reproductive toxicity of Cr(VI) in male somatic cells (mouse TM3 Leydig cells and TM4 Sertoli cells) and spermatogonial stem cells (SSCs) because damage to or dysfunction of these cells can directly affect spermatogenesis, resulting in male infertility. Cr(VI) by inducing oxidative stress was cytotoxic to both male somatic cells and SSCs in a dose-dependent manner, and induced mitochondria-dependent apoptosis. Although the mechanism of Cr(VI)-induced cytotoxicity was similar in both somatic cells, the differences in sensitivity of TM3 and TM4 cells to Cr(VI) could be attributed, at least in part, to cell-specific regulation of P-AKT1, P-ERK1/2, and P-P53 proteins. Cr(VI) affected the differentiation and self-renewal mechanisms of SSCs, disrupted steroidogenesis in TM3 cells, while in TM4 cells, the expression of tight junction signaling and cell receptor molecules was affected as well as the secretory functions were impaired. In conclusion, our results show that Cr(VI) is cytotoxic and impairs the physiological functions of male somatic cells and SSCs.
Collapse
Affiliation(s)
- Joydeep Das
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| | - Min-Hee Kang
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| | - Eunsu Kim
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| | - Deug-Nam Kwon
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| | - Yun-Jung Choi
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| | - Jin-Hoi Kim
- Department of Animal Biotechnology, College of Animal Bioscience and Biotechnology/Animal Resources Research Center, Konkuk University, Seoul 143-701, South Korea
| |
Collapse
|
24
|
Hu WS, Ting WJ, Chiang WD, Pai P, Yeh YL, Chang CH, Lin WT, Huang CY. The Heart Protection Effect of Alcalase Potato Protein Hydrolysate Is through IGF1R-PI3K-Akt Compensatory Reactivation in Aging Rats on High Fat Diets. Int J Mol Sci 2015; 16:10158-72. [PMID: 25950762 PMCID: PMC4463638 DOI: 10.3390/ijms160510158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity is high in older adults. Alcalase potato protein hydrolysate (APPH), a nutraceutical food, might have greater benefits and be more economical than hypolipidemic drugs. In this study, serum lipid profiles and heart protective effects were evaluated in high fat diet (HFD) induced hyperlipidemia in aging rats treated with APPH (15, 45 and 75 mg/kg/day) and probucol (500 mg/kg/day). APPH treatments reduced serum triacylglycerol (TG), total cholesterol (TC), and low density lipoprotein (LDL) levels to the normal levels expressed in the control group. Additionally, the IGF1R-PI3K-Akt survival pathway was reactivated, and Fas-FADD (Fas-associated death domain) induced apoptosis was inhibited by APPH treatments (15 and 45 mg/kg/day) in HFD aging rat hearts. APPH (75 mg/kg/day) rather than probucol (500 mg/kg/day) treatment could reduce serum lipids without affecting HDL expression. The heart protective effect of APPH in aging rats with hyperlipidemia was through lowering serum lipids and enhancing the activation of the compensatory IGF1R-PI3K-Akt survival pathway.
Collapse
Affiliation(s)
- Wei-Syun Hu
- PhD Program for Aging, China Medical University, Taichung 40402, Taiwan.
| | - Wei-Jen Ting
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Wen-Dee Chiang
- Department of Food Science, College of Agriculture, Tunghai University, Taichung 40704, Taiwan.
| | - Peiying Pai
- Division of Cardiology, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Yu-Lan Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan.
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town 35053, Taiwan.
| | - Wan-Teng Lin
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung 40704, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
25
|
Negraes PD, Trujillo CA, Pillat MM, Teng YD, Ulrich H. Roles of kinins in the nervous system. Cell Transplant 2015; 24:613-23. [PMID: 25839228 DOI: 10.3727/096368915x687778] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The kallikrein-kinin system (KKS) is an endogenous pathway involved in many biological processes. Although primarily related to blood pressure control and inflammation, its activation goes beyond these effects. Neurogenesis and neuroprotection might be stimulated by bradykinin being of great interest for clinical applications following brain injury. This peptide is also an important player in spinal cord injury pathophysiology and recovery, in which bradykinin receptor blockers represent substantial therapeutic potential. Here, we highlight the participation of kinin receptors and especially bradykinin in mediating ischemia pathophysiology in the central and peripheral nervous systems. Moreover, we explore the recent advances on mechanistic and therapeutic targets for biological, pathological, and neural repair processes involving kinins.
Collapse
Affiliation(s)
- Priscilla D Negraes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
26
|
Chao J, Bledsoe G, Chao L. Kallikrein-kinin in stem cell therapy. World J Stem Cells 2014; 6:448-457. [PMID: 25258666 PMCID: PMC4172673 DOI: 10.4252/wjsc.v6.i4.448] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/27/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
The tissue kallikrein-kinin system exerts a wide spectrum of biological activities in the cardiovascular, renal and central nervous systems. Tissue kallikrein-kinin modulates the proliferation, viability, mobility and functional activity of certain stem cell populations, namely mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), mononuclear cell subsets and neural stem cells. Stimulation of these stem cells by tissue kallikrein-kinin may lead to protection against renal, cardiovascular and neural damage by inhibiting apoptosis, inflammation, fibrosis and oxidative stress and promoting neovascularization. Moreover, MSCs and EPCs genetically modified with tissue kallikrein are resistant to hypoxia- and oxidative stress-induced apoptosis, and offer enhanced protective actions in animal models of heart and kidney injury and hindlimb ischemia. In addition, activation of the plasma kallikrein-kinin system promotes EPC recruitment to the inflamed synovium of arthritic rats. Conversely, cleaved high molecular weight kininogen, a product of plasma kallikrein, reduces the viability and vasculogenic activity of EPCs. Therefore, kallikrein-kinin provides a new approach in enhancing the efficacy of stem cell therapy for human diseases.
Collapse
|
27
|
Maneva-Radicheva L, Amatya C, Parker C, Ellefson J, Radichev I, Raghavan A, Charles ML, Williams MS, Robbins MS, Savinov AY. Autoimmune diabetes is suppressed by treatment with recombinant human tissue Kallikrein-1. PLoS One 2014; 9:e107213. [PMID: 25259810 PMCID: PMC4178025 DOI: 10.1371/journal.pone.0107213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 08/12/2014] [Indexed: 12/31/2022] Open
Abstract
The kallikrein-kinin system (KKS) comprises a cascade of proteolytic enzymes and biogenic peptides that regulate several physiological processes. Over-expression of tissue kallikrein-1 and modulation of the KKS shows beneficial effects on insulin sensitivity and other parameters relevant to type 2 diabetes mellitus. However, much less is known about the role of kallikreins, in particular tissue kallikrein-1, in type 1 diabetes mellitus (T1D). We report that chronic administration of recombinant human tissue kallikrein-1 protein (DM199) to non-obese diabetic mice delayed the onset of T1D, attenuated the degree of insulitis, and improved pancreatic beta cell mass in a dose- and treatment frequency-dependent manner. Suppression of the autoimmune reaction against pancreatic beta cells was evidenced by a reduction in the relative numbers of infiltrating cytotoxic lymphocytes and an increase in the relative numbers of regulatory T cells in the pancreas and pancreatic lymph nodes. These effects may be due in part to a DM199 treatment-dependent increase in active TGF-beta1. Treatment with DM199 also resulted in elevated C-peptide levels, elevated glucagon like peptide-1 levels and a reduction in dipeptidyl peptidase-4 activity. Overall, the data suggest that DM199 may have a beneficial effect on T1D by attenuating the autoimmune reaction and improving beta cell health.
Collapse
Affiliation(s)
- Lilia Maneva-Radicheva
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Christina Amatya
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Camille Parker
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Jacob Ellefson
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Ilian Radichev
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Arvind Raghavan
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
- * E-mail: (AR); (AYS)
| | | | - Mark S. Williams
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
| | - Mark S. Robbins
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
| | - Alexei Y. Savinov
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
- * E-mail: (AR); (AYS)
| |
Collapse
|
28
|
Chao J, Bledsoe G, Chao L. Tissue kallikrein-kinin therapy in hypertension and organ damage. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:37-57. [PMID: 25130039 DOI: 10.1007/978-3-319-06683-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Tissue kallikrein is a serine proteinase that cleaves low molecular weight kininogen to produce kinin peptides, which in turn activate kinin receptors to trigger multiple biological functions. In addition to its kinin-releasing activity, tissue kallikrein directly interacts with the kinin B2 receptor, protease-activated receptor-1, and gamma-epithelial Na channel. The tissue kallikrein-kinin system (KKS) elicits a wide spectrum of biological activities, including reducing hypertension, cardiac and renal damage, restenosis, ischemic stroke, and skin wound injury. Both loss-of-function and gain-of-function studies have shown that the KKS plays an important endogenous role in the protection against health pathologies. Tissue kallikrein/kinin treatment attenuates cardiovascular, renal, and brain injury by inhibiting oxidative stress, apoptosis, inflammation, hypertrophy, and fibrosis and promoting angiogenesis and neurogenesis. Approaches that augment tissue kallikrein-kinin activity might provide an effective strategy for the treatment of hypertension and associated organ damage.
Collapse
|
29
|
Rhaleb NE, Yang XP, Carretero OA. The kallikrein-kinin system as a regulator of cardiovascular and renal function. Compr Physiol 2013; 1:971-93. [PMID: 23737209 DOI: 10.1002/cphy.c100053] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autocrine, paracrine, endocrine, and neuroendocrine hormonal systems help regulate cardio-vascular and renal function. Any change in the balance among these systems may result in hypertension and target organ damage, whether the cause is genetic, environmental or a combination of the two. Endocrine and neuroendocrine vasopressor hormones such as the renin-angiotensin system (RAS), aldosterone, and catecholamines are important for regulation of blood pressure and pathogenesis of hypertension and target organ damage. While the role of vasodepressor autacoids such as kinins is not as well defined, there is increasing evidence that they are not only critical to blood pressure and renal function but may also oppose remodeling of the cardiovascular system. Here we will primarily be concerned with kinins, which are oligopeptides containing the aminoacid sequence of bradykinin. They are generated from precursors known as kininogens by enzymes such as tissue (glandular) and plasma kallikrein. Some of the effects of kinins are mediated via autacoids such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF), and/or tissue plasminogen activator (tPA). Kinins help protect against cardiac ischemia and play an important part in preconditioning as well as the cardiovascular and renal protective effects of angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARB). But the role of kinins in the pathogenesis of hypertension remains controversial. A study of Utah families revealed that a dominant kallikrein gene expressed as high urinary kallikrein excretion was associated with a decreased risk of essential hypertension. Moreover, researchers have identified a restriction fragment length polymorphism (RFLP) that distinguishes the kallikrein gene family found in one strain of spontaneously hypertensive rats (SHR) from a homologous gene in normotensive Brown Norway rats, and in recombinant inbred substrains derived from these SHR and Brown Norway rats this RFLP cosegregated with an increase in blood pressure. However, humans, rats and mice with a deficiency in one or more components of the kallikrein-kinin-system (KKS) or chronic KKS blockade do not have hypertension. In the kidney, kinins are essential for proper regulation of papillary blood flow and water and sodium excretion. B2-KO mice appear to be more sensitive to the hypertensinogenic effect of salt. Kinins are involved in the acute antihypertensive effects of ACE inhibitors but not their chronic effects (save for mineralocorticoid-salt-induced hypertension). Kinins appear to play a role in the pathogenesis of inflammatory diseases such as arthritis and skin inflammation; they act on innate immunity as mediators of inflammation by promoting maturation of dendritic cells, which activate the body's adaptive immune system and thereby stimulate mechanisms that promote inflammation. On the other hand, kinins acting via NO contribute to the vascular protective effect of ACE inhibitors during neointima formation. In myocardial infarction produced by ischemia/reperfusion, kinins help reduce infarct size following preconditioning or treatment with ACE inhibitors. In heart failure secondary to infarction, the therapeutic effects of ACE inhibitors are partially mediated by kinins via release of NO, while drugs that activate the angiotensin type 2 receptor act in part via kinins and NO. Thus kinins play an important role in regulation of cardiovascular and renal function as well as many of the beneficial effects of ACE inhibitors and ARBs on target organ damage in hypertension.
Collapse
Affiliation(s)
- Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.
| | | | | |
Collapse
|
30
|
Yao Y, Sheng Z, Li Y, Fu C, Ma G, Liu N, Chao J, Chao L. Tissue kallikrein-modified human endothelial progenitor cell implantation improves cardiac function via enhanced activation of akt and increased angiogenesis. J Transl Med 2013; 93:577-91. [PMID: 23508045 PMCID: PMC4051305 DOI: 10.1038/labinvest.2013.48] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endothelial progenitor cells (EPCs) have been shown to enhance angiogenesis not only by incorporating into the vasculature but also by secreting cytokines, thereby serving as an ideal vehicle for gene transfer. As tissue kallikrein (TK) has pleiotropic effects in inhibiting apoptosis and oxidative stress, and promoting angiogenesis, we evaluated the salutary potential of kallikrein-modified human EPCs (hEPCs; Ad.hTK-hEPCs) after acute myocardial infarction (MI). We genetically modified hEPCs with a TK gene and evaluated cell survival, engraftment, revascularization, and functional improvement in a nude mouse left anterior descending ligation model. hEPCs were manipulated to overexpress the TK gene. In vitro, the antiapoptotic and paracrine effects were assessed under oxidative stress. TK protects hEPCs from oxidative stress-induced apoptosis via inhibition of activation of caspase-3 and -9, induction of Akt phosphorylation, and secretion of vascular endothelial growth factor. In vivo, the Ad.hTK-hEPCs were transplanted after MI via intracardiac injection. The surviving cells were tracked after transplantation using near-infrared optical imaging. Left ventricular (LV) function was evaluated by transthoracic echocardiography. Capillary density was quantified using immunohistochemical staining. Engrafted Ad.hTK-hEPCs exhibited advanced protection against ischemia by increasing LV ejection fraction. Compared with Ad.Null-hEPCs, transplantation with Ad.hTK-hEPCs significantly decreased cardiomyocyte apoptosis in association with increased retention of transplanted EPCs in the myocardium. Capillary density and arteriolar density in the infarct border zone was significantly higher in Ad.hTK-hEPC-transplanted mice than in Ad.Null-hEPC-treated mice. Transplanted hEPCs were clearly incorporated into CD31(+) capillaries. These results indicate that implantation of kallikrein-modified EPCs in the heart provides advanced benefits in protection against ischemia-induced MI by enhanced angiogenesis and reducing apoptosis.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Inhibition of anti-apoptotic signals by Wortmannin induces apoptosis in the remote myocardium after LAD ligation: evidence for a protein kinase C-δ-dependent pathway. Mol Cell Biochem 2012; 372:275-83. [PMID: 23010893 DOI: 10.1007/s11010-012-1469-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 09/14/2012] [Indexed: 01/04/2023]
Abstract
It has been shown that, in the remote myocardium after infarction (MI), protein kinase C (PKC) inhibition reduces apoptosis both by blocking proapoptotic pathways and by activating antiapoptotic signals including the Akt pathway. However, it was open if vice versa, blockade of antiapoptotic pathways may influence proapoptotic signals. To clarify this, the present study tested the effects of the PI3-kinase blocker Wortmannin on proapoptotic signals and on apoptosis execution in the remote myocardium after infarction. Rats were subjected to MI by LAD ligation in situ. Some were pre-treated with Wortmannin alone or in combination with the PKC inhibitor Chelerythrine. After 24 h, pro- and anti-apoptotic signals (caspase-3, PKC isoforms, p38-MAPK, p42/44-MAPK, Akt, Bad), and marker of apoptosis execution (TUNEL) were quantified in the myocardium remote from the infarction. Wortmannin treatment increased apoptosis in the remote myocardium both at baseline and after MI, together with an activation of the PKC-δ/p38-MAPK-pathway. PKC-ε and p42/44-MAPK were unaffected. Combined treatment with Wortmannin and Chelerythrine fully reversed the pro-apoptotic effects of Wortmannin both at baseline and after MI. The PKC-δ-p38-MAPK-pathway as a strong signal for apoptosis in the non-infarcted myocardium can be influenced by targeting the anti-apoptotic PI3-kinase pathway. This gives evidence of a bi-directional crosstalk of pro- and anti-apoptotic signals after infarction.
Collapse
|
32
|
Zepeda R, Castillo P, Sáez D, Llanos MN, Ronco AM. Cardiac tissue injury resistance during myocardial infarction at adulthood by developmental exposure to cadmium. Cardiovasc Toxicol 2012; 12:64-72. [PMID: 21858600 DOI: 10.1007/s12012-011-9139-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
It has been suggested that prenatal exposure to cadmium may alter the cardiovascular function during adulthood. Using the left coronary artery ligation model of acute myocardial infarction, we studied the cardiac function of female adult offspring rats exposed to cadmium (30 ppm) during gestation. The cardiac ischemic zone in the control and cadmium-exposed groups was measured 72 h post-ligation using the TPT staining technique. Offspring from cadmium-treated dams showed a significantly smaller infarcted area compared with the control group (7.1 ± 1.5 vs. 19.6 ± 2.8%, P ≤ 0.05). We also performed echocardiographic and biochemical studies, which positively correlated with the differences observed previously. To evaluate whether the effects were associated to pre-infarct tissue damage and/or angiogenic molecules, we performed histological studies and measured the expression of vascular endothelial growth factor (VEGF), and platelet endothelial cellular adhesion molecule-1 (PECAM-1). Results revealed a higher heart vascularization in the exposed offspring that was associated with an increase in PECAM and a decrease in VEGF expression. We conclude that prenatal exposure to cadmium induces fetal adaptive responses involving changes in the expression of some cardiac angiogenic molecules resulting in long-term resistance to infarction.
Collapse
Affiliation(s)
- Ramiro Zepeda
- Laboratory of Nutrition and Metabolic Regulation, Institute of Nutrition and Food Technology (INTA), University of Chile, El Líbano, Casilla, Macul, Santiago, Chile
| | | | | | | | | |
Collapse
|
33
|
Shimizu Y, Mullins N, Blanchard Z, Elshamy WM. BRCA1/p220 loss triggers BRCA1-IRIS overexpression via mRNA stabilization in breast cancer cells. Oncotarget 2012; 3:299-313. [PMID: 22431556 PMCID: PMC3359886 DOI: 10.18632/oncotarget.462] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BRCA1/p220-assocaited and triple negative/basal-like (TN/BL) tumors are aggressive and incurable breast cancer diseases that share among other features the no/low BRCA1/p220 expression. Here we show that BRCA1/p220 silencing in normal human mammary epithelial (HME) cells reduces expression of two RNA-destabilizing proteins, namely AUF1 and pCBP2, both proteins bind and destabilize BRCA1-IRIS mRNA. BRCA1-IRIS overexpression in HME cells triggers expression of several TN/BL markers, e.g., cytokeratins 5 and 17, p-cadherin, EGFR and cyclin E as well as expression and activation of the pro-survival proteins; AKT and survivin. BRCA1-IRIS silencing in the TN/BL cell line, SUM149 or restoration of BRCA1/p220 expression in the mutant cell line, HCC1937 reduced expression of TN/BL markers, AKT and survivin and induced cell death. Collectively, we propose that BRCA1/p220 loss of expression or function triggers BRCA1-IRIS overexpression through a post-transcriptional mechanism, which in turn promotes formation of aggressive and invasive breast tumors by inducing expression of TN/BL and survival proteins.
Collapse
Affiliation(s)
- Yoshiko Shimizu
- Cancer Institute and Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | |
Collapse
|
34
|
Yao Y, Sheng Z, Li Y, Yan F, Fu C, Li Y, Ma G, Liu N, Chao J, Chao L. Tissue kallikrein promotes cardiac neovascularization by enhancing endothelial progenitor cell functional capacity. Hum Gene Ther 2012; 23:859-70. [PMID: 22435954 DOI: 10.1089/hum.2011.123] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue kallikrein (TK) has been demonstrated to improve neovasculogenesis after myocardial infarction (MI). In the present study, we examined the role and underlying mechanisms of TK in peripheral endothelial progenitor cell (EPC) function. Peripheral blood-derived mononuclear cells containing EPCs were isolated from rat. The in vitro effects of TK on EPC differentiation, apoptosis, migration, and vascular tube formation capacity were studied in the presence or absence of TK, kinin B(2) receptor antagonist (icatibant), and phosphatidylinositol-3 kinase inhibitor (LY294002). Apoptosis was evaluated by flow-cytometry analysis using Annexin V-FITC/PI staining, as well as western-blot analysis of Akt phosphorylation and cleaved caspase-3. Using an MI mouse model, we then examined the in vivo effects of human TK gene adenoviral vector (Ad.hTK) administration on the number of CD34(+)Flk-1(+) progenitors in the peripheral circulation, heart tissue, extent of vasculogenesis, and heart function. Administration of TK significantly increased the number of Dil-LDL/UEA-lectin double-positive early EPCs, as well as their migration and tube formation properties in vitro. Transduction of TK in cultured EPCs attenuated apoptosis induced by hypoxia and led to an increase in Akt phosphorylation and a decrease in cleaved caspase-3 levels. The beneficial effects of TK were blocked by pretreatment with icatibant and LY294002. The expression of recombinant human TK in the ischemic mouse heart significantly improved cardiac contractility and reduced infarct size 7 days after gene delivery. Compared with the Ad.Null group, Ad.hTK reduced mortality and preserved left ventricular function by increasing the number of CD34(+)Flk-1(+) EPCs and promoting the growth of capillaries and arterioles in the peri-infarct myocardium. These data provide direct evidence that TK promotes vessel growth by increasing the number of EPCs and enhancing their functional properties through the kinin B(2) receptor-Akt signaling pathway.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kinin-B2 receptor mediated neuroprotection after NMDA excitotoxicity is reversed in the presence of kinin-B1 receptor agonists. PLoS One 2012; 7:e30755. [PMID: 22348022 PMCID: PMC3277507 DOI: 10.1371/journal.pone.0030755] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/21/2011] [Indexed: 01/21/2023] Open
Abstract
Background Kinins, with bradykinin and des-Arg9-bradykinin being the most important ones, are pro-inflammatory peptides released after tissue injury including stroke. Although the actions of bradykinin are in general well characterized; it remains controversial whether the effects of bradykinin are beneficial or not. Kinin-B2 receptor activation participates in various physiological processes including hypotension, neurotransmission and neuronal differentiation. The bradykinin metabolite des-Arg9-bradykinin as well as Lys-des-Arg9-bradykinin activates the kinin-B1 receptor known to be expressed under inflammatory conditions. We have investigated the effects of kinin-B1 and B2 receptor activation on N-methyl-D-aspartate (NMDA)-induced excitotoxicity measured as decreased capacity to produce synaptically evoked population spikes in the CA1 area of rat hippocampal slices. Principal Findings Bradykinin at 10 nM and 1 µM concentrations triggered a neuroprotective cascade via kinin-B2 receptor activation which conferred protection against NMDA-induced excitotoxicity. Recovery of population spikes induced by 10 nM bradykinin was completely abolished when the peptide was co-applied with the selective kinin-B2 receptor antagonist HOE-140. Kinin-B2 receptor activation promoted survival of hippocampal neurons via phosphatidylinositol 3-kinase, while MEK/MAPK signaling was not involved in protection against NMDA-evoked excitotoxic effects. However, 100 nM Lys-des-Arg9-bradykinin, a potent kinin-B1 receptor agonist, reversed bradykinin-induced population spike recovery. The inhibition of population spikes recovery was reversed by PD98059, showing that MEK/MAPK was involved in the induction of apoptosis mediated by the B1 receptor. Conclusions Bradykinin exerted protection against NMDA-induced excitotoxicity which is reversed in the presence of a kinin-B1 receptor agonist. As bradykinin is converted to the kinin-B1 receptor metabolite des-Arg9-bradykinin by carboxypeptidases, present in different areas including in brain, our results provide a mechanism for the neuroprotective effect in vitro despite of the deleterious effect observed in vivo.
Collapse
|
36
|
Rust NM, Papa MP, Scovino AM, da Silva MMC, Calzavara-Silva CE, Marques ETDA, Peçanha LMT, Scharfstein J, Arruda LB. Bradykinin enhances Sindbis virus infection in human brain microvascular endothelial cells. Virology 2012; 422:81-91. [DOI: 10.1016/j.virol.2011.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/03/2011] [Accepted: 10/04/2011] [Indexed: 12/16/2022]
|
37
|
Regional imbalanced activation of the calcineurin/BAD apoptotic pathway and the PI3K/Akt survival pathway after myocardial infarction. Int J Cardiol 2011; 166:158-65. [PMID: 22088220 DOI: 10.1016/j.ijcard.2011.10.107] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 09/23/2011] [Accepted: 10/18/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND The underlying molecular mechanisms of the remodeling after myocardial infarction (MI) remain unclear. The purpose of this study was to investigate the role of a survival pathway (PI3K/Akt) and an apoptosis pathway (calcineurin/BAD) in the remodeling after MI in a large animal model. METHODS Ten Dorset hybrid sheep underwent 25% MI in the left ventricle (LV, n=10). Five sheep were used as sham control. The regional strain was calculated from sonomicrometry. Apoptosis and the activation of the PI3K/Akt and calcineurin/BAD pathways were evaluated in the non-ischemic adjacent zone and the remote zone relative to infarct by immunoblotting, immunoprecipitation, and immunofluorescence staining. RESULTS Dilation and dysfunction of LV were present at 12 weeks after MI. The regional strain in the adjacent zone was significantly higher than in the remote zone at 12 weeks (36.6 ± 4.0% vs 9.5 ± 3.6%, p<0.05). Apoptosis was more severe in the adjacent zone than in the remote zone. The PI3K/Akt and calcineurin/BAD pathways were activated in the adjacent zone. Dephosphorylation and translocation of BAD were evident in the adjacent zone. Regional correlation between the strain and the expression of calcineurin/BAD indicated that the activation was strain-related (R(2)=0.46, 0.48, 0.39 for calcineurin, BAD, mitochondrial BAD, respectively, p<0.05). CONCLUSIONS The PI3K/Akt survival and calcineurin/BAD apoptotic pathways were concomitantly activated in the non-ischemic adjacent zone after MI. The calcineurin/BAD pathway is strain related and its imbalanced activation may be one of the causes of progressive remodeling after MI.
Collapse
|
38
|
Watanabe K, Thandavarayan RA, Harima M, Sari FR, Gurusamy N, Veeraveedu PT, Mito S, Arozal W, Sukumaran V, Laksmanan AP, Soetikno V, Kodama M, Aizawa Y. Role of differential signaling pathways and oxidative stress in diabetic cardiomyopathy. Curr Cardiol Rev 2011; 6:280-90. [PMID: 22043204 PMCID: PMC3083809 DOI: 10.2174/157340310793566145] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 09/03/2010] [Accepted: 09/15/2010] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus increases the risk of heart failure independently of underlying coronary artery disease, and many believe that diabetes leads to cardiomyopathy. The underlying pathogenesis is partially understood. Several factors may contribute to the development of cardiac dysfunction in the absence of coronary artery disease in diabetes mellitus. There is growing evidence that excess generation of highly reactive free radicals, largely due to hyperglycemia, causes oxidative stress, which further exacerbates the development and progression of diabetes and its complications. Hyperglycemia-induced oxidative stress is a major risk factor for the development of micro-vascular pathogenesis in the diabetic myocardium, which results in myocardial cell death, hypertrophy, fibrosis, abnormalities of calcium homeostasis and endothelial dysfunction. Diabetes-mediated biochemical changes show cross-interaction and complex interplay culminating in the activation of several intracellular signaling molecules. Diabetic cardiomyopathy is characterized by morphologic and structural changes in the myocardium and coronary vasculature mediated by the activation of various signaling pathways. This review focuses on the oxidative stress and signaling pathways in the pathogenesis of the cardiovascular complications of diabetes, which underlie the development and progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Kenichi Watanabe
- Department of Clinical Pharmacology, Niigata University of Pharmacy and Applied Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata City, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
14-3-3 proteins in neurodegeneration. Semin Cell Dev Biol 2011; 22:696-704. [PMID: 21920445 DOI: 10.1016/j.semcdb.2011.08.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/11/2011] [Indexed: 11/23/2022]
Abstract
Among the first reported functions of 14-3-3 proteins was the regulation of tyrosine hydroxylase (TH) activity suggesting a possible involvement of 14-3-3 proteins in Parkinson's disease. Since then the relevance of 14-3-3 proteins in the pathogenesis of chronic as well as acute neurodegenerative diseases, including Alzheimer's disease, polyglutamine diseases, amyotrophic lateral sclerosis and stroke has been recognized. The reported function of 14-3-3 proteins in this context are as diverse as the mechanism involved in neurodegeneration, reaching from basal cellular processes like apoptosis, over involvement in features common to many neurodegenerative diseases, like protein stabilization and aggregation, to very specific processes responsible for the selective vulnerability of cellular populations in single neurodegenerative diseases. Here, we review what is currently known of the function of 14-3-3 proteins in nervous tissue focussing on the properties of 14-3-3 proteins important in neurodegenerative disease pathogenesis.
Collapse
|
40
|
Sussman MA, Völkers M, Fischer K, Bailey B, Cottage CT, Din S, Gude N, Avitabile D, Alvarez R, Sundararaman B, Quijada P, Mason M, Konstandin MH, Malhowski A, Cheng Z, Khan M, McGregor M. Myocardial AKT: the omnipresent nexus. Physiol Rev 2011; 91:1023-70. [PMID: 21742795 PMCID: PMC3674828 DOI: 10.1152/physrev.00024.2010] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.
Collapse
Affiliation(s)
- Mark A Sussman
- Department of Biology, San Diego State University, SDSU Heart Institute, San Diego, California 92182, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Das J, Ghosh J, Manna P, Sil PC. Taurine suppresses doxorubicin-triggered oxidative stress and cardiac apoptosis in rat via up-regulation of PI3-K/Akt and inhibition of p53, p38-JNK. Biochem Pharmacol 2011; 81:891-909. [PMID: 21295553 DOI: 10.1016/j.bcp.2011.01.008] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 12/21/2010] [Accepted: 01/18/2011] [Indexed: 12/16/2022]
Abstract
The objective of the present study was to investigate the signaling mechanisms involved in the beneficial role of taurine against doxorubicin-induced cardiac oxidative stress. Male rats were administered doxorubicin. Hearts were collected 3 weeks after the last dose of doxorubicin and were analyzed. Doxorubicin administration retarded the growth of the body and the heart and caused injury in the cardiac tissue because of increased oxidative stress. Similar experiments with doxorubicin showed reduced cell viability, increased ROS generation, intracellular Ca(2+) and DNA fragmentation, disrupted mitochondrial membrane potential and apoptotic cell death in primary cultured neonatal rat cardiomyocytes. Signal transduction studies showed that doxorubicin increased p53, JNK, p38 and NFκB phosphorylation; decreased the levels of phospho ERK and Akt; disturbed the Bcl-2 family protein balance; activated caspase 12, caspase 9 and caspase 3; and induced cleavage of the PARP protein. However, taurine treatment or cardiomyocyte incubation with taurine suppressed all of the adverse effects of doxorubicin. Studies with several inhibitors, including PS-1145 (an IKK inhibitor), SP600125 (a JNK inhibitor), SB203580 (a p38 inhibitor) and LY294002 (a PI3-K/Akt inhibitor), demonstrated that the mechanism of taurine-induced cardio protection involves activation of specific survival signals and PI3-K/Akt as well as the inhibition of p53, JNK, p38 and NFκB. These novel findings suggest that taurine might have clinical implications for the prevention of doxorubicin-induced cardiac oxidative stress.
Collapse
Affiliation(s)
- Joydeep Das
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | | | | | | |
Collapse
|
42
|
Hung PH, Chen YW, Cheng KC, Chou HC, Lyu PC, Lu YC, Lee YR, Wu CT, Chan HL. Plasma proteomic analysis of the critical limb ischemia markers in diabetic patients with hemodialysis. MOLECULAR BIOSYSTEMS 2011; 7:1990-8. [DOI: 10.1039/c1mb05055a] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
43
|
Fujita T, Ishikawa Y. Apoptosis in Heart Failure - The Role of the .BETA.-Adrenergic Receptor-Mediated Signaling Pathway and p53-Mediated Signaling Pathway in the Apoptosis of Cardiomyocytes -. Circ J 2011; 75:1811-1818. [DOI: 10.1253/circj.cj-11-0025] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine
| |
Collapse
|
44
|
Al-Seeni M, El-Sawi N, Shaker S, Al-Amoudi A. Investigation of the Biochemical and Histological Changes Induced by Zearalenone Mycotoxin on Liver in Male Mice and the Protective Role of Crude Venom Extracted from Jellyfish <i>Cassiopea Andromeda</i>. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/fns.2011.24045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Huang C, Gu H, Zhang W, Herrmann JL, Wang M. Testosterone-down-regulated Akt pathway during cardiac ischemia/reperfusion: a mechanism involving BAD, Bcl-2 and FOXO3a. J Surg Res 2010; 164:e1-11. [PMID: 20850791 DOI: 10.1016/j.jss.2010.07.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 07/13/2010] [Accepted: 06/06/2010] [Indexed: 01/01/2023]
Abstract
BACKGROUND Lower levels of myocardial Akt activity in males are associated with a higher incidence of heart failure and worsened cardiac function after ischemia/reperfusion (I/R). While Akt activation by estrogen provides cardioprotection in females, no information exists regarding the effect of testosterone on the myocardial Akt pathway following I/R. We hypothesized that following I/R: (1) endogenous testosterone will decrease myocardial Akt activation in male hearts; (2) endogenous testosterone will mediate downstream signals of Akt, including Bad, Bcl-2, and FOXO3a; (3) administration of exogenous testosterone will recapitulate negative effects on the Akt pathway in castrated male hearts. METHODS AND RESULTS Rat hearts from age-matched adult males, females, castrated males, males with androgen receptor blocker-flutamide, castrated males with chronic 5α-dihydrotestosterone (DHT) implantation, or acute testosterone infusion (ATI) (n = 9/group) were subjected to I/R (Langendorff). Castration or flutamide treatment significantly up-regulated myocardial Akt activation, increased downstream apoptosis-regulatory molecules p-Bad, Bcl-2, p-FOXO3a, but reduced Fas-L, consistent with decreased myocardial injury in male hearts following I/R. ATI administration, but not chronic DHT, reversed these effects on Akt signaling associated with further exacerbated cardiac dysfunction in castrated males. Notably, lower levels of MnSOD were observed in male hearts, and castration or flutamide treatment restored myocardial MnSOD expression to the levels of females in male hearts after I/R. CONCLUSION Our study represents the initial evidence of testosterone-induced down-regulation of the Akt pathway in male hearts following I/R, thereby mediating cardiac injury through decreased p-Bad, reduced ratio of Bcl-2/Bax in the cytoplasm, and increased FOXO3a in the nucleus.
Collapse
Affiliation(s)
- Chunyan Huang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|
46
|
Chao J, Shen B, Gao L, Xia CF, Bledsoe G, Chao L. Tissue kallikrein in cardiovascular, cerebrovascular and renal diseases and skin wound healing. Biol Chem 2010; 391:345-55. [PMID: 20180644 DOI: 10.1515/bc.2010.042] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tissue kallikrein (KLK1) processes low-molecular weight kininogen to produce vasoactive kinins, which exert biological functions via kinin receptor signaling. Using various delivery approaches, we have demonstrated that tissue kallikrein through kinin B2 receptor signaling exhibits a wide spectrum of beneficial effects by reducing cardiac and renal injuries, restenosis and ischemic stroke, and by promoting angiogenesis and skin wound healing, independent of blood pressure reduction. Protection by tissue kallikrein in oxidative organ damage is attributed to the inhibition of apoptosis, inflammation, hypertrophy and fibrosis. Tissue kallikrein also enhances neovascularization in ischemic heart and limb. Moreover, tissue kallikrein/kinin infusion not only prevents but also reverses kidney injury, inflammation and fibrosis in salt-induced hypertensive rats. Furthermore, there is a wide time window for kallikrein administration in protection against ischemic brain infarction, as delayed kallikrein infusion for 24 h after cerebral ischemia in rats is effective in reducing neurological deficits, infarct size, apoptosis and inflammation. Importantly, in the clinical setting, human tissue kallikrein has been proven to be effective in the treatment of patients with acute brain infarction when injected within 48 h after stroke onset. Finally, kallikrein promotes skin wound healing and keratinocyte migration by direct activation of protease-activated receptor 1.
Collapse
Affiliation(s)
- Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, 29425, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Watanabe K, Thandavarayan RA, Gurusamy N, Zhang S, Muslin AJ, Suzuki K, Tachikawa H, Kodama M, Aizawa Y. Role of 14-3-3 protein and oxidative stress in diabetic cardiomyopathy. ACTA ACUST UNITED AC 2009; 96:277-87. [PMID: 19706371 DOI: 10.1556/aphysiol.96.2009.3.3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide. Diabetes mellitus is a well-known and important risk factor for cardiovascular diseases. The occurrence of diabetic cardiomyopathy is independent of hypertension, coronary artery disease, or any other known cardiac diseases. There is growing evidence that excess generation of highly reactive free radicals, largely due to hyperglycemia, causes oxidative stress, which further exacerbates the development and progression of diabetes and its complications. Diabetic cardiomyopathy is characterized by morphologic and structural changes in the myocardium and coronary vasculature mediated by the activation of various signaling pathways. Myocardial apoptosis, hypertrophy and fibrosis are the most frequently proposed mechanisms to explain cardiac changes in diabetic cardiomyopathy. Mammalian 14-3-3 proteins are dimeric phosphoserine-binding proteins that participate in signal transduction and regulate several aspects of cellular biochemistry. 14-3-3 protein regulates diabetic cardiomyopathy via multiple signaling pathways. This review focuses on emerging evidence suggesting that 14-3-3 protein plays a key role in the pathogenesis of the cardiovascular complications of diabetes, which underlie the development and progression of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Kenichi Watanabe
- Department of Clinical Pharmacology, Niigata University of Pharmacy and Applied Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata City, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Recombinant adeno-associated virus-mediated human kallikrein gene therapy protects against hypertensive target organ injuries through inhibiting cell apoptosis. Acta Pharmacol Sin 2009; 30:1253-61. [PMID: 19684610 DOI: 10.1038/aps.2009.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIM Overexpression of human tissue kallikrein (HK), mediated by recombinant adeno-associated virus (rAAV), decreased blood pressure in spontaneous hypertensive rats (SHRs) and reduced injury to the heart, aorta and kidney. In this study, we used both an in vivo animal model and in vitro cell culture system to investigate whether rAAV-mediated HK gene therapy protects against organ damage by inhibiting cell apoptosis. METHODS rAAV encoding HK (rAAV-HK) or LacZ (rAAV-lacZ) were delivered as a control to spontaneously hypertensive rats (SHRs) and cultured human embryonic kidney (HEK) 293 cells. RESULTS Treatment with rAAV-HK decreased cell apoptosis in the target organs of SHRs and also inhibited lipopolysaccharide (LPS)-induced HEK 293 apoptosis. The rAAV-HK delivery system also increased the levels of apoptosis-inhibiting proteins bcl-2 and bcl-x(L), and decreased the level of Bax and the activity of caspase 3, two promoters of apoptosis. In addition to its role in the inhibition of apoptosis, rAAV-HK also activated the cell survival and proliferation signaling pathways ERK1/2 and PI3K/AKT. CONCLUSION rAAV-mediated HK gene delivery has multiple therapeutic possibilities for treating hypertension, not only by decreasing blood pressure, but also by directly inhibiting end-organ damage.
Collapse
|
49
|
Juhaszova M, Zorov DB, Yaniv Y, Nuss HB, Wang S, Sollott SJ. Role of glycogen synthase kinase-3beta in cardioprotection. Circ Res 2009; 104:1240-52. [PMID: 19498210 DOI: 10.1161/circresaha.109.197996] [Citation(s) in RCA: 294] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Limitation of infarct size by ischemic/pharmacological pre- and postconditioning involves activation of a complex set of cell-signaling pathways. Multiple lines of evidence implicate the mitochondrial permeability transition pore (mPTP) as a key end effector of ischemic/pharmacological pre- and postconditioning. Increasing the ROS threshold for mPTP induction enhances the resistance of cardiomyocytes to oxidant stress and results in infarct size reduction. Here, we survey and synthesize the present knowledge about the role of glycogen synthase kinase (GSK)-3beta in cardioprotection, including pre- and postconditioning. Activation of a wide spectrum of cardioprotective signaling pathways is associated with phosphorylation and inhibition of a discrete pool of GSK-3beta relevant to mitochondrial signaling. Therefore, GSK-3beta has emerged as the integration point of many of these pathways and plays a central role in transferring protective signals downstream to target(s) that act at or in proximity to the mPTP. Bcl-2 family proteins and mPTP-regulatory elements, such as adenine nucleotide translocator and cyclophilin D (possibly voltage-dependent anion channel), may be the functional downstream target(s) of GSK-3beta. Gaining a better understanding of these interactions to control and prevent mPTP induction when appropriate will enable us to decrease the negative impact of the reperfusion-induced ROS burst on the fate of mitochondria and perhaps allow us to limit propagation of damage throughout and between cells and consequently, to better limit infarct size.
Collapse
Affiliation(s)
- Magdalena Juhaszova
- Laboratory of Cardiovascular Science, Gerontology Research Center, Box 13, National Institute on Aging, NIH, Baltimore, Maryland 21224-6825, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Glycogen synthase kinase-3beta (GSK-3beta) is a multifunctional Ser/Thr kinase that plays important roles in necrosis and apoptosis of cardiomyocytes. A major mechanism of cell necrosis is the opening of the mitochondrial permeability transition pore (mPTP), which consists of multiple protein subunits, including adenine nucleotide translocase (ANT). The threshold for mPTP opening is elevated by phosphorylation of GSK-3beta at Ser9, which reduces activity of this kinase. How inactivation of GSK-3beta suppresses mPTP opening has not been fully understood, but evidence to date suggests that preservation of hexokinase-II in the mPTP complex, inhibition of cyclophilin-D-ANT binding, inhibition of p53 and inhibition of ANT into the mitochondria are contributory. GSK-3beta phosphorylation is a step to which multiple protective signaling pathways converge, and thus GSK-3beta phosphorylation is crucial in cardioprotection of a variety of interventions against ischemia/reperfusion injury. Apoptosis of cardiomyocytes by pressure overload or ischemia/reperfusion is also suppressed by inactivation of GSK-3beta, in which reduced phosphorylation of p53, heat shock factor-1 and myeloid cell leukemia sequence-1 and inhibition of Bax translocation might be involved. Considering predominant roles of GSK-3beta in cardiomyocyte death, manipulation of this protein kinase is a promising strategy for myocardial protection in coronary artery disease and heart failure.
Collapse
Affiliation(s)
- Tetsuji Miura
- Division of Cardiology, Second Department of Internal Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo, Japan.
| | | |
Collapse
|