151
|
Normobaric oxygen therapy inhibits HIF-1α and VEGF expression in perihematoma and reduces neurological function defects. Neuroreport 2016; 27:329-36. [PMID: 26872098 DOI: 10.1097/wnr.0000000000000542] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
It is unknown whether normobaric oxygen (NBO) therapy exerts neuroprotective effects against human intracerebral hemorrhage (ICH). In this study, the potential of NBO therapy for salvaging brain damage following ICH was investigated in a rodent model with oxygen delivered at different concentrations. A total of 164 male Sprague-Dawley rats were induced with ICH using a collagenase injection and divided randomly into one ICH control group (no treatment, n=86) and three NBO treatment groups (35, 50, or 90% oxygen, n=26/group). Twenty-six rats were used as sham controls. The Neurological Severity Score (NSS) was evaluated. Contents of brain water, hypoxia-inducible factor 1α (HIF-1α), and vascular endothelial growth factor (VEGF) were measured in the perihematoma. A cellular apoptosis assay was performed. Compared with the sham control group, the ICH control group had higher NSS following ICH, which peaked at 24 h and began to decrease after 72 h. ICH rats also showed higher contents of brain water, HIF-1α, and VEGF (peaked at 72 h) in the ipsilateral perihematoma tissue than in the contralateral brain tissue. Compared with the ICH control group, all NBO groups showed improved NSSs, decreased contents of brain water, HIF-1α and VEGF, and fewer apoptotic cells in the perihematoma at 72 h after ICH, but statistical significance of these changes was achieved only with oxygen delivered at 90% (P<0.05, two-way analysis of variance). These results suggest that NBO therapy with oxygen delivered at 90% conferred best neuroprotection to ICH rats, potentially through amelioration of brain edema by suppressing HIF-1α and VEGF expression in the perihematoma.
Collapse
|
152
|
Sinha-Hikim I, Friedman TC, Falz M, Chalfant V, Hasan MK, Espinoza-Derout J, Lee DL, Sims C, Tran P, Mahata SK, Sinha-Hikim AP. Nicotine plus a high-fat diet triggers cardiomyocyte apoptosis. Cell Tissue Res 2016; 368:159-170. [PMID: 27917437 DOI: 10.1007/s00441-016-2536-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/11/2016] [Indexed: 12/29/2022]
Abstract
Cigarette smoking is an important risk factor for diabetes, cardiovascular disease and non-alcoholic fatty liver disease. The health risk associated with smoking can be aggravated by obesity. Smoking might also trigger cardiomyocyte (CM) apoptosis. Given that CM apoptosis has been implicated as a potential mechanism in the development of cardiomyopathy and heart failure, we characterize the key signaling pathways in nicotine plus high-fat diet (HFD)-induced CM apoptosis. Adult C57BL6 male mice were fed a normal diet (ND) or HFD and received twice-daily intraperitoneal (IP) injections of nicotine (0.75 mg/kg body weight [BW]) or saline for 16 weeks. An additional group of nicotine-treated mice on HFD received twice-daily IP injections of mecamylamine (1 mg/kg BW), a non-selective nicotinic acetylcholine receptor antagonist, for 16 weeks. Nicotine when combined with HFD led to a massive increase in CM apoptosis that was fully prevented by mecamylamine treatment. Induction of CM apoptosis was associated with increased oxidative stress and activation of caspase-2-mediated intrinsic pathway signaling coupled with inactivation of AMP-activated protein kinase (AMPK). Furthermore, nicotine treatment significantly (P < 0.05) attenuated the HFD-induced decrease in fibroblast growth factor 21 (FGF21) and silent information regulator 1 (SIRT1). We conclude that nicotine, when combined with HFD, triggers CM apoptosis through the generation of oxidative stress and inactivation of AMPK together with the activation of caspase-2-mediated intrinsic apoptotic signaling independently of FGF21 and SIRT1.
Collapse
Affiliation(s)
- Indrani Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA.,David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA.,David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark Falz
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Victor Chalfant
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Mohammad Kamrul Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Desean L Lee
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Carl Sims
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Peter Tran
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA
| | - Sushil K Mahata
- VA San Diego Health Care System and University of California, San Diego, Calif., USA
| | - Amiya P Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA 90059, USA. .,David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
153
|
Zeng M, Wei X, Wu Z, Li W, Zheng Y, Li B, Meng X, Fu X, Fei Y. Simulated ischemia/reperfusion-induced p65-Beclin 1-dependent autophagic cell death in human umbilical vein endothelial cells. Sci Rep 2016; 6:37448. [PMID: 27857190 PMCID: PMC5114588 DOI: 10.1038/srep37448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 10/31/2016] [Indexed: 12/15/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury detrimentally alters the prognosis of patients undergoing revascularization after acute myocardial infarction. Our previous study demonstrated that NF-κB-induced autophagy plays a detrimental role in cardiac I/R injury using a rabbit myocardial I/R model. In this study, we sought to explore the specific mechanism of this autophagy-mediated cell damage in an in vitro simulated ischemia/reperfusion (sI/R) model using human umbilical vein endothelial cells. Our current study demonstrates that simulated I/R induces autophagy in a p65-Beclin 1-dependent manner, which can be suppressed with the inhibition of NF-κB. Furthermore, rapamycin which promotes autophagy, exacerbates sI/R-induced cell death. While 3-methyladenine rescues cell damage. Our data thus suggest that I/R promotes NF-κB p65 activity mediated Beclin 1-mediated autophagic flux, thereby exacerbating myocardial injury.
Collapse
Affiliation(s)
- Min Zeng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xin Wei
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Zhiyong Wu
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Wei Li
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Yin Zheng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Bing Li
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xuqing Meng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xiuhong Fu
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Yi Fei
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| |
Collapse
|
154
|
Feng J, Wang W. Hypoxia pretreatment and EPO-modification enhance the protective effects of MSC on neuron-like PC12 cells in a similar way. Biochem Biophys Res Commun 2016; 482:232-238. [PMID: 27845038 DOI: 10.1016/j.bbrc.2016.11.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/08/2016] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) based cell transplantation therapy is proved to be an attractive strategy with great potential for improvement of hypoxia induced neural damage. In the present study, MSCs were co-culture with PC12 to investigate its protective effects against hypoxia pretreatment, and the Lactate dehydrogenase (LDH) release assay, MTT and Anexin V staining were performed to analysis the cellular damage or apoptotic. RT-PCR and Western blotting were further used to investigate the underlying mechanism. The results indicate that hypoxia treatment results in the decrease of PC12 cell viability, yet co-culture with MSC could protect the PC12 from hypoxia induced damage. Hypoxia pre-activated or EPO transduced MSC with up-regulated erythropoietin (EPO) expression could further enhance MSC's protective effect against hypoxia induced cell damage, which was associated with high level of anti-apoptotic p-Akt and ration Bcl-2/Bax, and decreased Caspase 3 in PC12. Taken together, these data suggests high levels of MSC-mediated cyto-protection is closely tied to high gene expression levels of EPO. The up-regulation of EPO for enhanced MSC-mediated cyto-protection may has great potential for the MSC cellular therapy of neural or neuronal injuries induced by hypoxia.
Collapse
Affiliation(s)
- Jinli Feng
- Department of Neurology, The 309th Hospital of PLA, NO17, Heishanhu Road, Haidian District, Beijing, China.
| | - Wei Wang
- Department of Neurology, The 309th Hospital of PLA, NO17, Heishanhu Road, Haidian District, Beijing, China.
| |
Collapse
|
155
|
Bibli SI, Andreadou I, Glynos C, Chatzianastasiou A, Toumpanakis D, Zakynthinos S, Vasilakopoulos T, Iliodromitis EK, Papapetropoulos A. Exposure to cigarette smoke abrogates the beneficial effect of ischemic postconditioning. Am J Physiol Heart Circ Physiol 2016; 311:H1321-H1332. [PMID: 27694220 DOI: 10.1152/ajpheart.00925.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/06/2016] [Indexed: 02/08/2023]
Abstract
Cigarette smoking is one of the risk factors for coronary artery disease. Although conditioning decreases infarct size in hearts from healthy animals, comorbidities may render it ineffective. We investigated the effects of cigarette smoke (CS) exposure on intracellular myocardial signaling, infarct size after ischemia-reperfusion, and the potential interference with ischemic conditioning. Exposure of mice to CS increased blood pressure, caused cardiac hypertrophy, and upregulated the nitric oxide synthatse (NOS)/soluble guanylate cyclase (sGC)/cGMP pathway. To test the effect of CS exposure on the endogenous cardioprotective mechanisms, mice were subjected to regional myocardial ischemia and reperfusion with no further intervention or application of preconditioning (PreC) or postconditioning (PostC). Exposure to CS did not increase the infarction compared with the room air (RA)-exposed group. PreC was beneficial for both CS and RA vs. nonconditioned animals. PostC was effective only in RA animals, while the infarct size-limiting effect was not preserved in the CS group. Differences in oxidative stress markers, Akt, and endothelial NOS phosphorylation and cGMP levels were observed between RA and CS groups subjected to PostC. In conclusion, exposure to CS does not per se increase infarct size. The beneficial effect of ischemic PreC is preserved in mice exposed to CS, as it does not affect the cardioprotective signaling; in contrast, PostC fails to protect CS-exposed mice due to impaired activation of the Akt/eNOS/cGMP axis that occurs in parallel to enhanced oxidative stress.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Glynos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Athanasia Chatzianastasiou
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Dimitris Toumpanakis
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Spyros Zakynthinos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Theodoros Vasilakopoulos
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| | - Efstathios K Iliodromitis
- Faculty of Medicine, 2nd Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece;
- "George P. Livanos and Marianthi Simou Laboratories," 1st Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece; and
| |
Collapse
|
156
|
Guarini G, Huqi A, Morrone D, Marzilli M. Pharmacological Agents Targeting Myocardial Metabolism for the Management of Chronic Stable Angina : an Update. Cardiovasc Drugs Ther 2016; 30:379-391. [DOI: 10.1007/s10557-016-6677-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
157
|
Chen ZD, Xu L, Tang KK, Gong FX, Liu JQ, Ni Y, Jiang LZ, Hong J, Han F, Li Q, Yang XH, Sun RH, Mo SJ. NF-κB-dependent transcriptional upregulation of cyclin D1 exerts cytoprotection against hypoxic injury upon EGFR activation. Exp Cell Res 2016; 347:52-59. [PMID: 27443256 DOI: 10.1016/j.yexcr.2016.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/27/2016] [Accepted: 07/12/2016] [Indexed: 12/28/2022]
Abstract
Apoptosis of neural cells is one of the main pathological features in hypoxic/ischemic brain injury. Nuclear factor-κB (NF-κB) might be a potential therapeutic target for hypoxic/ischemic brain injury since NF-κB has been found to be inactivated after hypoxia exposure, yet the underlying molecular mechanisms of NF-κB inactivation are largely unknown. Here we report that epidermal growth factor receptor (EGFR) activation prevents neuron-like PC12 cells apoptosis in response to hypoxia via restoring NF-κB-dependent transcriptional upregulation of cyclin D1. Functionally, EGFR activation by EGF stimulation mitigates hypoxia-induced PC12 cells apoptosis in both dose- and time-dependent manner. Of note, EGFR activation elevates IKKβ phosphorylation, increases IκBα ubiquitination, promotes P65 nuclear translocation and recruitment at cyclin D1 gene promoter as well as upregulates cyclin D1 expression. EGFR activation also abrogates the decrease of IKKβ phosphorylation, reduction of IκBα ubiquitination, blockade of P65 nuclear translocation and recruitment at cyclin D1 gene promoter as well as downregulation of cyclin D1 expression induced by hypoxia. Furthermore, NF-κB-dependent upregulation of cyclin D1 is instrumental for the EGFR-mediated cytoprotection against hypoxic apoptosis. In addition, the dephosphorylation of EGFR induced by either EGF siRNA transfection or anti-HB-EGF neutralization antibody treatment enhances hypoxic cytotoxicity, which are attenuated by EGF administration. Our results highlight the essential role of NF-κB-dependent transcriptional upregulation of cyclin D1 in EGFR-mediated cytoprotective effects under hypoxic preconditioning and support further investigation of EGF in clinical trials of patients with hypoxic/ischemic brain injury.
Collapse
Affiliation(s)
- Zhi-Dong Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Huzhou Normal College, Huzhou 313000, Zhejiang, China
| | - Liang Xu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Kan-Kai Tang
- Department of Critical Care Medicine, The First Affiliated Hospital of Huzhou Normal College, Huzhou 313000, Zhejiang, China
| | - Fang-Xiao Gong
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Jing-Quan Liu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Yin Ni
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Ling-Zhi Jiang
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Jun Hong
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Fang Han
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Qian Li
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Xiang-Hong Yang
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China
| | - Ren-Hua Sun
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China.
| | - Shi-Jing Mo
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang, China.
| |
Collapse
|
158
|
Li SY, Qi Y, Hu SH, Piao FY, Guan H, Wang ZM, Chen RL, Liu S. Mesenchymal stem cells-conditioned medium protects PC12 cells against 2,5-hexanedione-induced apoptosis via inhibiting mitochondria-dependent caspase 3 pathway. Toxicol Ind Health 2016; 33:107-118. [PMID: 26419259 DOI: 10.1177/0748233715598267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Studies suggested that the conditioned medium of mesenchymal stem cells (MSC-CM) inhibited the increased apoptosis in various cells. However, there are no reports underlying the protection of MSC-CM against 2,5-hexanedione (HD)-induced apoptosis in neural cells. In the present study, the viability was observed in PC12 cells that received HD alone or with MSC-CM by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Apoptosis was estimated by Hoechst 33342 staining and flow cytometry. Mitochondrial transmembrane potential was examined by rhodamine 123. Moreover, we investigated the expression of Bax and Bcl-2, cytochrome c translocation, and caspase 3 activity by real-time polymerase chain reaction, Western blot, and immunochemistry. Nerve growth factor (NGF) was examined in MSCs and MSC-CM. Our results showed that MSC-CM promoted cell survival and reduced apoptosis in HD-exposed PC12 cells. Moreover, MSC-CM significantly reversed disturbance of Bax and Bcl-2, ameliorated disruption of mitochondrial transmembrane potential, and reduced release of cytochrome c and activity of caspase 3 in HD-exposed PC12 cells. In the meantime, NGF was detected in MSCs and MSC-CM. These findings demonstrate that MSC-CM protects against HD-induced apoptosis in PC12 cells via inhibiting mitochondrial pathway. Our results indicate that NGF in MSC-CM may be involved in the protection of MSC-CM against HD-induced apoptosis. Our study clarifies the protection of MSC-CM on HD neurotoxicity and its underlying mechanism.
Collapse
Affiliation(s)
- Shuang-Yue Li
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| | - Yuan Qi
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| | - Shu-Hai Hu
- 2 College of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Feng-Yuan Piao
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| | - Huai Guan
- 3 Department of Obstetrics and Gynecology, Dalian, Liaoning, China
| | - Zhe-Min Wang
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| | - Ruo-Lin Chen
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| | - Shuang Liu
- 1 Department of Occupational and Environmental Health, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
159
|
Cubedo J, Vilahur G, Casaní L, Mendieta G, Gómez-Jabalera E, Juan-Babot O, Padró T, Badimon L. Targeting the molecular mechanisms of ischemic damage: Protective effects of alpha-crystallin-B. Int J Cardiol 2016; 215:406-16. [DOI: 10.1016/j.ijcard.2016.04.072] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022]
|
160
|
Regenerative repair of Pifithrin-α in cerebral ischemia via VEGF dependent manner. Sci Rep 2016; 6:26295. [PMID: 27212231 PMCID: PMC4876321 DOI: 10.1038/srep26295] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/29/2016] [Indexed: 01/01/2023] Open
Abstract
Promoting regenerative repair, including neurogenesis and angiogenesis, may provide a new therapeutic strategy for treatment of stroke. P53, a well-documented transcription factor, has been reported to be involved in cerebral ischemia and also serves as an important regulator of vascular endothelial growth factor (VEGF). However, the role of p53 in endogenous regenerative repair after brain ischemia is poorly understood. In this study, we investigated the effects of PFT-α, a specific p53 inhibitor on neurogenesis and angiogenesis improvement and associated signal pathways in rats impaired by cerebral artery occlusion (MCAo). PFT-α induced neuroprotection, reduced infarct volume and neurological functional impairment after ischemic stroke. More importantly, neurogenesis and angiogenesis were greatly enhanced by PFT-α, and accompanied by increased expression of VEGF. Moreover, we got consistent results in neural stem cells (NSCs) isolated from fetal rats. In contrast, application of the anti-VEGF neutralizing antibody (RB-222) partially reversed PFT-α-induced neuroprotection and rescued p53 expression. Noteworthily, inhibition of p53 after ischemic stroke in these rats improved their outcomes via promotion of regenerative repair. In conclusion, PFT-α could serve as a promising therapeutic strategy for ischemic stroke by promoting regenerative repair.
Collapse
|
161
|
Abstract
This study was designed to determine if Resolvin D1 (RvD1), a pro-resolution metabolite of the omega-3 polyunsaturated fatty acid docosahexaenoic acid, could decrease myocardial infarct size with delivered at the onset of ischemia. Male Sprague Dawley rats underwent 40 minutes of myocardial ischemia followed by reperfusion. These animals received 1 intraventricular injection of RvD1 (0.01, 0.1, or 0.3 μg RvD1) or vehicle (saline) before coronary occlusion. Infarct size and neutrophil accumulation were evaluated 24 hours after the onset of reperfusion. Caspase-3, caspase-8, protein kinase B (Akt) activities were evaluated 30 minutes after the reperfusion. Rats receiving 0.1 or 0.3 μg RvD1 showed a significant decrease of infarct size and caspase-3 and caspase-8 activities compared with the vehicle controls. Neutrophil accumulations were reduced in rats administered RvD1 compared with vehicle, independently of dose level. Akt activation was increased only in animals receiving 0.1 or 0.3 μg, whereas no change was observed in the 0.01 μg group. When they were treated with LY-294002, a phosphoinositide 3-kinase (PI3K)/Akt inhibitor, cardioprotection by RvD1 was abrogated. RvD1 treatment at the onset of ischemia decreases infarct size by a mechanism involving the PI3K/Akt pathway.
Collapse
|
162
|
Erythropoietin reduces storage lesions and decreases apoptosis indices in blood bank red blood cells. Rev Bras Hematol Hemoter 2016; 38:15-20. [PMID: 26969770 PMCID: PMC4786759 DOI: 10.1016/j.bjhh.2015.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 09/14/2015] [Accepted: 10/19/2015] [Indexed: 11/20/2022] Open
Abstract
Background Recent evidence shows a selective destruction of the youngest circulating red blood cells (neocytolysis) trigged by a drop in erythropoietin levels. Objective The aim of this study was to evaluate the effect of recombinant human erythropoietin beta on the red blood cell storage lesion and apoptosis indices under blood bank conditions. Methods Each one of ten red blood cell units preserved in additive solution 5 was divided in two volumes of 100 mL and assigned to one of two groups: erythropoietin (addition of 665 IU of recombinant human erythropoietin) and control (isotonic buffer solution was added). The pharmacokinetic parameters of erythropoietin were estimated and the following parameters were measured weekly, for six weeks: Immunoreactive erythropoietin, hemolysis, percentage of non-discocytes, adenosine triphosphate, glucose, lactate, lactate dehydrogenase, and annexin-V/esterase activity. The t-test or Wilcoxon's test was used for statistical analysis with significance being set for a p-value <0.05. Results Erythropoietin, when added to red blood cell units, has a half-life >6 weeks under blood bank conditions, with persistent supernatant concentrations of erythropoietin during the entire storage period. Adenosine triphosphate was higher in the Erythropoietin Group in Week 6 (4.19 ± 0.05 μmol/L vs. 3.53 ± 0.02 μmol/L; p-value = 0.009). The number of viable cells in the Erythropoietin Group was higher than in the Control Group (77% ± 3.8% vs. 71% ± 2.3%; p-value <0.05), while the number of apoptotic cells was lower (9.4% ± 0.3% vs. 22% ± 0.8%; p-value <0.05). Conclusions Under standard blood bank conditions, an important proportion of red blood cells satisfy the criteria of apoptosis. Recombinant human erythropoietin beta seems to improve storage lesion parameters and mitigate apoptosis.
Collapse
|
163
|
Jackson R, Brennan S, Fielding P, Sims MW, Challiss RAJ, Adlam D, Squire IB, Rainbow RD. Distinct and complementary roles for α and β isoenzymes of PKC in mediating vasoconstrictor responses to acutely elevated glucose. Br J Pharmacol 2016; 173:870-87. [PMID: 26660275 DOI: 10.1111/bph.13399] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/23/2015] [Accepted: 11/30/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE We investigated the hypothesis that elevated glucose increases contractile responses in vascular smooth muscle and that this enhanced constriction occurs due to the glucose-induced PKC-dependent inhibition of voltage-gated potassium channels. EXPERIMENTAL APPROACH Patch-clamp electrophysiology in rat isolated mesenteric arterial myocytes was performed to investigate the glucose-induced inhibition of voltage-gated potassium (Kv ) current. To determine the effects of glucose in whole vessel, wire myography was performed in rat mesenteric, porcine coronary and human internal mammary arteries. KEY RESULTS Glucose-induced inhibition of Kv was PKC-dependent and could be pharmacologically dissected using PKC isoenzyme-specific inhibitors to reveal a PKCβ-dependent component of Kv inhibition dominating between 0 and 10 mM glucose with an additional PKCα-dependent component becoming evident at concentrations greater than 10 mM. These findings were supported using wire myography in all artery types used, where contractile responses to vessel depolarization and vasoconstrictors were enhanced by increasing bathing glucose concentration, again with evidence for distinct and complementary PKCα/PKCβ-mediated components. CONCLUSIONS AND IMPLICATIONS Our results provide compelling evidence that glucose-induced PKCα/PKCβ-mediated inhibition of Kv current in vascular smooth muscle causes an enhanced constrictor response. Inhibition of Kv current causes a significant depolarization of vascular myocytes leading to marked vasoconstriction. The PKC dependence of this enhanced constrictor response may present a potential therapeutic target for improving microvascular perfusion following percutaneous coronary intervention after myocardial infarction in hyperglycaemic patients.
Collapse
Affiliation(s)
- Robert Jackson
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Sean Brennan
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Peter Fielding
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Mark W Sims
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - R A John Challiss
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - David Adlam
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Richard D Rainbow
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| |
Collapse
|
164
|
Liou K, Jepson N, Buckley N, Chen V, Thomas S, Russell EA, Ooi SY. Design and Rationale for the Endothelin-1 Receptor Antagonism in the Prevention of Microvascular Injury in Patients with non-ST Elevation Acute Coronary Syndrome Undergoing Percutaneous Coronary Intervention (ENDORA-PCI) Trial. Cardiovasc Drugs Ther 2016; 30:169-75. [DOI: 10.1007/s10557-016-6641-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
165
|
Xing Y, Tang B, Zhu C, Li W, Li Z, Zhao J, Gong WD, Wu ZQ, Zhu CC, Zhang YQ. N-myc downstream-regulated gene 4, up-regulated by tumor necrosis factor-α and nuclear factor kappa B, aggravates cardiac ischemia/reperfusion injury by inhibiting reperfusion injury salvage kinase pathway. Basic Res Cardiol 2016; 111:11. [PMID: 26780215 DOI: 10.1007/s00395-015-0519-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/08/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022]
Abstract
N-myc downstream-regulated gene 4 (NDRG4) is expressed weakly in heart and has been reported to modulate cardiac development and QT interval duration, but the role of NDRG4 in myocardial ischemia/reperfusion (I/R) injury remains unknown. In the present study, we analyzed the expression as well as potential function of cardiac NDRG4 and investigated how NDRG4 expression is regulated by inflammation. We found that NDRG4 was weakly expressed in cardiomyocytes and that its expression increased significantly both in I/R injured heart and in hypoxia-reoxygenation (H/R) injured neonatal rat ventricular myocytes (NRVMs). The increased NDRG4 expression aggravated myocardial I/R injury by inhibiting the activation of the reperfusion injury salvage kinase (RISK) pathway. Forced over-expression of NDRG4 inhibited RISK activation and exacerbated injury not only in I/R injured heart, but also in H/R treated NRVMs, whereas short hairpin RNA (shRNA)-mediated knock-down of NDRG4 enhanced RISK activation and attenuated injury. Upon injury, myocardial NDRG4 expression was induced by tumor necrosis factor-α (TNF-α) through nuclear factor kappa B (NF-κB), and we found that pre-treatment with inhibitors of either TNF-α or NF-κB blocked NDRG4 expression as well as I/R injury in vivo and H/R injury in vitro. Our study indicates that up-regulation of NDRG4 aggravates myocardial I/R injury by inhibiting activation of the RISK pathway, thereby identifying NDRG4 as a potential therapeutic target in I/R injury.
Collapse
Affiliation(s)
- Yuan Xing
- Department of Physiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Tang
- Department of International Medical, China-Japan Frindship Hospital, Beijing, 100029, China
| | - Chao Zhu
- Institute of Orthopaedics, Xi'jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Li
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhen Li
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Zhao
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei-dong Gong
- Department of Interventional Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zhi-qun Wu
- Department of Interventional Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Chu-chao Zhu
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yuan-qiang Zhang
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
166
|
Chu SF, Zhang Z, Zhang W, Zhang MJ, Gao Y, Han N, Zuo W, Huang HY, Chen NH. Upregulating the Expression of Survivin-HBXIP Complex Contributes to the Protective Role of IMM-H004 in Transient Global Cerebral Ischemia/Reperfusion. Mol Neurobiol 2016; 54:524-540. [DOI: 10.1007/s12035-015-9673-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/17/2015] [Indexed: 12/30/2022]
|
167
|
Li N, Si B, Ju JF, Zhu M, You F, Wang D, Ren J, Ning YS, Zhang FQ, Dong K, Huang J, Yu WQ, Wang TJ, Qiao B. Nicotine Induces Cardiomyocyte Hypertrophy Through TRPC3-Mediated Ca 2+/NFAT Signalling Pathway. Can J Cardiol 2015; 32:1260.e1-1260.e10. [PMID: 26952156 DOI: 10.1016/j.cjca.2015.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/11/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Nicotine is thought to be an important risk factor for the development of cardiovascular diseases. However, the effects of nicotine on cardiomyocyte hypertrophy are poorly understood. The present study was designed to explore the role of nicotine in cardiomyocyte hypertrophy and its underlying mechanism. METHODS We used primary cardiomyocytes isolated from Wistar rats to examine the effects of nicotine on intracellular Ca2+ mobilization and hypertrophy determined by immunofluorescence, quantitative polymerase chain reaction, and western blot analysis. A luciferase reporter assay was used to examine the activity of NFAT signalling. RESULTS We found that nicotine caused cardiomyocyte hypertrophy, which was accompanied by increased intracellular Ca2+. Nicotine-enhanced intracellular Ca2+ concentration ([Ca2+]i) was significantly abolished by store-operated Ca2+ entry (SOCE) and TRPC inhibitors. Knockdown of TRPC3 significantly decreased nicotine-induced SOCE and hypertrophy. Moreover, calcineurin-nuclear factor of activated T cells (NFAT) is involved in TRPC3-mediated Ca2+ signalling and cardiomyocyte hypertrophy. Notably, upregulation of TRPC3 by nicotine requires TRPC3-mediated Ca2+ influx and calcineurin-NFAT signalling activation. CONCLUSIONS Our findings demonstrate that the prohypertrophic effect of nicotine on cardiomyocytes is dependent on enhanced TRPC3 expression through a calcium-dependent regulatory loop, which could become a potential target for prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Na Li
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Biao Si
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Ji-Feng Ju
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Meng Zhu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng You
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Dong Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jie Ren
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Yan-Song Ning
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng-Quan Zhang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Kai Dong
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jing Huang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Wen-Qian Yu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Tong-Jian Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| | - Bin Qiao
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| |
Collapse
|
168
|
Sanhueza C, Wehinger S, Castillo Bennett J, Valenzuela M, Owen GI, Quest AFG. The twisted survivin connection to angiogenesis. Mol Cancer 2015; 14:198. [PMID: 26584646 PMCID: PMC4653922 DOI: 10.1186/s12943-015-0467-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Survivin, a member of the inhibitor of apoptosis family of proteins (IAPs) that controls cell division, apoptosis, metastasis and angiogenesis, is overexpressed in essentially all human cancers. As a consequence, the gene/protein is considered an attractive target for cancer treatment. Here, we discuss recent findings related to the regulation of survivin expression and its role in angiogenesis, particularly in the context of hypoxia. We propose a novel role for survivin in cancer, whereby expression of the protein in tumor cells promotes VEGF synthesis, secretion and angiogenesis. Mechanistically, we propose the existence of a positive feed-back loop involving PI3-kinase/Akt activation and enhanced β-Catenin-TCF/LEF-dependent VEGF expression followed by secretion. Finally, we elaborate on the possibility that this mechanism operating in cancer cells may contribute to enhanced tumor vascularization by vasculogenic mimicry together with conventional angiogenesis.
Collapse
Affiliation(s)
- C Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330024, Chile
| | - S Wehinger
- Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile
| | - J Castillo Bennett
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - M Valenzuela
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - G I Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Facultad de Ciencias Biológicas & Center UC Investigation in Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A F G Quest
- Cellular Communication Laboratory, Center for Molecular Studies of the Cell (CEMC), Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Av. Independencia 1027, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.
| |
Collapse
|
169
|
Ryan TE, Schmidt CA, Green TD, Brown DA, Neufer PD, McClung JM. Mitochondrial Regulation of the Muscle Microenvironment in Critical Limb Ischemia. Front Physiol 2015; 6:336. [PMID: 26635622 PMCID: PMC4649016 DOI: 10.3389/fphys.2015.00336] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe clinical presentation of peripheral arterial disease and manifests as chronic limb pain at rest and/or tissue necrosis. Current clinical interventions are largely ineffective and therapeutic angiogenesis based trials have shown little efficacy, highlighting the dire need for new ideas and novel therapeutic approaches. Despite a decade of research related to skeletal muscle as a determinant of morbidity and mortality outcomes in CLI, very little progress has been made toward an effective therapy aimed directly at the muscle myopathies of this disease. Within the muscle cell, mitochondria are well positioned to modulate the ischemic cellular response, as they are the principal sites of cellular energy production and the major regulators of cellular redox charge and cell death. In this mini review, we update the crucial importance of skeletal muscle to CLI pathology and examine the evolving influence of muscle and endothelial cell mitochondria in the complex ischemic microenvironment. Finally, we discuss the novelty of muscle mitochondria as a therapeutic target for ischemic pathology in the context of the complex co-morbidities often associated with CLI.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
170
|
Flenner F, Friedrich FW, Ungeheuer N, Christ T, Geertz B, Reischmann S, Wagner S, Stathopoulou K, Söhren KD, Weinberger F, Schwedhelm E, Cuello F, Maier LS, Eschenhagen T, Carrier L. Ranolazine antagonizes catecholamine-induced dysfunction in isolated cardiomyocytes, but lacks long-term therapeutic effects in vivo in a mouse model of hypertrophic cardiomyopathy. Cardiovasc Res 2015; 109:90-102. [PMID: 26531128 DOI: 10.1093/cvr/cvv247] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 10/27/2015] [Indexed: 12/19/2022] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is often accompanied by increased myofilament Ca(2+) sensitivity and diastolic dysfunction. Recent findings indicate increased late Na(+) current density in human HCM cardiomyocytes. Since ranolazine has the potential to decrease myofilament Ca(2+) sensitivity and late Na(+) current, we investigated its effects in an Mybpc3-targeted knock-in (KI) mouse model of HCM. METHODS AND RESULTS Unloaded sarcomere shortening and Ca(2+) transients were measured in KI and wild-type (WT) cardiomyocytes. Measurements were performed at baseline (1 Hz) and under increased workload (30 nM isoprenaline (ISO), 5 Hz) in the absence or presence of 10 µM ranolazine. KI myocytes showed shorter diastolic sarcomere length at baseline, stronger inotropic response to ISO, and drastic drop of diastolic sarcomere length under increased workload. Ranolazine attenuated ISO responses in WT and KI cells and prevented workload-induced diastolic failure in KI. Late Na(+) current density was diminished and insensitive to ranolazine in KI cardiomyocytes. Ca(2+) sensitivity of skinned KI trabeculae was slightly decreased by ranolazine. Phosphorylation analysis of cAMP-dependent protein kinase A-target proteins and ISO concentration-response measurements on muscle strips indicated antagonism at β-adrenoceptors with 10 µM ranolazine shifting the ISO response by 0.6 log units. Six-month treatment with ranolazine (plasma level >20 µM) demonstrated a β-blocking effect, but did not reverse cardiac hypertrophy or dysfunction in KI mice. CONCLUSION Ranolazine improved tolerance to high workload in mouse HCM cardiomyocytes, not by blocking late Na(+) current, but by antagonizing β-adrenergic stimulation and slightly desensitizing myofilaments to Ca(2+). This effect did not translate in therapeutic efficacy in vivo.
Collapse
Affiliation(s)
- Frederik Flenner
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Felix W Friedrich
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Nele Ungeheuer
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Silke Reischmann
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stefan Wagner
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany Department for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Konstantina Stathopoulou
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Klaus-Dieter Söhren
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany Department of Clinical Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Cuello
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lars S Maier
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany Department for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
171
|
Mo SJ, Hong J, Chen X, Han F, Ni Y, Zheng Y, Liu JQ, Xu L, Li Q, Yang XH, Sun RH, Yin XY. VEGF-mediated NF-κB activation protects PC12 cells from damage induced by hypoxia. Neurosci Lett 2015; 610:54-9. [PMID: 26518240 DOI: 10.1016/j.neulet.2015.10.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/17/2022]
Abstract
Neuronal apoptosis is a contributing cause of disability and death in cerebral ischemia. Nuclear factor-κB (NF-κB) may become a potential therapeutic target for hypoxic/ischemic neuron damage because NF-κB is inactivated after hypoxia exposure. Vascular endothelial growth factor (VEGF) has been found to improve neurological function recovery in cerebral ischemic injury although the exact molecular mechanisms that underlie the neuroprotective function of VEGF remain largely unknown. Here we defined the mechanism by which VEGF antagonized neuron-like PC12 cells apoptosis induced by hypoxia mimetic agent cobalt chloride (CoCl2) is through restoration of NF-κB activity. Depletion of VEGF with small interfering RNA (siRNA) in PC12 cells conferred CoCl2-induced cytotoxicity which was mitigated by VEGF administration. Treatment of PC12 cells with VEGF attenuated the CoCl2-induced cytotoxicity in both dose- and time-dependent manner. Mechanistically, VEGF increased IκBα phosphorylation and ubiquitination, promoted P65 nuclear translocation as well as upregulated XIAP and CCND1 expression. Meanwhile, VEGF administration reversed the dysregulation of IκBα phosphorylation and ubiquitination, P65 nuclear translocation as well as XIAP and CCND1 expression induced by CoCl2. Notably, the VEGF-dependent cytoprotection was abolished by pretreatment with BAY 11-7085, a specific inhibitor of NF-κB. Our data suggest that VEGF/NF-κB signalling pathway represents an adaptive mechanism that protects neural cells against hypoxic damage.
Collapse
Affiliation(s)
- Shi-Jing Mo
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Jun Hong
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Xu Chen
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Fang Han
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Yin Ni
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Yang Zheng
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Jing-Quan Liu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Liang Xu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Qian Li
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Xiang-Hong Yang
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China
| | - Ren-Hua Sun
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.
| | - Xiao-Yu Yin
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
172
|
Gu Y, Zhang Y, Bi Y, Liu J, Tan B, Gong M, Li T, Chen J. Mesenchymal stem cells suppress neuronal apoptosis and decrease IL-10 release via the TLR2/NFκB pathway in rats with hypoxic-ischemic brain damage. Mol Brain 2015; 8:65. [PMID: 26475712 PMCID: PMC4609057 DOI: 10.1186/s13041-015-0157-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/10/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic brain damage (HIBD) is a major cause of infant mortality and neurological disability in children. Many studies have demonstrated that mesenchymal stem cell (MSC) transplantation facilitates the restoration of the biological function of injured tissue following HIBD via immunomodulation. This study aimed to elucidate the mechanisms by which MSCs mediate immunomodulation via the key effectors Toll-like receptor 2 (TLR2) and interleukin-10 (IL-10). RESULTS We showed that TLR2 expression in the brain of HIBD rats was upregulated following HIBD and that MSC transplantation suppressed the expression of TLR2 and the release of IL-10, thereby alleviating the learning-memory deficits of HIBD rats. Following treatment with the specific TLR2 agonist Pam3CSK4 to activate TLR2, learning-memory function became further impaired, and the levels of nuclear factor kappa B (NFκB) and Bax expression and IL-10 release were significantly increased compared with those in HIBD rats that did not receive Pam3CSK4. In vitro, we found that MSC co-culture downregulated TLR2/NFκB signaling and repressed Bax expression and IL-10 secretion in oxygen and glucose deprivation (OGD)-injured adrenal pheochromocytoma (PC12) cells. Furthermore, NFκB and Bax expression and IL-10 release were enhanced following Pam3CSK4 treatment and were decreased following siTLR2 treatment in OGD-injured PC12 cells in the presence or absence of MSCs. CONCLUSIONS Our data indicate that TLR2 is involved in HIBD and that MSCs decrease apoptosis and improve learning-memory function in HIBD rats by suppressing the TLR2/NFκB signaling pathway via a feedback mechanism that reduces IL-10 release. These findings strongly suggest that MSC transplantation improves HIBD via the inhibition of the TLR2/NFκB pathway.
Collapse
Affiliation(s)
- Yan Gu
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Yun Zhang
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Yang Bi
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Jingjing Liu
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Bin Tan
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Min Gong
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Tingyu Li
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, 400014, China.
| | - Jie Chen
- Children Nutrition Research Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Chongqing Stem Cell Therapy Engineering Technical Centre, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
173
|
Grose JH, Langston K, Wang X, Squires S, Mustafi SB, Hayes W, Neubert J, Fischer SK, Fasano M, Saunders GM, Dai Q, Christians E, Lewandowski ED, Ping P, Benjamin IJ. Characterization of the Cardiac Overexpression of HSPB2 Reveals Mitochondrial and Myogenic Roles Supported by a Cardiac HspB2 Interactome. PLoS One 2015; 10:e0133994. [PMID: 26465331 PMCID: PMC4605610 DOI: 10.1371/journal.pone.0133994] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 07/03/2015] [Indexed: 01/26/2023] Open
Abstract
Small Heat Shock Proteins (sHSPs) are molecular chaperones that transiently interact with other proteins, thereby assisting with quality control of proper protein folding and/or degradation. They are also recruited to protect cells from a variety of stresses in response to extreme heat, heavy metals, and oxidative-reductive stress. Although ten human sHSPs have been identified, their likely diverse biological functions remain an enigma in health and disease, and much less is known about non-redundant roles in selective cells and tissues. Herein, we set out to comprehensively characterize the cardiac-restricted Heat Shock Protein B-2 (HspB2), which exhibited ischemic cardioprotection in transgenic overexpressing mice including reduced infarct size and maintenance of ATP levels. Global yeast two-hybrid analysis using HspB2 (bait) and a human cardiac library (prey) coupled with co-immunoprecipitation studies for mitochondrial target validation revealed the first HspB2 “cardiac interactome” to contain many myofibril and mitochondrial-binding partners consistent with the overexpression phenotype. This interactome has been submitted to the Biological General Repository for Interaction Datasets (BioGRID). A related sHSP chaperone HspB5 had only partially overlapping binding partners, supporting specificity of the interactome as well as non-redundant roles reported for these sHSPs. Evidence that the cardiac yeast two-hybrid HspB2 interactome targets resident mitochondrial client proteins is consistent with the role of HspB2 in maintaining ATP levels and suggests new chaperone-dependent functions for metabolic homeostasis. One of the HspB2 targets, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), has reported roles in HspB2 associated phenotypes including cardiac ATP production, mitochondrial function, and apoptosis, and was validated as a potential client protein of HspB2 through chaperone assays. From the clientele and phenotypes identified herein, it is tempting to speculate that small molecule activators of HspB2 might be deployed to mitigate mitochondrial related diseases such as cardiomyopathy and neurodegenerative disease.
Collapse
Affiliation(s)
- Julianne H. Grose
- Microbiology and Molecular Biology Department, Brigham Young University, Provo, UT, 84602, United States of America
- * E-mail: (JHG); (IJB)
| | - Kelsey Langston
- Microbiology and Molecular Biology Department, Brigham Young University, Provo, UT, 84602, United States of America
| | - Xiaohui Wang
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
| | - Shayne Squires
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
- Division of Cardiovascular Medicine, Dept. of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, United States of America
| | - Soumyajit Banerjee Mustafi
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
| | - Whitney Hayes
- Microbiology and Molecular Biology Department, Brigham Young University, Provo, UT, 84602, United States of America
| | - Jonathan Neubert
- Microbiology and Molecular Biology Department, Brigham Young University, Provo, UT, 84602, United States of America
| | - Susan K. Fischer
- Program in Integrative Cardiac Metabolism, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, United States of America
| | - Matthew Fasano
- Program in Integrative Cardiac Metabolism, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, United States of America
| | - Gina Moore Saunders
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
| | - Qiang Dai
- Division of Cardiovascular Medicine, Dept. of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, United States of America
| | - Elisabeth Christians
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
| | - E. Douglas Lewandowski
- Program in Integrative Cardiac Metabolism, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, United States of America
| | - Peipei Ping
- UCLA Departments of Physiology, Medicine, and Cardiology, Los Angeles, CA, 90095, United States of America
| | - Ivor J. Benjamin
- Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, United States of America
- Division of Cardiovascular Medicine, Dept. of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, United States of America
- * E-mail: (JHG); (IJB)
| |
Collapse
|
174
|
Abstract
Myocardial infarction is defined as sudden ischemic death of myocardial tissue. In the clinical context, myocardial infarction is usually due to thrombotic occlusion of a coronary vessel caused by rupture of a vulnerable plaque. Ischemia induces profound metabolic and ionic perturbations in the affected myocardium and causes rapid depression of systolic function. Prolonged myocardial ischemia activates a "wavefront" of cardiomyocyte death that extends from the subendocardium to the subepicardium. Mitochondrial alterations are prominently involved in apoptosis and necrosis of cardiomyocytes in the infarcted heart. The adult mammalian heart has negligible regenerative capacity, thus the infarcted myocardium heals through formation of a scar. Infarct healing is dependent on an inflammatory cascade, triggered by alarmins released by dying cells. Clearance of dead cells and matrix debris by infiltrating phagocytes activates anti-inflammatory pathways leading to suppression of cytokine and chemokine signaling. Activation of the renin-angiotensin-aldosterone system and release of transforming growth factor-β induce conversion of fibroblasts into myofibroblasts, promoting deposition of extracellular matrix proteins. Infarct healing is intertwined with geometric remodeling of the chamber, characterized by dilation, hypertrophy of viable segments, and progressive dysfunction. This review manuscript describes the molecular signals and cellular effectors implicated in injury, repair, and remodeling of the infarcted heart, the mechanistic basis of the most common complications associated with myocardial infarction, and the pathophysiologic effects of established treatment strategies. Moreover, we discuss the implications of pathophysiological insights in design and implementation of new promising therapeutic approaches for patients with myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
175
|
Ghosh A, Pechota LVTA, Upchurch GR, Eliason JL. Cross-talk between macrophages, smooth muscle cells, and endothelial cells in response to cigarette smoke: the effects on MMP2 and 9. Mol Cell Biochem 2015; 410:75-84. [DOI: 10.1007/s11010-015-2539-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/18/2015] [Indexed: 01/26/2023]
|
176
|
Kleinbongard P, Gedik N, Witting P, Freedman B, Klöcker N, Heusch G. Pleiotropic, heart rate-independent cardioprotection by ivabradine. Br J Pharmacol 2015; 172:4380-90. [PMID: 26076181 DOI: 10.1111/bph.13220] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/27/2015] [Accepted: 06/06/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE In pigs, ivabradine reduces infarct size even when given only at reperfusion and in the absence of heart rate reduction. The mechanism of this non-heart rate-related cardioprotection is unknown. Hence, in the present study we assessed the pleiotropic action of ivabradine in more detail. EXPERIMENTAL APPROACH Anaesthetized mice were pretreated with ivabradine (1.7 mg · kg(-1) i.v.) or placebo (control) before a cycle of coronary occlusion/reperfusion (30/120 min ± left atrial pacing). Infarct size was determined. Isolated ventricular cardiomyocytes were exposed to simulated ischaemia/reperfusion (60/5 min) in the absence and presence of ivabradine, viability was then quantified and intra- and extracellular reactive oxygen species (ROS) formation was detected. Mitochondria were isolated from mouse hearts and exposed to simulated ischaemia/reperfusion (6/3 min) in glutamate/malate- and ADP-containing buffer in the absence and presence of ivabradine respectively. Mitochondrial respiration, extramitochondrial ROS, mitochondrial ATP production and calcium retention capacity (CRC) were assessed. KEY RESULTS Ivabradine decreased infarct size even with atrial pacing. Cardiomyocyte viability after simulated ischaemia/reperfusion was better preserved with ivabradine, the accumulation of intra- and extracellular ROS decreased in parallel. Mitochondrial complex I respiration was not different without/with ivabradine, but ivabradine significantly inhibited the accumulation of extramitochondrial ROS, increased mitochondrial ATP production and increased CRC. CONCLUSION AND IMPLICATIONS Ivabradine reduces infarct size independently of a reduction in heart rate and improves ventricular cardiomyocyte viability, possibly by reducing mitochondrial ROS formation, increasing ATP production and CRC.
Collapse
Affiliation(s)
- P Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - N Gedik
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - P Witting
- Discipline of Pathology, The Charles Perkins Centre, The University of Sydney Medical School, Sydney, NSW, Australia
| | - B Freedman
- Concord Repatriation General Hospital, Vascular Biology Group, ANZAC Research Institute, Concord, NSW, Australia
| | - N Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - G Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| |
Collapse
|
177
|
Xu D, Wang A, Jiang F, Hu J, Zhang X. Effects of interleukin-37 on cardiac function after myocardial infarction in mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5247-5251. [PMID: 26191225 PMCID: PMC4503097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/26/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Interleukin-37 (IL-37) is a new discovered member of the interleukin family and plays anti-inflammatory effect in some inflammatory disease. A recent study found that IL-37 elevated significantly in peripheral blood of patients with acute myocardial infarction. We aimed to explore the effect IL-37 on cardiac function after mice myocardial infarction (MI) and its mechanism. METHODS Acute MI mouse model was established and divided into three groups: sham group, MI group and IL-37 treatment group. MPO expression was detected by immunohistochemistry; NF-κB signaling pathway was tested by Western blot; and cardiac function was measured by echocardiography. RESULTS Compared with MI mice, IL-37 treatment showed an obvious decrease of MPO expression, suppression of p-p65 expression, and improved cardiac function by decreasing left ventricular shortening fraction (LVFS). CONCLUSION IL-37 may improve MI mice cardiac function via inhibition of inflammatory NF-κB signaling pathway.
Collapse
Affiliation(s)
- Daoying Xu
- Department of Cardiology, Binzhou People’s HospitalBinzhou City 256610, Shandong Province, China
| | - Aiqin Wang
- Department of Critical Care Medicine, Binzhou People’s HospitalBinzhou City 256610, Shandong Province, China
| | - Fengqin Jiang
- Department of Cardiology, Binzhou People’s HospitalBinzhou City 256610, Shandong Province, China
| | - Junhong Hu
- Department of Cardiology, Binzhou People’s HospitalBinzhou City 256610, Shandong Province, China
| | - Xiuzhou Zhang
- Department of Cardiology, Binzhou People’s HospitalBinzhou City 256610, Shandong Province, China
| |
Collapse
|
178
|
Nemeth Z, Cziraki A, Szabados S, Horvath I, Koller A. Pericardial fluid of cardiac patients elicits arterial constriction: role of endothelin-1. Can J Physiol Pharmacol 2015; 93:779-85. [PMID: 26322806 DOI: 10.1139/cjpp-2015-0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recently, several vasoactive molecules have been found in pericardial fluid (PF). Thus, we hypothesized that in coronary artery disease due to ischemia or ischemia-reperfusion, the level of vasoconstrictors, mainly endothelin-1 (ET-1), increases in PF, which can increase the vasomotor tone of arteries. Experiments were performed using an isometric myograph. Vasomotor effects of PF from patients undergoing coronary artery bypass graft (PFCABG, n = 14) or valve replacement (PFVR, n = 7) surgery were examined in isolated rat carotid arteries (N = 14; n = 26). Vasomotor responses to KCl (40 or 60 mmol/L) were also tested. The selective endothelin A receptor antagonist BQ123 (10(-6) mol/L) was used to elucidate the role of ET-1. Both the first and the second additions of KCl elicited increases in the isometric force of the isolated arteries (KCl1, 6.1 ± 0.2 mN; KCl2, 6.5 ± 0.9 mN). PFCABG and PFVR elicited substantial increases in the isometric force of arteries (PFCABG, 3.1 ± 0.7 mN; PFVR, 3.0 ± 0.9 mN; p > 0.05). The presence of the selective endothelin A receptor blocker significantly reduced arterial contractions to PFCABG (before BQ123, 2.6 ± 0.5 mN vs. after BQ123, 0.8 ± 0.1 mN; p < 0.05). This study is the first to demonstrate that PFs of patients elicit substantial arterial constrictions, which is mediated primarily by ET-1. Interfering with the vasoconstrictor action of PF could be a potential therapeutic target to improve coronary blood flow in cardiac patients.
Collapse
Affiliation(s)
- Zoltan Nemeth
- a University of Pecs, Medical School, Department of Pathophysiology and Gerontology and Szentagothai Research Centre, Pecs, Hungary
| | - Attila Cziraki
- b University of Pecs, Medical School, Heart Institute, Pecs, Hungary
| | - Sandor Szabados
- b University of Pecs, Medical School, Heart Institute, Pecs, Hungary
| | - Ivan Horvath
- b University of Pecs, Medical School, Heart Institute, Pecs, Hungary
| | - Akos Koller
- a University of Pecs, Medical School, Department of Pathophysiology and Gerontology and Szentagothai Research Centre, Pecs, Hungary.,c University of Physical Education, Institute of Natural Sciences, Budapest, Hungary.,d Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
179
|
Rohrbach S, Troidl C, Hamm C, Schulz R. Ischemia and reperfusion related myocardial inflammation: A network of cells and mediators targeting the cardiomyocyte. IUBMB Life 2015; 67:110-9. [PMID: 25850820 DOI: 10.1002/iub.1352] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/14/2015] [Indexed: 12/15/2022]
Abstract
Occlusion of a coronary artery if maintained for longer period of time results in damage of the cardiac tissue. However, restoration of blood flow to previously ischemic tissue can itself induce further cardiac damage, a phenomenon known as myocardial reperfusion injury. Cardiac homoeostasis is supported by a network of direct and indirect interactions between cardiomyocytes and resident cell types such as fibroblasts, adipocytes, and endothelial cells or invading blood cells. This review will discuss the role of the cellular interplay in ischemia-reperfusion injury from a cardiomyocyte-centered view, although we are aware that other cellular interactions are equally important. We will try to work out currently unresolved questions and potential future directions in the field.
Collapse
Affiliation(s)
- Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | | | | | | |
Collapse
|
180
|
MCP-1-induced protein attenuates post-infarct cardiac remodeling and dysfunction through mitigating NF-κB activation and suppressing inflammation-associated microRNA expression. Basic Res Cardiol 2015; 110:26. [PMID: 25840774 DOI: 10.1007/s00395-015-0483-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/21/2022]
Abstract
MCP-1-induced protein (MCPIP, also known as ZC3H12A) has recently been uncovered to act as a negative regulator of inflammation. Expression of MCPIP was elevated in the ventricular myocardium of patients with ischemic heart failure. However, the role of MCPIP in the development of post-infarct cardiac inflammation and remodeling is unknown. The objective of the present study was to investigate whether MCPIP exerts an inhibitory effect on the cardiac inflammatory response and adverse remodeling after myocardial infarction (MI). Mice with cardiomyocyte-specific expression of MCPIP and their wild-type littermates (FVB/N) were subjected to permanent ligation of left coronary artery. The levels of MCPIP were significantly increased in the ischemic myocardium and sustained for 4 weeks after MI. Acute infarct size was comparable between groups. However, constitutive overexpression of MCPIP in the murine heart resulted in improved survival rate, decreased cardiac hypertrophy, less of fibrosis and scar formation, and better cardiac performance at 28 days after MI, along with a markedly reduced monocytic cell infiltration, less cytokine expression, decreased caspase-3/7 activities and apoptotic cell death compared to the wild-type hearts. Cardiomyocyte-specific expression of MCPIP also attenuated activation of cardiac NF-κB signaling and expression of inflammation-associated microRNAs (miR-126, -146a, -155, and -199a) when compared with the post-infarct wild-type hearts. In vitro, MCPIP expression suppressed hypoxia-induced NF-κB-luciferase activity in cardiomyocytes. In conclusion, MCPIP expression in the ischemic myocardium protects against adverse cardiac remodeling and dysfunction following MI by modulation of local myocardial inflammation, possibly through mitigating NF-κB signaling and suppressing inflammation-associated microRNA expression.
Collapse
|
181
|
Gupta T, Khera S, Kolte D, Aronow WS, Iwai S. Antiarrhythmic properties of ranolazine: A review of the current evidence. Int J Cardiol 2015; 187:66-74. [PMID: 25828315 DOI: 10.1016/j.ijcard.2015.03.324] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/20/2015] [Indexed: 12/19/2022]
Abstract
Ranolazine was developed as an antianginal agent and was approved by the Food and Drug Administration in 2006 for use in chronic stable angina pectoris. Experimental and clinical studies have shown that it also has antiarrhythmic properties based on the frequency-dependent blockade of peak sodium channel current (peak INa) and rapidly activating delayed rectifier potassium current (IKr) in the atria and blockade of late phase of the inward sodium current (late INa) in the ventricles. Recent clinical studies have revealed the efficacy of ranolazine in prevention of atrial fibrillation in patients with acute coronary syndromes, prevention as well as conversion of postoperative atrial fibrillation after cardiac surgery, conversion of recent-onset atrial fibrillation and maintenance of sinus rhythm in recurrent atrial fibrillation. Ranolazine has also been shown to reduce ventricular tachycardia and drug-refractory implantable cardioverter defibrillator shocks. The antiarrhythmic effect of ranolazine is preserved in the setting of chronic heart failure and clinical studies have demonstrated its safety in patients with heart failure. This review discusses the available preclinical and clinical data on the antiarrhythmic effects of this novel antianginal agent.
Collapse
Affiliation(s)
- Tanush Gupta
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Sahil Khera
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States.
| | - Dhaval Kolte
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Wilbert S Aronow
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| | - Sei Iwai
- Division of Cardiology, Department of Medicine, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
182
|
Li G, Shan C, Liu L, Zhou T, Zhou J, Hu X, Chen Y, Cui H, Gao N. Tanshinone IIA inhibits HIF-1α and VEGF expression in breast cancer cells via mTOR/p70S6K/RPS6/4E-BP1 signaling pathway. PLoS One 2015; 10:e0117440. [PMID: 25659153 PMCID: PMC4320086 DOI: 10.1371/journal.pone.0117440] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/23/2014] [Indexed: 01/08/2023] Open
Abstract
Hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) play important roles in angiogenesis and tumor growth. Tanshinone IIA (T2A) is a novel antiangiogenic agent with promising antitumor effects; however, the molecular mechanism underlying the antiangiogenic effects of T2A remains unclear. In the present study, we provided evidence showing that T2A inhibited angiogenesis and breast cancer growth by down-regulating VEGF expression. Specifically, T2A repressed HIF-1α expression at the translational level and inhibited the transcriptional activity of HIF-1α, which led to the down-regulation of VEGF expression. Suppression of HIF-1α synthesis by T2A correlated with strong dephosphorylation of mammalian target of rapamycin (mTOR) and its effectors ribosomal protein S6 kinase (p70S6K) and eukaryotic initiation factor 4E-binding protein-1 (4E-BP1), a pathway regulating HIF-1α expression at the translational level. In addition, we also found that T2A inhibited the angiogenesis and growth of human breast cancer xenografts in nude mice through suppression of HIF-1α and VEGF. Our study provides novel perspectives and potential targets for the treatment of human breast cancer.
Collapse
MESH Headings
- Abietanes/pharmacology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antineoplastic Agents, Phytogenic
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Cycle Proteins
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Mice
- Mice, Nude
- Neoplasm Proteins/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Phosphoproteins/metabolism
- Ribosomal Protein S6/metabolism
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Vascular Endothelial Growth Factor A/biosynthesis
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guobing Li
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Changyu Shan
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Lei Liu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ting Zhou
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jing Zhou
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaoye Hu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yibiao Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Ning Gao
- College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
183
|
Dental pulp stem cells derived conditioned medium promotes angiogenesis in hindlimb ischemia. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-014-9053-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
184
|
Abstract
Despite the growing number of patients affected, the understanding of diastolic dysfunction and heart failure with preserved ejection fraction (HFpEF) is still poor. Clinical trials, largely based on successful treatments for systolic heart failure, have been disappointing, suggesting that HFpEF has a different pathology to that of systolic dysfunction. In this review, general concepts, epidemiology, diagnosis, and treatment of diastolic dysfunction are summarized, with an emphasis on new experiments suggesting that oxidative stress plays a crucial role in the pathogenesis of at least some forms of the disease. This observation has lead to potential new diagnostics and therapeutics for diastolic dysfunction and heart failure caused by diastolic dysfunction.
Collapse
Affiliation(s)
- Euy-Myoung Jeong
- Cardiovascular Research Center and Cardiovascular Institute of Lifespan, The Warren Alpert Medical School, Brown University
| | | |
Collapse
|
185
|
Lahiani A, Zahavi E, Netzer N, Ofir R, Pinzur L, Raveh S, Arien-Zakay H, Yavin E, Lazarovici P. Human PLacental eXpanded (PLX) mesenchymal-like adherent stromal cells confer neuroprotection to nerve growth factor (NGF)-differentiated PC12 cells exposed to ischemia by secretion of IL-6 and VEGF. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:422-30. [DOI: 10.1016/j.bbamcr.2014.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/03/2014] [Accepted: 11/10/2014] [Indexed: 12/21/2022]
|
186
|
Zhu J, Liu Q, Jiang Y, Wu L, Xu G, Liu X. Enhanced angiogenesis promoted by human umbilical mesenchymal stem cell transplantation in stroked mouse is Notch1 signaling associated. Neuroscience 2015; 290:288-99. [PMID: 25637797 DOI: 10.1016/j.neuroscience.2015.01.038] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/18/2014] [Accepted: 01/08/2015] [Indexed: 12/16/2022]
Abstract
Cellular therapy has provided hope for restoring neurological function post stroke through promoting endogenous neurogenesis, angiogenesis and synaptogenesis. The current study was based on the observation that transplantation of human umbilical cord mesenchymal stem cells (hUCMSCs) promoted the neurological function improvement in stroked mice and meanwhile enhanced angiogenesis in the stroked hemisphere. Grafted hUCMSCs secreted human vascular endothelial growth factor A (VEGF-A). Notch1 signaling was activated after stroke and also in the grafted hUCMSCs. To address the potential mechanism that might mediate such pro-angiogenic effect, we established a hUCMSC-neuron co-culture system. Neurons were subjected to oxygen glucose deprivation (OGD) injury before co-culturing to mimic the in vivo cell transplantation. Consistent with the in vivo data, co-culture medium claimed from hUCMSC-OGD neuron co-culture system significantly promoted the capillary-like tube formation of brain-derived endothelial cells. Moreover, coincident with our in vivo data, Notch 1 signaling activation was detected in hUCMSCs after co-cultured with OGD neurons as demonstrated by the up-regulation of key Notch1 signaling components Notch1 and Notch1 intercellular domain (NICD). In addition, OGD-neuron co-culture also increased the VEGF-A production by hUCMSCs. To verify whether Notch1 activation was involved in the pro-angiogenic effect, γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) was added into the co-culture medium before co-culture. It turned out that DAPT significantly prevented the Notch1 activation in hUCMSCs after co-culture with OGD neurons. More importantly, the pro-angiogenic effect of hUCMSCs was remarkably abolished by DAPT addition as demonstrated by inhibited capillary-like tube formation and less VEGF-A production. Regarding how Notch1 signaling was linked with VEGF-A secretion, we provided some clue that Notch1 effector Hes1 mRNA expression was significantly up-regulated by OGD-neuron co-culturing and down-regulated after additional treatment of DAPT. In summary, our data provided evidence that the VEGF-A secretion from hUCMSCs after being triggered by OGD neurons is Notch1 signaling associated. This might be a possible mechanism that contributes to the angiogenic effect of hUCMSC transplantation in stroked brain.
Collapse
Affiliation(s)
- J Zhu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - Q Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - Y Jiang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - L Wu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - G Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - X Liu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| |
Collapse
|
187
|
Myricanol induces apoptotic cell death and anti-tumor activity in non-small cell lung carcinoma in vivo. Int J Mol Sci 2015; 16:2717-31. [PMID: 25629230 PMCID: PMC4346861 DOI: 10.3390/ijms16022717] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/21/2015] [Indexed: 12/17/2022] Open
Abstract
This study explored the inhibiting effect and mechanism of myricanol on lung adenocarcinoma A549 xenografts in nude mice. Forty nude mice with subcutaneous A549 xenografts were randomly divided into five groups: high-dose myricanol (40 mg/kg body weight) group; middle-dose myricanol (20 mg/kg body weight) group; low-dose myricanol (10 mg/kg body weight) group; polyethylene glycol 400 vehicle group (1 mL/kg); and tumor model group. Nude mice were sacrificed after 14 days of treatment and the tumor inhibition rate (TIR, %) was then calculated. The relative mRNA expression levels of Bax, Bcl-2, VEGF, HIF-1α, and survivin in the tumor tissues were determined by real-time PCR. TUNEL assay was applied to determine cellular apoptosis, while IHC test was performed to detect the protein expression levels of Bax, Bcl-2, VEGF, HIF-1α, and survivin. The TIR of the three myricanol-treated groups ranged from 14.9% to 38.5%. The IHC results showed that the protein expression of Bcl-2, VEGF, HIF-1α, and survivin were consistently downregulated, whereas that of Bax was upregulated after myricanol treatment. Myricanol also significantly upregulated the mRNA expression of Bax and downregulated that of Bcl-2, VEGF, HIF-1α, and survivin in a dose-dependent manner (p < 0.05 to 0.001). These results are consistent with those of IHC. The TUNEL assay results indicated that apoptotic-positive cells significantly increased in the myricanol-treated tumor tissues compared with the cells of the vehicle control group (p < 0.01 to 0.001). These data suggest that myricanol could significantly decelerate tumor growth in vivo by inducing apoptosis.
Collapse
|
188
|
Lu M, Tang F, Zhang J, Luan A, Mei M, Xu C, Zhang S, Wang H, Maslov LN. Astragaloside IV attenuates injury caused by myocardial ischemia/reperfusion in rats via regulation of toll-like receptor 4/nuclear factor-κB signaling pathway. Phytother Res 2015; 29:599-606. [PMID: 25604645 DOI: 10.1002/ptr.5297] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/25/2014] [Accepted: 12/15/2014] [Indexed: 12/17/2022]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury, in which inflammatory response and cell apoptosis play a vital role, is frequently encountered in clinical practice. Astragaloside IV (AsIV), a small molecular saponin of Astragalus membranaceus, has been shown to confer protective effects against many cardiovascular diseases. The present study was aimed to investigate the antiinflammatory and antiapoptotic effects and the possible mechanism of AsIV on MI/R injury in rats. Rats were randomly divided into sham operation group, MI/R group and groups with combinations of MI/R and different doses of AsIV. The results showed that the expressions of myocardial toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) were significantly increased, and apoptosis of cardiomyocytes was induced in MI/R group compared with that in sham operation group. Administration of AsIV attenuated MI/R injury, downregulated the expressions of TLR4 and NF-κB and inhibited cell apoptosis as evidenced by decreased terminal deoxynucleotidyl transferase dUTP nick end labeling positive cells, B-cell lymphoma-2 associated X protein and caspase-3 expressions and increased B-cell lymphoma-2 expression compared with that in MI/R group. In addition, AsIV treatment reduced levels of inflammatory cytokines induced by MI/R injury. In conclusion, our results demonstrated that AsIV downregulates TLR4/NF-κB signaling pathway and inhibits cell apoptosis, subsequently attenuating MI/R injury in rats.
Collapse
Affiliation(s)
- Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Liaoning Medical University, Jinzhou, 121001, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Relationship between serum endothelin-1 level and spontaneous reperfusion in patients with acute myocardial infarction. Coron Artery Dis 2015; 26:37-41. [DOI: 10.1097/mca.0000000000000175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
190
|
From Chinese Science Bulletin to Science Bulletin: celebrate the coming 50th birthday. Sci Bull (Beijing) 2015; 60:2145-2150. [PMID: 32215225 PMCID: PMC7089006 DOI: 10.1007/s11434-015-0974-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
191
|
Javan H, Szucsik AM, Li L, Schaaf CL, Salama ME, Selzman CH. Cardiomyocyte p65 nuclear factor-κB is necessary for compensatory adaptation to pressure overload. Circ Heart Fail 2014; 8:109-18. [PMID: 25480781 DOI: 10.1161/circheartfailure.114.001297] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Nuclear factor κB (NF-κB) is often implicated in contributing to the detrimental effects of cardiac injury. This ostensibly negative view of NF-κB competes with its important role in the normal host inflammatory and immune response. We have previously demonstrated that pharmacological inhibition of NF-κB at the time of acute pressure overload accelerates the progression of left ventricular hypertrophy to heart failure in mice. NF-κB regulates angiogenesis and other factors responsible for compensatory reaction to intracellular hypoxia. We hypothesized that impaired angiogenesis may be the trigger, not the result, of pathological left ventricular hypertrophy through NF-κB-related pathways. METHODS AND RESULTS Transgenic mice were generated with cardiomyocyte-specific deletion of the p65 subunit of NF-κB. Mice underwent transverse aortic constriction and serially followed up with echocardiography for 6 weeks. Cardiomyocyte p65 NF-κB deletion promoted maladaptive left ventricular hypertrophy and accelerated progression toward heart failure as measured by ejection fraction, left ventricular mass, and lung congestion. Transgenic mice had higher levels of fibrosis and periostin expression. Whole-field digital microscopy revealed increased capillary domain areas in knockout mice while concurrently demonstrating decreased microvessel density. This observation was associated with decreased expression of hypoxia-inducible factor 1α. CONCLUSIONS Rather than developing compensatory left ventricular hypertrophy, pressure overload in cardiomyocyte NF-κB-deficient mice resulted in functional deterioration that was associated with increased fibrosis, decreased hypoxia-inducible factor expression, and decreased microvessel density. These observations mechanistically implicate NF-κB, and its regulation of hypoxic stress, as an important factor determining the path between adaptive hypertrophy and maladaptive heart failure.
Collapse
Affiliation(s)
- Hadi Javan
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City
| | - Amanda M Szucsik
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City
| | - Ling Li
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City
| | - Christin L Schaaf
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City
| | - Mohamed E Salama
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City
| | - Craig H Selzman
- From the Division of Cardiothoracic Surgery, Department of Surgery and Molecular Medicine (H.J., A.M.S., L.L., C.L.S., C.H.S.) and Department of Pathology, ARUP Institute for Research and Development (M.E.S.), University of Utah, Salt Lake City.
| |
Collapse
|
192
|
Salem AM, Ahmed HH, Atta HM, Ghazy MA, Aglan HA. Potential of bone marrow mesenchymal stem cells in management of Alzheimer's disease in female rats. Cell Biol Int 2014; 38:1367-83. [PMID: 25044885 DOI: 10.1002/cbin.10331] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 05/17/2014] [Indexed: 07/27/2024]
Abstract
Alzheimer's disease (AD) has been called the disease of the century with significant clinical and socioeconomic impacts. Pharmacological treatment has limited efficacy and only provides symptomatic relief without long-term cure. Accordingly, there is an urgent need to develop novel and effective medications for AD. Stem cell-based therapy is a promising approach to handling neurodegenerative diseases. Therefore, the current study aimed to explore the possible therapeutic role of single intravenous injection of bone marrow derived mesenchymal stem cells (BM-MSCs) after 4 months in management of AD in the experimental model. The work also extended to compare the therapeutic potential of BM-MSCs with 2 conventional therapies of AD; rivastigmine and cerebrolysin administered daily. BM-MSCs were able to home at the injured brains and produced significant increases in the number of positive cells for choline acetyltransferase (ChAT) and survivin expression, as well as selective AD indicator-1 (seladin-1) and nestin gene expression. Histopathological examination indicated that BM-MSCs could remove beta-amyloid plaques from hippocampus. Significant improvement in these biomarkers was similar to or better sometimes than the reference drugs, clearly showing the potential therapeutic role of BM-MSCs against AD through their anti-apoptotic, neurogenic and immunomodulatory properties.
Collapse
Affiliation(s)
- Ahmed M Salem
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
193
|
Ghosh A, Pechota A, Coleman D, Upchurch GR, Eliason JL. Cigarette smoke-induced MMP2 and MMP9 secretion from aortic vascular smooth cells is mediated via the Jak/Stat pathway. Hum Pathol 2014; 46:284-94. [PMID: 25537973 DOI: 10.1016/j.humpath.2014.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 11/10/2014] [Accepted: 11/14/2014] [Indexed: 10/24/2022]
Abstract
It is hypothesized that cigarette smoke may increase MMP2 and MMP9 secretion through Jak/Stat pathway in the aorta, thereby facilitating abdominal aortic aneurysm (AAA) formation/progression in smokers. We observed through zymograms that treatment of male rat aortic vascular smooth muscle cells (RASMC) with an aqueous extract of cigarette smoke (CSE) for 24 hours resulted in a significant increase in pro-MMP9 (P = .005) and a modest increase in pro-MMP2 (P = .055) production. Western blot with protein extracts from CSE-treated RASMC showed up-regulation of pStat3, pJak2, and T-Jak2 and unchanged levels of T-Stat3. Transfection of RASMC with small interfering RNAs for Jak2, Stat3, or both Jak2 and Stat3 significantly reduced pro-MMP9 (P < .005) and pro-MMP2 (P < .05) in medium of CSE-treated RASMC compared with control small interfering RNA-transfected cells. Immunoprecipitation with total Jak2 antibody showed increased pStat3 and T-Stat3 in the cytoplasm and nucleus of CSE-treated RASMC. Immunofluorescence revealed increased presence of pJak2, T-Jak2, pStat3, and T-Stat3 in the cytoplasm and nucleus of the CSE-treated cells. Treatment of control human tissues with CSE resulted in pro-MMP9 secretion and up-regulation of the Jak/Stat proteins. In addition, AAA tissues showed more pJak2 and pStat3 than control human tissues. Therefore, inhibiting the Jak/Stat pathway could be a potential therapeutic approach in the treatment of AAA.
Collapse
Affiliation(s)
- Abhijit Ghosh
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Angela Pechota
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Dawn Coleman
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Gilbert R Upchurch
- University of Virginia, Division of Vascular and Endovascular Surgery, Charlottesville, VA 800679
| | - Jonathan L Eliason
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867.
| |
Collapse
|
194
|
Horvath B, Bers DM. The late sodium current in heart failure: pathophysiology and clinical relevance. ESC Heart Fail 2014; 1:26-40. [PMID: 28834665 DOI: 10.1002/ehf2.12003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
Large and growing body of data suggest that an increased late sodium current (INa,late ) can have a significant pathophysiological role in heart failure and other heart diseases. The first goal of this article is to describe how INa,late functions under physiological circumstances. The second goal is to show the wide range of cellular mechanisms that can increase INa,late in cardiac disease, and also to describe how the up-regulated INa,late contributes to the pathophysiology of heart failure. The final section of the article discusses the possible use of INa,late -modifying drugs in heart failure, on the basis of experimental and preclinical data.
Collapse
Affiliation(s)
- Balazs Horvath
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
195
|
Papp Z, Borbély A, Paulus WJ. CrossTalk opposing view: the late sodium current is not an important player in the development of diastolic heart failure (heart failure with a preserved ejection fraction). J Physiol 2014; 592:415-7. [PMID: 24488067 DOI: 10.1113/jphysiol.2013.264242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
196
|
Awad H, Nolette N, Hinton M, Dakshinamurti S. AMPK and FoxO1 regulate catalase expression in hypoxic pulmonary arterial smooth muscle. Pediatr Pulmonol 2014; 49:885-97. [PMID: 24167160 DOI: 10.1002/ppul.22919] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 08/13/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Hypoxia and reactive oxygen species (ROS) including H(2)O(2) play major roles in triggering and progression of pulmonary vascular remodeling in persistent pulmonary hypertension. Catalase (CAT), the major endogenous enzyme scavenging H(2)O(2), is regulated in a tissue- and context-specific manner. OBJECTIVE To investigate mechanisms by which hypoxia and H(2)O(2) regulate catalase expression, and the role of AMPK-FoxO pathway, in neonatal porcine pulmonary artery smooth muscle (PASMC). DESIGN/METHODS PASMC were grown in hypoxia (10% O(2)) or normoxia (21% O(2)) for 72 hr. We measured catalase activity and lipid peroxidation; CAT, FoxO1, and FoxO3a expression by qPCR; protein contents of CAT, FoxOs, p-AMPK, p-AKT, p-JNK, p-ERK1/2 in whole lysates, and FoxOs in nuclear extracts, by immunoblot; and FoxO-1 nuclear localization by immunocytochemistry, quantified by laser scanning cytometry. RESULTS Hypoxia upregulated CAT transcription, content and activity, by increasing CAT transcription factors FoxO1 and FoxO3a mRNA, and promoting nuclear translocation of FoxO1. However, lipid peroxidation increased in hypoxic PASMC. Among candidate FoxO regulatory kinases, hypoxia activated AMPK, and decreased p-Akt and ERK1/2. AMPK activation increased FoxO1 (total and nuclear) and CAT, while AMPK inhibition inhibited FoxO1 and CAT, but not FoxO3a. Exogenous H(2)O(2) decreased p-AMPK and increased p-AKT in hypoxic PASMC. This decreased active FoxO1, and reduced mRNA and protein content of CAT. Hypoxic induction of CAT, AKT inhibition (LY294002), or addition of PEG-catalase partly ameliorated the H(2)O(2) -mediated loss of nuclear FoxO1. CONCLUSIONS Hypoxia induces catalase expression, though this adaptation is insufficient to protect PASMC from hypoxia-induced lipid peroxidation. This occurs via hypoxic activation of AMPK, which promotes nuclear FoxO1 and thus catalase expression. Exogenous ROS may downregulate cellular antioxidant defenses; H(2)O(2) activates survival factor Akt, decreasing nuclear FoxO1 and thus catalase.
Collapse
Affiliation(s)
- Hanan Awad
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Canada
| | | | | | | |
Collapse
|
197
|
Mayyas F, Al-Jarrah M, Ibrahim K, Mfady D, Van Wagoner DR. The significance of circulating endothelin-1 as a predictor of coronary artery disease status and clinical outcomes following coronary artery catheterization. Cardiovasc Pathol 2014; 24:19-25. [PMID: 25213716 DOI: 10.1016/j.carpath.2014.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/16/2014] [Accepted: 08/08/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND/OBJECTIVES Coronary artery disease (CAD) is responsible for significant morbidity and mortality. Inflammatory, pro-thrombotic and structural factors contribute to the etiology of CAD. This study sought to determine the relationship of plasma endothelin-1 (pET-1), a potent vasoconstrictor, mitogen and modulator of cardiac inflammation, to clinical characteristics and outcomes of CAD patients. METHODS Blood samples were collected from 336 patients with underlying chest pain or recent myocardial infarction (MI), prior to coronary catheterization. pET-1 was correlated with clinical characteristics and outcomes following catheterization and at 30-day follow-up. RESULTS pET-1 was higher in recent MI patients than in patients with CAD (coronary occlusion≥50%) or without CAD (<50%) (Mean±sem (pg/ml): 2.12±0.13, 1.51±0.10, 1.21±0.06; 95% confidence interval (1.85-2.38, 1.31-1.72, 1.07-1.32; respectively, P<.0001). Patients with ST elevation MI (STEMI) had higher pET-1 than non-STEMI (P=.008). pET-1 was associated with heart failure (HF) and low left ventricular ejection fraction (LVEF) and was highest in MI patients presented with acute HF. At 30-day follow up, pET-1 was not associated with the change in LVEF. In multivariate analysis, pET-1 was positively associated with age, smoking, HF, CAD status, and need for revascularization by coronary artery bypass surgery (CABG). pET-1 was negatively correlated with LVEF and preoperative statin use. CONCLUSIONS pET-1 is associated with recent MI, HF, age, smoking, CABG, and low LVEF. Preoperative statin use was associated with lower pET-1. pET-1 may serve as a risk marker and a potential therapeutic target in CAD patients.
Collapse
Affiliation(s)
- Fadia Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Mohammad Al-Jarrah
- Department of General Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan; Department of Internal Medicine, Division of Cardiology, Faculty of Medicine, King Abdullah University Hospital, Irbid, Jordan
| | - Khalid Ibrahim
- Department of General Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan; Department of General Surgery, Division of Cardiovascular Surgery, Faculty of Medicine, King Abdullah University Hospital, Irbid, Jordan
| | - Doaa Mfady
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - David R Van Wagoner
- Department of Molecular Cardiology, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
198
|
Thomas CM, Yong QC, Rosa RM, Seqqat R, Gopal S, Casarini DE, Jones WK, Gupta S, Baker KM, Kumar R. Cardiac-specific suppression of NF-κB signaling prevents diabetic cardiomyopathy via inhibition of the renin-angiotensin system. Am J Physiol Heart Circ Physiol 2014; 307:H1036-45. [PMID: 25085967 DOI: 10.1152/ajpheart.00340.2014] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of NF-κB signaling in the heart may be protective or deleterious depending on the pathological context. In diabetes, the role of NF-κB in cardiac dysfunction has been investigated using pharmacological approaches that have a limitation of being nonspecific. Furthermore, the specific cellular pathways by which NF-κB modulates heart function in diabetes have not been identified. To address these questions, we used a transgenic mouse line expressing mutated IκB-α in the heart (3M mice), which prevented activation of canonical NF-κB signaling. Diabetes was developed by streptozotocin injections in wild-type (WT) and 3M mice. Diabetic WT mice developed systolic and diastolic cardiac dysfunction by the 12th week, as measured by echocardiography. In contrast, cardiac function was preserved in 3M mice up to 24 wk of diabetes. Diabetes induced an elevation in cardiac oxidative stress in diabetic WT mice but not 3M mice compared with nondiabetic control mice. In diabetic WT mice, an increase in the phospholamban/sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 ratio and decrease in ryanodine receptor expression were observed, whereas diabetic 3M mice showed an opposite effect on these parameters of Ca(2+) handling. Significantly, renin-angiotensin system activity was suppressed in diabetic 3M mice compared with an increase in WT animals. In conclusion, these results demonstrate that inhibition of NF-κB signaling in the heart prevents diabetes-induced cardiac dysfunction through preserved Ca(2+) handling and inhibition of the cardiac renin-angiotensin system.
Collapse
Affiliation(s)
- Candice M Thomas
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Qian Chen Yong
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rodolfo M Rosa
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - Rachid Seqqat
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Shanthi Gopal
- Central Texas Veterans Health Care System, Temple, Texas
| | - Dulce E Casarini
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - W Keith Jones
- Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, Illinois
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rajesh Kumar
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas;
| |
Collapse
|
199
|
Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B. Mesenchymal stem cells and hypoxia: where are we? Mitochondrion 2014; 19 Pt A:105-12. [PMID: 25034305 DOI: 10.1016/j.mito.2014.07.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/09/2014] [Indexed: 12/23/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are involved in the organization and maintenance of tissue integrity. MSCs have also attracted attention as a promising tool for cell therapy and regenerative medicine. However, their usage is limited due to cell impairment induced by an extremely harsh microenvironment during transplantation ex vivo. The microenvironment of MSCs in tissue depots is characterized by rather low oxygen consumption, demonstrating that MSCs might be quite resistant to oxygen limitation. However, accumulated data revealed that the response of MSCs to hypoxic conditions is rather controversial, demonstrating both damaging and ameliorating effects. Here, we make an attempt to summarize recent knowledge on the survival of MSCs under low oxygen conditions of varying duration and severity and to elucidate the mechanisms of MSC resistance/sensitivity to hypoxic impact.
Collapse
Affiliation(s)
- L B Buravkova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia; Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia
| | - E R Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - V Gogvadze
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden
| | - B Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| |
Collapse
|
200
|
Ma J, Song Y, Shryock JC, Hu L, Wang W, Yan X, Zhang P, Belardinelli L. Ranolazine Attenuates Hypoxia- and Hydrogen Peroxide-induced Increases in Sodium Channel Late Openings in Ventricular Myocytes. J Cardiovasc Pharmacol 2014; 64:60-8. [DOI: 10.1097/fjc.0000000000000090] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|