101
|
Vasani A, Kumar MS. Advances in the proteomics of amniotic fluid to detect biomarkers for chromosomal abnormalities and fetomaternal complications during pregnancy. Expert Rev Proteomics 2019; 16:277-286. [PMID: 30722712 DOI: 10.1080/14789450.2019.1578213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Amniotic fluid (AF) is a dynamic and complex mixture that reflects the physiological condition of developing fetus. In the last decade, proteomic analysis of AF for 16-18 weeks normal pregnancy has been done for the composition and functions of this fluid. Other body fluids such as urine, sweat, tears, etc. are being used for diagnosis of disease, but an insight into protein biomarkers of amniotic fluid can save the fetus and mother from future complications. Areas covered: We have covered the proteomics of amniotic fluid done since 2000, in order to strengthen the establishment of these techniques as a recognized diagnostic tool in the field. After classifying the diseases based on chromosomal aneuploidies, gestational changes, and inflammation caused during pregnancy; we have focused on amniotic fluid to detect various complications during and post pregnancy and its effect on the fetomaternal relationship. Expert comment: The main protein biomarkers responsible for various syndromes, diseases, and complications have been summarized. Major proteins identified for gestational conditions are IGFBP-1, fibrinogen, neutrophil defensins like calgranulins A and C, cathelicidin, APOA1, TRFE, etc. Validation of particular technique and establishing a single standardized biomarker for the diagnosis to avoid any overlapping for different diseases is required. After certain improvements, proteomics approach can be considered for diagnosis of diseases associated with fetal-maternal health.
Collapse
Affiliation(s)
- Aayushi Vasani
- a Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management , SVKM'S NMIMS , V.L. Mehta Road, Vile Parle west, Mumbai - 400056 , India
| | - Maushmi S Kumar
- a Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management , SVKM'S NMIMS , V.L. Mehta Road, Vile Parle west, Mumbai - 400056 , India
| |
Collapse
|
102
|
Xu M, Liu PP, Li H. Innate Immune Signaling and Its Role in Metabolic and Cardiovascular Diseases. Physiol Rev 2019; 99:893-948. [PMID: 30565509 DOI: 10.1152/physrev.00065.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is an evolutionarily conserved system that senses and defends against infection and irritation. Innate immune signaling is a complex cascade that quickly recognizes infectious threats through multiple germline-encoded cell surface or cytoplasmic receptors and transmits signals for the deployment of proper countermeasures through adaptors, kinases, and transcription factors, resulting in the production of cytokines. As the first response of the innate immune system to pathogenic signals, inflammatory responses must be rapid and specific to establish a physical barrier against the spread of infection and must subsequently be terminated once the pathogens have been cleared. Long-lasting and low-grade chronic inflammation is a distinguishing feature of type 2 diabetes and cardiovascular diseases, which are currently major public health problems. Cardiometabolic stress-induced inflammatory responses activate innate immune signaling, which directly contributes to the development of cardiometabolic diseases. Additionally, although the innate immune elements are highly conserved in higher-order jawed vertebrates, lower-grade jawless vertebrates lack several transcription factors and inflammatory cytokine genes downstream of the Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) pathways, suggesting that innate immune signaling components may additionally function in an immune-independent way. Notably, recent studies from our group and others have revealed that innate immune signaling can function as a vital regulator of cardiometabolic homeostasis independent of its immune function. Therefore, further investigation of innate immune signaling in cardiometabolic systems may facilitate the discovery of new strategies to manage the initiation and progression of cardiometabolic disorders, leading to better treatments for these diseases. In this review, we summarize the current progress in innate immune signaling studies and the regulatory function of innate immunity in cardiometabolic diseases. Notably, we highlight the immune-independent effects of innate immune signaling components on the development of cardiometabolic disorders.
Collapse
Affiliation(s)
- Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Peter P Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| |
Collapse
|
103
|
Esmaeilzade B, Artimani T, Amiri I, Najafi R, Shahidi S, Sabec M, Farzadinia P, Zare M, Zahiri M, Soleimani Asl S. Dimethyloxalylglycine preconditioning enhances protective effects of bone marrow-derived mesenchymal stem cells in Aβ- induced Alzheimer disease. Physiol Behav 2019; 199:265-272. [PMID: 30500334 DOI: 10.1016/j.physbeh.2018.11.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cell (MSC) transplantation therapy has been proposed as a promising approach for the treatment of neurodegenerative disease. Chemical and pharmacological preconditioning before transplantation could optimize the therapeutic properties of transplanted MSCs. In this study, we hypothesized that preconditioning treatment with a prolyl hydroxylase inhibitor, dimethyloxalylglycine (DMOG), will increase MSC efficacy and paracrine effects in an amyloid-β (Aβ)-injected Alzheimer rat model. MSCs were incubated in different concentrations of DMOG for 24 h. Cell viability, migration, and antioxidant capacity was assessed in DMOG-treated and non-treated MSCs before transplantation into Aβ-injected rats. In vitro analysis revealed that DMOG treatment increased cell viability, migration, and expression of CXCR4, CCR2, Nrf2, and HIF-1α in the MSCs. Our in vivo results show that DMOG preconditioning enhances a MSC-mediated rescue of learning and memory function in Aβ-injected rats. Furthermore, we found an increased level of BDNF and total antioxidant capacity in the hippocampus of Aβ-injected rats following transplantation of preconditioned relative to untreated MSCs. Our results suggest that preconditioning MSCs with DMOG before transplantation may enhance the efficacy of stem cell based therapy in neurodegenerative disease.
Collapse
Affiliation(s)
| | - Tayebe Artimani
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Iran; Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Amiri
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Iran; Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Centre for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marie Sabec
- Institut Pasteur, Neurobiologie Intégrative des Systèmes Cholinergiques, Paris, France
| | - Parviz Farzadinia
- Department of Anatomy, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadali Zare
- Department of Anatomy, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maria Zahiri
- Department of Anatomy, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Sara Soleimani Asl
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Iran; Neurophysiology Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
104
|
Wang Z, Li C, Xing R, Shao Y, Zhao X, Zhang W, Guo M. β-Integrin mediates LPS-induced coelomocyte apoptosis in sea cucumber Apostichopus japonicus via the integrin/FAK/caspase-3 signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 91:26-36. [PMID: 30339873 DOI: 10.1016/j.dci.2018.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/13/2018] [Accepted: 10/15/2018] [Indexed: 06/08/2023]
Abstract
Lipopolysaccharides (LPS) can induce the apoptosis of coelomocytes in Apostichopus japonicus (A. japonicus), and β-integrin serves as an apoptotic inhibitor during this process. However, the underlying mechanism in invertebrates is largely unknown. Integrin/focal adhesion kinase (FAK) signaling pathway modulates the apoptosis in vertebrates. In this study, a novel FAK was identified from A. japonicus (designated as AjFAK) by β-integrin (designated as AjITGB) -mediated GST-pull down assay. This interaction was further validated in the LPS-exposed coelomocytes through co-immunoprecipitation and immunofluorescence analyses. To investigate the functional role of AjFAK in AjITGB-mediated coelomocyte apoptosis, we cloned the full-length cDNA of AjFAK and characterized its relationship with AjITGB through real-time PCR. The mRNA expression levels of AjFAK exhibited consistent expression patterns with those of AjITGB in our previous work with 0.48- and 0.22-fold decreases at 12 and 96 h in LPS-exposed coelomocytes and in Vibrio splendidus challenged sea cucumber, respectively. Moreover, the expression level of AjFAK decreased to 0.35-fold in AjITGB knockdown treatment by specific small interference RNA (siRNA). We further performed an assay for the apoptotic rate of coelomocytes in AjITGB, AjFAK, and AjITGB/AjFAK silencing conditions and found that their apoptotic percentages increased to 26%, 25%, and 30%, respectively, compared with those of the control. Finally, the expression levels of four caspases from A. japonicus were also investigated to determine the apoptotic effector. After AjITGB or AjFAK was silenced, the mRNA levels of caspase-3 were 6.6-fold and 2.5-fold higher than those of the control, respectively. In addition, the enzymatic activity of caspase-3 was enhanced to 1.82- and 1.79-fold that of the control in the two groups. However, no significant changes were detected in caspase-2/6/8. All our results supported that β-integrin mediated the LPS-induced coelomocyte apoptosis in sea cucumber via the integrin/FAK/caspase-3 signaling pathway.
Collapse
Affiliation(s)
- Zhenhui Wang
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China; College of Life Sciences, Yantai University, Yantai, 264005, PR China.
| | - Ronglian Xing
- College of Life Sciences, Yantai University, Yantai, 264005, PR China
| | - Yina Shao
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| | - Ming Guo
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, PR China
| |
Collapse
|
105
|
Yan X, Xie B, Wu G, Hu J, Wang D, Cai X, Li J. Interleukin-37: The Effect of Anti-Inflammatory Response in Human Coronary Artery Endothelial Cells. Mediators Inflamm 2019; 2019:2650590. [PMID: 30728750 PMCID: PMC6341264 DOI: 10.1155/2019/2650590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/07/2018] [Accepted: 11/06/2018] [Indexed: 02/05/2023] Open
Abstract
Interleukin-37 (IL-37) is unique in the IL-1 family since it broadly suppresses innate immunity and elevates in humans with inflammatory and autoimmune diseases. IL-37 shows definite groups and transcripts for human IL37 gene, but it is still not completely understood the effect and mechanisms of inflammatory response in endothelial cells. It is well accepted that endothelial dysfunction caused by inflammation is a key initiating event in atherosclerotic plaque formation, which leads to the occurrence and development of the cardiovascular adverse events in clinical since the inflammatory responses of endothelial cells could induce and enhance the deposition of extensive lipid and the formation of atherosclerotic plaque in the intima. Thus, it is essential to investigate the role and potential mechanisms in endothelial inflammatory response to prevent the formation and development of many cardiovascular diseases including atherosclerosis. So far, the recent studies have revealed that IL-37 is able to inhibit inflammatory response by suppressing the TLR2-NF-κB-ICAM-1 pathway intracellularly in human coronary artery endothelial cells (HCAECs). Further, the role of IL-37 may be related to the IL-18 pathway extracellularly and involved in the adhesion and transmigration of neutrophils in HCAECs.
Collapse
Affiliation(s)
- Xianfeng Yan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bin Xie
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Guihai Wu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jing Hu
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, China
| | - Di Wang
- Department of Dermatovenereology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiangna Cai
- Department of Plastic Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jilin Li
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
106
|
Patel S, Athirasala A, Menezes PP, Ashwanikumar N, Zou T, Sahay G, Bertassoni LE. Messenger RNA Delivery for Tissue Engineering and Regenerative Medicine Applications. Tissue Eng Part A 2019; 25:91-112. [PMID: 29661055 PMCID: PMC6352544 DOI: 10.1089/ten.tea.2017.0444] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/09/2018] [Indexed: 12/25/2022] Open
Abstract
The ability to control cellular processes and precisely direct cellular reprogramming has revolutionized regenerative medicine. Recent advances in in vitro transcribed (IVT) mRNA technology with chemical modifications have led to development of methods that control spatiotemporal gene expression. Additionally, there is a current thrust toward the development of safe, integration-free approaches to gene therapy for translational purposes. In this review, we describe strategies of synthetic IVT mRNA modifications and nonviral technologies for intracellular delivery. We provide insights into the current tissue engineering approaches that use a hydrogel scaffold with genetic material. Furthermore, we discuss the transformative potential of novel mRNA formulations that when embedded in hydrogels can trigger controlled genetic manipulation to regenerate tissues and organs in vitro and in vivo. The role of mRNA delivery in vascularization, cytoprotection, and Cas9-mediated xenotransplantation is additionally highlighted. Harmonizing mRNA delivery vehicle interactions with polymeric scaffolds can be used to present genetic cues that lead to precise command over cellular reprogramming, differentiation, and secretome activity of stem cells-an ultimate goal for tissue engineering.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
| | - Paula P. Menezes
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Postgraduate Program in Health Sciences, Department of Pharmacy, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - N. Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Ting Zou
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
| | - Luiz E. Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
107
|
Wu Q, Zheng R, Wang J, Wang J, Li S. CT perfusion imaging of cerebral microcirculatory changes following subarachnoid hemorrhage in rabbits: Specific role of endothelin-1 receptor antagonist. Brain Res 2018; 1701:196-203. [PMID: 30244111 DOI: 10.1016/j.brainres.2018.09.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cerebral vasospasm may lead to delayed ischemic neurological deficits following subarachnoid hemorrhage (SAH). Endothelin (ET-1) is an important factor participating in cerebral vasospasm underlying SAH. We used a specific endothelin receptor antagonist, BQ123 to assess the specific role of endothelin-1 receptor antagonist in cerebral vasospasm in a rabbit model of SAH by examining plasma ET-1 levels and the principal CT perfusion (CTP) parameters pertinent to the hemodynamic status of microcirculation following SAH. METHODS 102 male New Zealand white rabbits were divided into control, SAH and SAH + BQ123 intervention group (BQ123 group). Rabbit SAH model was established by double hemorrhage injection of autologous blood into the cisterna magna; Aquilion ONE was used to collect cerebral blood flow (CBF), cerebral blood volume (CBV), and mean transit time (MTT) which were used to evaluate cerebral microcirculation hemodynamics; Elisa was used to assess plasma ET-1 levels. Data were collected on days 1, 4, 7 and 14 following SAH, respectively. RESULTS Compared with the control group, the CBF in the SAH group was significantly lower, while the MTT was significantly higher. The CBF decreased on the 4th day and reached the lowest on the 7th day. The MTT began to rise on the 4th day and peaked on the 7th day. While in the BQ123 intervention group, the CBF significantly increased while the MTT significantly decreased on the 1st and the 4th days, respectively. Compared with SAH group, plasma ET-1 levels in BQ123 group significantly increased on the earlier (1st and 4th days) but not later days (between the 7th and 14th days). In addition, the inflammatory infiltration of brain tissues in rabbits treated with BQ123 post-SAH was significantly reduced compared with SAH group. CONCLUSION CTP can quantify the therapeutic effect of BQ123 after SAH; Selective blockade of ET-1 endothelin receptor, BQ123 significantly improved microcirculatory perfusion along with a reduction in resultant vasogenic inflammatory responses. The effect of BQ123 on the cerebral microcirculation was lobe dependent.
Collapse
Affiliation(s)
- Quanyang Wu
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Ruibin Zheng
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Jiao Wang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Jiaqi Wang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Songbai Li
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China.
| |
Collapse
|
108
|
Ostrowski RP, Zhang JH. The insights into molecular pathways of hypoxia-inducible factor in the brain. J Neurosci Res 2018; 98:57-76. [PMID: 30548473 DOI: 10.1002/jnr.24366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022]
Abstract
The objectives of this present work were to review recent developments on the role of hypoxia-inducible factor (HIF) in the survival of cells under normoxic versus hypoxic and inflammatory brain conditions. The dual nature of HIF effects appears well established, based on the accumulated evidence of HIF playing both the role of adaptive factor and mediator of cell demise. Cellular HIF responses depend on pathophysiological conditions, developmental phase, comorbidities, and administered medications. In addition, HIF-1α and HIF-2α actions may vary in the same tissues. The multiple roles of HIF in stem cells are emerging. HIF not only regulates expression of target genes and thereby influences resultant protein levels but also contributes to epigenetic changes that may reciprocally provide feedback regulations loops. These HIF-dependent alterations in neurological diseases and its responses to treatments in vivo need to be examined alongside with a functional status of subjects involved in such studies. The knowledge of HIF pathways might be helpful in devising HIF-mimetics and modulating drugs, acting on the molecular level to improve clinical outcomes, as exemplified here by clinical and experimental data of selected brain diseases, occasionally corroborated by the data from disorders of other organs. Because of complex role of HIF in brain injuries, prospective therapeutic interventions need to differentially target HIF responses depending on their roles in the molecular mechanisms of neurologic diseases.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - John H Zhang
- Departments of Anesthesiology and Physiology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
109
|
Huang Q, Lan T, Lu J, Zhang H, Zhang D, Lou T, Xu P, Ren J, Zhao D, Sun L, Li X, Wang J. DiDang Tang Inhibits Endoplasmic Reticulum Stress-Mediated Apoptosis Induced by Oxygen Glucose Deprivation and Intracerebral Hemorrhage Through Blockade of the GRP78-IRE1/PERK Pathways. Front Pharmacol 2018; 9:1423. [PMID: 30564125 PMCID: PMC6288198 DOI: 10.3389/fphar.2018.01423] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022] Open
Abstract
DiDang Tang (DDT), a Chinese traditional medicine formula, contains 4 Chinese traditional medicine substances, has been widely used to treat intracerebral hemorrhage (ICH) patients. However, the molecular mechanisms of DDT for protecting neurons from oxygen and glucose deprivation (OGD)-induced endoplasmic reticulum (ER) stress and apoptosis after ICH still remains elusive. In this study, high-performance liquid chromatography fingerprint analysis was performed to learn the features of the chemical compositions of DDT. OGD-induced ER stress, Ca2+ overload, and mitochondrial apoptosis were investigated in nerve growth factor -induced PC12, primary neuronal cells, and ICH rats to evaluate the protective effect of DDT. We found that DDT treatment protected neurons against OGD-induced damage and apoptosis by increasing cell viability and reducing the release of lactate dehydrogenase. DDT decreased OGD-induced Ca2+ overload and ER stress through the blockade of the glucose-regulated protein 78 (GRP78)- inositol-requiring protein 1α (IRE1)/ protein kinase RNA-like ER kinase (PERK) pathways and also inhibited apoptosis by decreasing mitochondrial damage. Moreover, we observed similar findings when we studied DDT for inhibition of ER stress in a rat model of ICH. In addition, our experiments further confirmed the neuroprotective potential of DDT against tunicamycin (TM)-induced neural damage. Our in vitro and in vivo results indicated that the neuroprotective effect of DDT against ER stress damage and apoptosis occurred mainly by blocking the GPR78-IRE1/PERK pathways. Taken together, it provides reliable experimental evidence and explains the molecular mechanism of DDT for the treatment of patients with ICH.
Collapse
Affiliation(s)
- Qingxia Huang
- Research Center of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Tianye Lan
- Department of Encephalopathy, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - He Zhang
- Research Center of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Scientific Research Office, Changchun University of Chinese Medicine, Changchun, China
| | - Tingting Lou
- Research Center of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Peng Xu
- Department of Encephalopathy, Changchun University of Chinese Medicine, Changchun, China
| | - Jixiang Ren
- Department of Encephalopathy, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
110
|
Vaněčková I, Hojná S, Kadlecová M, Vernerová Z, Kopkan L, Červenka L, Zicha J. Renoprotective effects of ET(A) receptor antagonists therapy in experimental non-diabetic chronic kidney disease: Is there still hope for the future? Physiol Res 2018; 67:S55-S67. [PMID: 29947528 DOI: 10.33549/physiolres.933898] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) is a life-threatening disease arising as a frequent complication of diabetes, obesity and hypertension. Since it is typically undetected for long periods, it often progresses to end-stage renal disease. CKD is characterized by the development of progressive glomerulosclerosis, interstitial fibrosis and tubular atrophy along with a decreased glomerular filtration rate. This is associated with podocyte injury and a progressive rise in proteinuria. As endothelin-1 (ET-1) through the activation of endothelin receptor type A (ET(A)) promotes renal cell injury, inflammation, and fibrosis which finally lead to proteinuria, it is not surprising that ET(A) receptors antagonists have been proven to have beneficial renoprotective effects in both experimental and clinical studies in diabetic and non-diabetic CKD. Unfortunately, fluid retention encountered in large clinical trials in diabetic CKD led to the termination of these studies. Therefore, several advances, including the synthesis of new antagonists with enhanced pharmacological activity, the use of lower doses of ET antagonists, the addition of diuretics, plus simply searching for distinct pathological states to be treated, are promising targets for future experimental studies. In support of these approaches, our group demonstrated in adult subtotally nephrectomized Ren-2 transgenic rats that the addition of a diuretic on top of renin-angiotensin and ET(A) blockade led to a further decrease of proteinuria. This effect was independent of blood pressure which was normalized in all treated groups. Recent data in non-diabetic CKD, therefore, indicate a new potential for ET(A) antagonists, at least under certain pathological conditions.
Collapse
Affiliation(s)
- I Vaněčková
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
111
|
Mao S, Wang L, Chen P, Lan Y, Guo R, Zhang M. Nanoparticle-mediated delivery of Tanshinone IIA reduces adverse cardiac remodeling following myocardial infarctions in a mice model: role of NF-κB pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S707-S716. [PMID: 30284484 DOI: 10.1080/21691401.2018.1508028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Our previous works have shown that tanshinone IIA inhibited maladaptive extracellular matrix remodeling in cardiac fibroblasts implicating its potential role in treating of pathologic cardiac remodeling. However, the intrinsically poor solubility and bioavailability of tanshinone IIA hindered its clinical application. Here we develop monomethoxy-poly (ethylene glycol)-poly (lactic acid)-D-α-Tocopheryl polyethylene glycol 1000 succinate (mPEG-PLA-TPGS) nanoparticle incorporating tanshinone IIA (tanshinone IIA-NPs) and study its efficacy in post-infarction left ventricular (LV) remodeling. Male C57BL/6 mice underwent left coronary artery ligation followed by subsequent intravenously injected tanshinone IIA-NPs therapy for 5 consecutive days. Treatment with tanshinone IIA-NP improved cardiac function, limited infarct expansion, and prevented left ventricle dilation at 4 weeks post-MI. Furthermore, cardiomyocytes inflammation, apoptosis and myocardial fibrosis were significantly attenuated in tanshinone IIA-NP treated mice. These effects also correlated with inhibition of IκB protein phosphorylation and NF-κB activation, leading to suppression of proinflammatory cytokine expression. Together, these results demonstrate tanshinone IIA-NP attenuated adverse cardiac remodeling and dysfunction mediated through prevention of IκB phosphorylation and NF-κB activation. Tanshinone IIA-NP is a novel approach to treat myocardial IR injury in patients with MI.
Collapse
Affiliation(s)
- Shuai Mao
- a Key Discipline of Integrated Chinese and Western Medicine , Second Clinical College, Guangzhou University of Chinese Medicine , Guangzhou , China.,b AMI Key laboratory of Chinese Medicine in Guangzhou , Guangdong Provincial Hospital of Chinese Medicine , Guangzhou , China
| | - Lei Wang
- b AMI Key laboratory of Chinese Medicine in Guangzhou , Guangdong Provincial Hospital of Chinese Medicine , Guangzhou , China
| | - Peipei Chen
- a Key Discipline of Integrated Chinese and Western Medicine , Second Clinical College, Guangzhou University of Chinese Medicine , Guangzhou , China.,b AMI Key laboratory of Chinese Medicine in Guangzhou , Guangdong Provincial Hospital of Chinese Medicine , Guangzhou , China
| | - Yong Lan
- c Beogene Biotech (Guangzhou) CO., LTD , Guangzhou , China
| | - Rui Guo
- d Department of Biomedical Engineering , Jinan University , Guangzhou , China
| | - Minzhou Zhang
- a Key Discipline of Integrated Chinese and Western Medicine , Second Clinical College, Guangzhou University of Chinese Medicine , Guangzhou , China.,b AMI Key laboratory of Chinese Medicine in Guangzhou , Guangdong Provincial Hospital of Chinese Medicine , Guangzhou , China
| |
Collapse
|
112
|
Zitouni H, Ben Ali Gannoum M, Raguema N, Maleh W, Zouari I, Faleh RE, Guibourdenche J, Almawi WY, Mahjoub T. Contribution of angiotensinogen M235T and T174M gene variants and haplotypes to preeclampsia and its severity in (North African) Tunisians. J Renin Angiotensin Aldosterone Syst 2018; 19:1470320317753924. [PMID: 29366364 PMCID: PMC5843851 DOI: 10.1177/1470320317753924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Preeclampsia (PE) is a pregnancy-associated hypertensive disorder and a leading cause of maternal and neonatal morbidity and mortality. While its pathogenesis remains ill defined, several candidate genes for PE have been identified, but results remain inconclusive. We investigated the association of the angiotensinogen ( AGT) gene variants M235T and T174M with PE, and we analyzed the contribution of both variants to the severity of PE. METHODS This case-control study enrolled 550 Tunisian pregnant women: 272 with PE, of whom 147 presented with mild, and 125 with severe PE, along with 278 unrelated age- and ethnically matched control women. AGT genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism. RESULTS Significantly higher M235T minor allele frequency (MAF) was associated with increased risk of PE ( p < 0.001). Decreased frequency of heterozygous T174M genotype carriers were found in control women ( p = 0.015), suggesting a protective effect of this genotype (odds ratio (95% confidence interval) = 0.51 (0.29-0.89)). Two-locus haplotype analysis demonstrated MM and TT haplotypes to be negatively and positively associated with PE, respectively. MAF of M253T, but not T174M, was higher in the severe PE group, and carrying M235T or T174M minor allele was associated with increased body mass index ( p < 0.001) among unselected PE women. CONCLUSIONS AGT M235T and T174M variants contribute to an increased risk of developing PE, and for M235T to PE severity.
Collapse
Affiliation(s)
- Hedia Zitouni
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia.,3 INSERM UMR-S1139 College of Pharmacy, Paris Descartes University, France
| | - Marwa Ben Ali Gannoum
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Nozha Raguema
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia.,2 Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Wided Maleh
- 4 Centre of Maternity and Neonatology, Tunisia
| | - Ines Zouari
- 4 Centre of Maternity and Neonatology, Tunisia
| | | | - Jean Guibourdenche
- 3 INSERM UMR-S1139 College of Pharmacy, Paris Descartes University, France
| | - Wassim Y Almawi
- 5 Faculty of Science of Tunis, University of Tunis El Manar, Tunisia
| | - Touhami Mahjoub
- 1 Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), College of Pharmacy, University of Monastir, Tunisia
| |
Collapse
|
113
|
Peterson JM, Wang DJ, Shettigar V, Roof SR, Canan BD, Bakkar N, Shintaku J, Gu JM, Little SC, Ratnam NM, Londhe P, Lu L, Gaw CE, Petrosino JM, Liyanarachchi S, Wang H, Janssen PML, Davis JP, Ziolo MT, Sharma SM, Guttridge DC. NF-κB inhibition rescues cardiac function by remodeling calcium genes in a Duchenne muscular dystrophy model. Nat Commun 2018; 9:3431. [PMID: 30143619 PMCID: PMC6109146 DOI: 10.1038/s41467-018-05910-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder causing progressive muscle degeneration. Although cardiomyopathy is a leading mortality cause in DMD patients, the mechanisms underlying heart failure are not well understood. Previously, we showed that NF-κB exacerbates DMD skeletal muscle pathology by promoting inflammation and impairing new muscle growth. Here, we show that NF-κB is activated in murine dystrophic (mdx) hearts, and that cardiomyocyte ablation of NF-κB rescues cardiac function. This physiological improvement is associated with a signature of upregulated calcium genes, coinciding with global enrichment of permissive H3K27 acetylation chromatin marks and depletion of the transcriptional repressors CCCTC-binding factor, SIN3 transcription regulator family member A, and histone deacetylase 1. In this respect, in DMD hearts, NF-κB acts differently from its established role as a transcriptional activator, instead promoting global changes in the chromatin landscape to regulate calcium genes and cardiac function.
Collapse
Affiliation(s)
- Jennifer M Peterson
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, NY, 13902, USA
| | - David J Wang
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Vikram Shettigar
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Steve R Roof
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA.,Q Test Labs, Columbus, OH, 43235, USA
| | - Benjamin D Canan
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Nadine Bakkar
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Neurobiology, St Joseph's Hospital and Medical Center-Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| | - Jonathan Shintaku
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Jin-Mo Gu
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Biomedical Engineering and Pediatrics, Emory University, Decatur, GA, 30322, USA
| | - Sean C Little
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA.,Bristol-Myers Squibb, Wallingford, CT, 06492, USA
| | - Nivedita M Ratnam
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Priya Londhe
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Leina Lu
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Christopher E Gaw
- The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jennifer M Petrosino
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Sandya Liyanarachchi
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul M L Janssen
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Jonathan P Davis
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Mark T Ziolo
- Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA.,The Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, Ohio, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Denis C Guttridge
- Department of Cancer Biology and Genetics, Columbus, OH, 43210, USA. .,Center for Muscle Health and Neuromuscular Disorders, Columbus, OH, 43210, USA. .,The Ohio State University Medical Center, Columbus, OH, 43210, USA. .,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.
| |
Collapse
|
114
|
The mechanistic role of oxidative stress in cigarette smoke-induced cardiac stem cell dysfunction and prevention by ascorbic acid. Cell Biol Toxicol 2018; 35:111-127. [DOI: 10.1007/s10565-018-9437-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022]
|
115
|
White Wine Consumption Influences Inflammatory Phase of Repair After Myocardial Infarction in Rats. J Cardiovasc Pharmacol 2018; 70:293-299. [PMID: 28731891 DOI: 10.1097/fjc.0000000000000519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Effects of white wine (WW) consumption on the expression of inflammatory markers/mediators (MMP-2, MMP-9, NF-ĸB p65 and TGF-β1) in myocardial tissue after experimentally induced permanent myocardial ischemia was investigated. Male Sprague-Dawley rats were given either a combination of WW and water or only water, for 28 days. After coronary ligation, animals were left to survive for 24 hours. Three representative areas: infarct/ischemic, peri-infarct/border zone, and control/non-ischemic zones were analyzed for expression of immunoreactivity by measuring the threshold area % of signal density. For MMP-9, significantly smaller expression was found in all 3 zones of wine drinking animals (P < 0.001). There was no difference in MMP-2 immunoreactivity between the 2 groups, except in peri-infarct zones, where the signal was significantly decreased (P < 0.001). The same pattern of expression was found for the NF-κB p65 signal, although no differences between experimental groups were observed for TGF-β1. White wine consumption decreases the expression of the 3 investigated inflammatory markers/mediators in the peri-infarct zone, suggesting its significant modulatory effect. For MMP-9 and MMP-2, expression was similar to the effect of postischemic reperfusion. No effect on TGF-β1 was observed, highlighting its role in being the master-switch, changing from the inflammatory to the proliferative stage of infarct healing.
Collapse
|
116
|
Song X, Su L, Yin H, Dai J, Wei H. Effects of HSYA on the proliferation and apoptosis of MSCs exposed to hypoxic and serum deprivation conditions. Exp Ther Med 2018; 15:5251-5260. [PMID: 29904409 PMCID: PMC5996714 DOI: 10.3892/etm.2018.6125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
As a primary active ingredient of safflor yellow, hydroxysafflor yellow A (HSYA) exhibits notable antioxidative and neuroprotective effects. The aim of the present study was to investigate the protective effects of HSYA in mesenchymal stem cells (MSCs) exposed to hypoxia (5% O2) and serum deprivation (H/SD), and to explore the mechanisms underlying HSYA-mediated protection. Under H/SD conditions, HSYA was applied to protect MSCs against injury. Cell viability, proliferation, apoptosis and reactive oxygen species (ROS) levels were determined using an 5-ethynyl-2′-deoxyuridine assay, MTT assay, Hoechst 33342/propidium iodide and 2′,7′-dichlorodihydrofluorescein diacetate staining, respectively. The results revealed that 160 mg/l HSYA significantly reduced apoptosis and ROS levels compared with the H/SD group; however, HSYA demonstrated minimal effects on cell proliferation. A western blot assay demonstrated that HSYA reduced cleaved caspase-3 expression and cytC release from the mitochondria to the cytoplasm when compared with the H/SD group. In addition, western blotting and RT-qPCR analyses revealed that HSYA treatment significantly increased the expression of hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). In conclusion, the results of the current study demonstrated that HSYA exerts protective effects against H/SD-induced apoptosis in MSCs potentially via activation of the HIF-1α/VEGF signaling pathway and stabilization of the mitochondrial membrane.
Collapse
Affiliation(s)
- Xiaoqing Song
- Biology Office, Basic Medical College of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Lining Su
- Biology Office, Basic Medical College of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Haifeng Yin
- Biology Office, Basic Medical College of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Jin Dai
- Biology Office, Basic Medical College of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Huiping Wei
- Biology Office, Basic Medical College of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
117
|
de Godoy CMG, Vasques ÊR, Caricati-Neto A, Tavares JGP, Alves BJ, Duarte J, Miranda-Ferreira R, Lima MA, Nader HB, Tersariol ILDS. Heparin Oligosaccharides Have Antiarrhythmic Effect by Accelerating the Sodium-Calcium Exchanger. Front Cardiovasc Med 2018; 5:67. [PMID: 29930947 PMCID: PMC5999778 DOI: 10.3389/fcvm.2018.00067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/17/2018] [Indexed: 01/15/2023] Open
Abstract
Background: Blockage of the Na+/Ca2+ exchanger (NCX) is used to determine the role of NCX in arrhythmogenesis. Trisulfated heparin disaccharide (TD) and Low Molecular Weight Heparins (LMWHs) can directly interact with the NCX and accelerate its activity. Objective: In this work, we investigated the antiarrhythmic effect of heparin oligosaccharides related to the NCX activity. Methods: The effects of heparin oligosaccharides were tested on the NCX current (patch clamping) and intracellular calcium transient in rat cardiomyocytes. The effects of heparin oligosaccharides were further investigated in arrhythmia induced in isolated rat atria and rats in vivo. Results: The intracellular Ca2+ concentration decreases upon treatment with either enoxaparin or ardeparin. These drugs abolished arrhythmia induction in isolated atria. The NCX antagonist KB-R7943 abolished the enoxaparin or ardeparin antiarrhythmic effects in isolated atria. In the in vivo measurements, injection of TD 15 min both before coronary occlusion or immediately after reperfusion, significantly prevented the occurrence of reperfusion-induced arrhythmias (ventricular arrhythmia and total AV block) and reduced the lethality rate. The patch clamping experiments showed that, mechanistically, TD increases the forward mode NCX current. Conclusion: Together, the data shows that heparin oligosaccharides may constitute a new class of antiarrhythmic drug that acts by accelerating the forward mode NCX under calcium overload.
Collapse
Affiliation(s)
- Carlos M G de Godoy
- Institute of Science and Technology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ênio R Vasques
- Department of Gastroenterology (LIM 37), Medical School, University of São Paulo, São Paulo, Brazil.,Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
| | - Afonso Caricati-Neto
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José G P Tavares
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz J Alves
- Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana Duarte
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
| | | | - Marcelo A Lima
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Helena B Nader
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ivarne L Dos Santos Tersariol
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil.,Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
| |
Collapse
|
118
|
Farghaly HSM, Ashry IESM, Hareedy MS. High doses of digoxin increase the myocardial nuclear factor-kB and CaV1.2 channels in healthy mice. A possible mechanism of digitalis toxicity. Biomed Pharmacother 2018; 105:533-539. [PMID: 29885637 DOI: 10.1016/j.biopha.2018.05.137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Toxic effects of digoxin may occur with normal therapeutic serum level. However, the underlying mechanisms are not fully understood. Nuclear factor kappa-B (NF-kB) is an important transcription factor in most organ systems and is often implicated in the harmful effects of cardiac injury. NF-kB promotes inflammatory responses, mediates adverse cardiac remodeling and has a function correlation with calcium. The voltage-gated L-type calcium channel CaV1.2 mediates the influx of Ca+2 into the cell in response to membrane depolarization. Our aim was to characterize the role of NF-kB during digoxin toxicity and to assess its correlation with Cav 1.2 in healthy mice in vivo. METHODS To address these questions, digoxin was administered in doses of 0.1, 1 or 5 mg/kg orally daily for seven days to the animals. Serum digoxin, serum calcium, atrial and ventricular calcium levels were measured. We, also, looked for NF-kB and CaV1.2 channel expression in cardiac muscle of mice. RESULTS Digoxin at a dose of 0.1 mg/kg did not enhance serum, atrial, and ventricular Ca+2 levels, but were increased when digoxin dose of 1 and 5 mg/kg were administered. Histologically, myocardial necrosis and cellular infiltration on day 7 were significantly more severe in the 5 mg/kg/day digoxin group. Immunohistochemical studies showed more expression of both NF-kB and CaV1.2 in 1 and 5 mg/kg/day digoxin groups. CONCLUSIONS These data suggest that NF-kB may be responsible for digoxin toxicity, at least partially via modulation of CaV1.2 and intracellular calcium homeostasis in the myocardium.
Collapse
|
119
|
Sydykov A, Mamazhakypov A, Petrovic A, Kosanovic D, Sarybaev AS, Weissmann N, Ghofrani HA, Schermuly RT. Inflammatory Mediators Drive Adverse Right Ventricular Remodeling and Dysfunction and Serve as Potential Biomarkers. Front Physiol 2018; 9:609. [PMID: 29875701 PMCID: PMC5974151 DOI: 10.3389/fphys.2018.00609] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/04/2018] [Indexed: 01/07/2023] Open
Abstract
Adverse right ventricular (RV) remodeling leads to ventricular dysfunction and failure that represents an important determinant of outcome in patients with pulmonary hypertension (PH). Recent evidence indicates that inflammatory activation contributes to the pathogenesis of adverse RV remodeling and dysfunction. It has been shown that accumulation of inflammatory cells such as macrophages and mast cells in the right ventricle is associated with maladaptive RV remodeling. In addition, inhibition of inflammation in animal models of RV failure ameliorated RV structural and functional impairment. Furthermore, a number of circulating inflammatory mediators have been demonstrated to be associated with RV performance. This work reviews the role of inflammation in RV remodeling and dysfunction and discusses anti-inflammatory strategies that may attenuate adverse structural alterations while promoting improvement of RV function.
Collapse
Affiliation(s)
- Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany.,Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Argen Mamazhakypov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Aleksandar Petrovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
120
|
Over-expression of growth differentiation factor 15 (GDF15) preventing cold ischemia reperfusion (I/R) injury in heart transplantation through Foxo3a signaling. Oncotarget 2018; 8:36531-36544. [PMID: 28388574 PMCID: PMC5482674 DOI: 10.18632/oncotarget.16607] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/02/2017] [Indexed: 12/21/2022] Open
Abstract
Ischemia reperfusion (I/R) injury which inevitably occurs during heart transplantation is the major factor leading to organ failure and graft rejection. In order to develop new therapies to prevent I/R injury, we used both a murine heart transplantation model with 24 hour cold I/R and an in vitro cell culture system to determine whether growth differentiation factor 15 (GDF15) is a protective factor in preventing I/R injury in heart transplantation and to further investigate underlying mechanisms of I/R injury. We found that cold I/R caused severe damage to the endocardium, epicardium and myocardium of heart grafts from wild type C57BL/6 mice, whereas grafts from GDF15 transgenic (TG) mice showed less damage as demonstrated by decreased cell apoptosis/death, decreased neutrophils infiltration and the preservation of the normal structure of the heart. Over-expression of GDF15 reduced expression of phosphorylated RelA p65, pre-inflammatory and pro-apoptotic genes while it enhanced Foxo3a phosphorylation in vitro and in vivo. Over-expression of GDF15 inhibited cell apoptosis/death and reduced neutrophil infiltration. In conclusion, this study, for the first time, demonstrates that GDF15 is a promising target for preventing cold I/R injury in heart transplantation. This study also shows that the resultant protective effects are mediated by the Foxo3 and NFκB signaling pathways.
Collapse
|
121
|
Zhang Q, Liu S, Li T, Yuan L, Liu H, Wang X, Wang F, Wang S, Hao A, Liu D, Wang Z. Preconditioning of bone marrow mesenchymal stem cells with hydrogen sulfide improves their therapeutic potential. Oncotarget 2018; 7:58089-58104. [PMID: 27517324 PMCID: PMC5295414 DOI: 10.18632/oncotarget.11166] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/27/2016] [Indexed: 12/15/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) transplantation has shown great promises for treating various brain diseases. However, poor viability of transplanted BMSCs in injured brain has limited the therapeutic efficiency. Hypoxia-ischemic injury is one of major mechanisms underlying the survival of transplanted BMSCs. We investigated the mechanism of preconditioning of BMSCs with hydrogen sulfide (H2S), which has been proposed as a novel therapeutic strategy for hypoxia-ischemic injury. In this study, we demonstrated that preconditioning of NaHS, a H2S donor, effectively suppressed hypoxia-ischemic-induced apoptosis whereby the rise in Bax/Bcl-2 ratio. Further analyses revealed Akt and ERK1/2 pathways were involved in the protective effects of NaHS. In addition, NaHS preconditioning increased secretion of BDNF and VEGF in BMSCs. Consistent with in vitro data, transplantation of NaHS preconditioned BMSCs in vivo further enhanced the therapeutic effects of BMSCs on neuronal injury and neurological recovery, associated with increased vessel density and upregulation of BDNF and VEGF in the ischemic tissue. These findings suggest that H2S could enhance the therapeutic effects of BMSCs. The underlying mechanisms might be due to enhanced capacity of BMSCs and upregulation of protective cytokines in the hypoxia tissue.
Collapse
Affiliation(s)
- Qun Zhang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Song Liu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Tong Li
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Lin Yuan
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Hansen Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Xueer Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Fuwu Wang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Shuanglian Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Aijun Hao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Dexiang Liu
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Zhen Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China.,Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
122
|
Wongkhum C, Chotigeat W, Kedjarune-Leggat U. Effect of recombinant vascular endothelial growth factor and translationally controlled tumor protein on 2‑hydroxyethyl methacrylate‑treated pulp cells. Mol Med Rep 2018; 17:6100-6108. [PMID: 29436669 DOI: 10.3892/mmr.2018.8593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/06/2018] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)-A is a potential signaling protein that may promote angiogenesis. VEGF also helps cells survive in stressfull or hazardous conditions. The present study aimed to compare the effect of VEGF with translationally controlled tumor protein (TCTP), an anti‑apoptotic protein in human dental pulp cells (HDPCs), following exposure to 2‑hydroxyethyl methacrylate (HEMA), which is a major residual monomer from resin restorative dental materials. Cell viability, alkaline phosphatase (ALP) activity, mineralization and gene expressions for odontogenic and osteogenic differentiation markers of HDPCs were investigated, following exposure to HEMA and in combination with TCTP and VEGF. The results revealed that TCTP at 1 ng/ml and VEGF at 10 ng/ml significantly promoted the proliferation of HDPCs (P<0.05). TCTP (1 ng/ml) and VEGF (10 ng/ml) maintained the cell viability of 4 mM HEMA‑treated cells at the same percentage as the control. However, cells treated with HEMA+TCTP+VEGF had a lower cell viability than the groups treated with HEMA and TCTP or VEGF alone. TCTP and VEGF promoted cell proliferation, ALP activity and mineralization, and upregulated of DSPP, DMP‑1, BMP‑2, and ALP mRNA expression compared with the control. Furthermore, the HEMA+TCTP and HEMA+VEGF groups had significantly higher percentages of calcium deposition than HEMA‑treated cells (P<0.001). HEMA was cytotoxic to HDPCs, reduced ALP activity and caused the significant downregulation of odontogenic and osteogenic gene expressions (P<0.05). It was concluded that VEGF and TCTP promoted pulp cell growth and the survival of HEMA‑treated cells without synergistic effects. TCTP was required in lower concentrations for these effects. VEGF and TCTP enhanced cell differentiation and mineralization.
Collapse
Affiliation(s)
- Chunyanut Wongkhum
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Wilaiwan Chotigeat
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Ureporn Kedjarune-Leggat
- Department of Oral Biology and Occlusion, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| |
Collapse
|
123
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
124
|
Wang L, Quan Y, Yue Y, Heng X, Che F. Interleukin-37: A crucial cytokine with multiple roles in disease and potentially clinical therapy. Oncol Lett 2018; 15:4711-4719. [PMID: 29552110 DOI: 10.3892/ol.2018.7982] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Interleukin (IL)-37, a new IL-1 family member, has received increasing attention in recent years. In the past decade, it has been determined that IL-37 is expressed in various normal cells and tissues and is regulated by inflammatory stimuli and pro-cytokines via different signal transduction pathways. Recently, it has been found that IL-37 is expressed in a variety of cancers, chronic inflammatory and autoimmune disorders, and exerts anti-inflammatory effects. Moreover, a growing body of literature demonstrates that IL-37 plays a vital role in inhibiting both innate and adaptive immune responses as well as inflammatory reactions. In addition, IL-37 may prove to be a new and potentially useful target for effective cytokine therapy. Further evidence is needed to clarify in more detail the effects of IL-37 in experimental and clinical studies. Based on an extensive summary of published data, the aim of this review is to outline the current knowledge of IL-37, including the location, structure, expression, regulation and function, as well as the potential clinical applications of this cytokine.
Collapse
Affiliation(s)
- Lijuan Wang
- Central Laboratory, Hematology Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276000, P.R. China.,Department of Hematology, Hematology Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276000, P.R. China
| | - Yanchun Quan
- Central Laboratory, Hematology Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276000, P.R. China
| | - Yongfang Yue
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xueyuan Heng
- Department of Neurosurgery, Linyi People's Hospital, Shandong University, Linyi, Shandong 276000, P.R. China
| | - Fengyuan Che
- Central Laboratory, Hematology Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
125
|
Bcl-2-associated athanogene 3 (BAG3) is an enhancer of small heat shock protein turnover via activation of autophagy in the heart. Biochem Biophys Res Commun 2018; 496:1141-1147. [DOI: 10.1016/j.bbrc.2018.01.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 01/25/2018] [Indexed: 11/17/2022]
|
126
|
Guarini G, Huqi A, Morrone D, Capozza PFG, Marzilli M. Trimetazidine and Other Metabolic Modifiers. Eur Cardiol 2018; 13:104-111. [PMID: 30697354 DOI: 10.15420/ecr.2018.15.2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Treatment goals for people with chronic angina should focus on the relief of symptoms and improving mortality rates so the patient can feel better and live longer. The traditional haemodynamic approach to ischaemic heart disease was based on the assumption that increasing oxygen supply and decreasing oxygen demand would improve symptoms. However, data from clinical trials, show that about one third of people continue to have angina despite a successful percutaneous coronary intervention and medical therapy. Moreover, several trials on chronic stable angina therapy and revascularisation have failed to show benefits in terms of primary outcome (survival, cardiovascular death, all-cause mortality), symptom relief or echocardiographic parameters. Failure to significantly improve quality of life and prognosis may be attributed in part to a limited understanding of ischaemic heart disease, by neglecting the fact that ischaemia is a metabolic disorder. Shifting cardiac metabolism from free fatty acids towards glucose is a promising approach for the treatment of patients with stable angina, independent of the underlying disease (macrovascular and/or microvascular disease). Cardiac metabolic modulators open the way to a greater understanding of ischaemic heart disease and its common clinical manifestations as an energetic disorder rather than an imbalance between the demand and supply of oxygen and metabolites.
Collapse
Affiliation(s)
- Giacinta Guarini
- Cardiovascular Medicine Division, Cardiothoracic and Vascular Department, University of Pisa Italy
| | - Alda Huqi
- Cardiovascular Medicine Division, Ospedale della Versilia, Lido di Camaiore Italy
| | - Doralisa Morrone
- Cardiovascular Medicine Division, Cardiothoracic and Vascular Department, University of Pisa Italy
| | | | - Mario Marzilli
- Cardiovascular Medicine Division, Cardiothoracic and Vascular Department, University of Pisa Italy
| |
Collapse
|
127
|
Deng M, Xiao H, Peng H, Yuan H, Xu Y, Zhang G, Tang J, Hu Z. Preservation of neuronal functions by exosomes derived from different human neural cell types under ischemic conditions. Eur J Neurosci 2017; 47:150-157. [PMID: 29178548 DOI: 10.1111/ejn.13784] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapies have been reported in protecting cerebral infarction-induced neuronal dysfunction and death. However, most studies used rat/mouse neuron as model cell when treated with stem cell or exosomes. Whether these findings can be translated from rodent to humans has been in doubt. Here, we used human embryonic stem cell-derived neurons to detect the protective potential of exosomes against ischemia. Neurons were treated with in vitro oxygen-glucose deprivation (OGD) for 1 h. For treatment group, different exosomes were derived from neuron, embryonic stem cell, neural progenitor cell and astrocyte differentiated from H9 human embryonic stem cell and added to culture medium 30 min after OGD (100 μg/mL). Western blotting was performed 12 h after OGD, while cell counting and electrophysiological recording were performed 48 h after OGD. We found that these exosomes attenuated OGD-induced neuronal death, Mammalian target of rapamycin (mTOR), pro-inflammatory and apoptotic signaling pathway changes, as well as basal spontaneous synaptic transmission inhibition in varying degrees. The results implicate the protective effect of exosomes on OGD-induced neuronal death and dysfunction in human embryonic stem cell-derived neurons, potentially through their modulation on mTOR, pro-inflammatory and apoptotic signaling pathways.
Collapse
Affiliation(s)
- Mingyang Deng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Han Xiao
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huan Yuan
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunxiao Xu
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guangsen Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianguang Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
128
|
Gong W, Qie S, Huang P, Xi J. Protective Effect of miR-374a on Chemical Hypoxia-Induced Damage of PC12 Cells In Vitro via the GADD45α/JNK Signaling Pathway. Neurochem Res 2017; 43:581-590. [PMID: 29247275 DOI: 10.1007/s11064-017-2452-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 12/05/2017] [Accepted: 12/09/2017] [Indexed: 12/31/2022]
Abstract
To explore the effect of microRNA-374a (miR-374a) on chemical hypoxia-induced pheochromocytoma (PC12) cell damage by mediating growth arrest and the DNA damage-45 alpha (GADD45α)/c-Jun N-terminal kinase (JNK) signaling pathway. PC12 cells were divided into a Control group (no treatment), Model group (treated with CoCl2 for 24 h), negative control (NC) group (transfected with miR-374a negative control sequence and treated with CoCl2 for 24 h), and miR-374a mimic group (transfected with miR-374a mimics and treated with CoCl2 for 24 h). The viability and apoptosis of PC12 cells were determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and flow cytometry, while the mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) content were assessed by Rh123 and dichloro-dihydro-fluorescein diacetate (DCFH-DA) methods. The expression of miR-374a and GADD45α/JNK proteins was detected using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. A significant decrease was found in the survival rate, MMP and miR-374a expression, while an increase was shown in the ROS content and GADD45α and p-JNK expression in hypoxic PC12 cells (all P < 0.05). A luciferase reporter gene assay demonstrated that GADD45α is the target gene of miR-374a. When transfected with miR-374a mimics, hypoxic PC12 cells showed an obvious elevation in survival rate and MMP but a great reduction in cell apoptosis and ROS content, as well as in the expression of GADD45α and p-JNK proteins (all P < 0.05). MiR-374a can protect PC12 cells against hypoxia-induced injury by inhibiting the GADD45α/JNK pathway, enhancing cell viability, suppressing oxidative stress, and inhibiting cell apoptosis, thereby becoming a potential therapeutic target for hypoxic damage.
Collapse
Affiliation(s)
- Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Shuyan Qie
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Peiling Huang
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, 100069, China
| | - Jianing Xi
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Xixiazhuang, Badachu Road, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
129
|
Late sodium current associated cardiac electrophysiological and mechanical dysfunction. Pflugers Arch 2017; 470:461-469. [DOI: 10.1007/s00424-017-2079-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
130
|
Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac remodelling. Cardiovasc Res 2017; 113:1538-1550. [PMID: 29088374 DOI: 10.1093/cvr/cvx130] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 01/03/2025] Open
Abstract
Canonical innate immune signalling involves complex cascades: multiple germline-encoded pattern recognition receptors rapidly recognize pathogen-associated or damage-associated molecular patterns to induce the production of cytokines, which bind to their corresponding receptors to orchestrate subsequent host defense phases. Inflammation is a healthy response to pathogenic signals, which are typically rapid and specific, and they terminate once the threat has passed. However, excessive activation or suppression of innate immune or inflammatory responses can lead to considerable human suffering, such as cardiac remodelling. Interestingly, recent studies have revealed that innate immune molecules in the parenchymal cells of the heart influence cardiac homeostasis not only by directly regulating innate immune responses but also through reprogrammed signalling pathways, which are independent of conventional innate immune signalling. Elucidating 'innate immune signalling reprogramming' events will help us better understand the functions of innate immune molecules and, moreover, the pathogenesis of cardiac diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| | - Zan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- College of Life Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| |
Collapse
|
131
|
Falik-Zaccai TC, Barsheshet Y, Mandel H, Segev M, Lorber A, Gelberg S, Kalfon L, Ben Haroush S, Shalata A, Gelernter-Yaniv L, Chaim S, Raviv Shay D, Khayat M, Werbner M, Levi I, Shoval Y, Tal G, Shalev S, Reuveni E, Avitan-Hersh E, Vlodavsky E, Appl-Sarid L, Goldsher D, Bergman R, Segal Z, Bitterman-Deutsch O, Avni O. Sequence variation in PPP1R13L results in a novel form of cardio-cutaneous syndrome. EMBO Mol Med 2017; 9:319-336. [PMID: 28069640 PMCID: PMC5331242 DOI: 10.15252/emmm.201606523] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a life-threatening disorder whose genetic basis is heterogeneous and mostly unknown. Five Arab Christian infants, aged 4-30 months from four families, were diagnosed with DCM associated with mild skin, teeth, and hair abnormalities. All passed away before age 3. A homozygous sequence variation creating a premature stop codon at PPP1R13L encoding the iASPP protein was identified in three infants and in the mother of the other two. Patients' fibroblasts and PPP1R13L-knocked down human fibroblasts presented higher expression levels of pro-inflammatory cytokine genes in response to lipopolysaccharide, as well as Ppp1r13l-knocked down murine cardiomyocytes and hearts of Ppp1r13l-deficient mice. The hypersensitivity to lipopolysaccharide was NF-κB-dependent, and its inducible binding activity to promoters of pro-inflammatory cytokine genes was elevated in patients' fibroblasts. RNA sequencing of Ppp1r13l-knocked down murine cardiomyocytes and of hearts derived from different stages of DCM development in Ppp1r13l-deficient mice revealed the crucial role of iASPP in dampening cardiac inflammatory response. Our results determined PPP1R13L as the gene underlying a novel autosomal-recessive cardio-cutaneous syndrome in humans and strongly suggest that the fatal DCM during infancy is a consequence of failure to regulate transcriptional pathways necessary for tuning cardiac threshold response to common inflammatory stressors.
Collapse
Affiliation(s)
- Tzipora C Falik-Zaccai
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel .,Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Yiftah Barsheshet
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Hanna Mandel
- Metabolic Disease Unit, Rambam Health Care Campus, Haifa, Israel.,Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Meital Segev
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Avraham Lorber
- Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Pediatric Cardiology, Rambam Health Care Campus, Haifa, Israel
| | - Shachaf Gelberg
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Limor Kalfon
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Shani Ben Haroush
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Adel Shalata
- The Winter Genetic Institute, Bnei Zion Medical Center, Haifa, Israel
| | | | - Sarah Chaim
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Dorith Raviv Shay
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Morad Khayat
- The Genetic Institute, Ha'emek Medical Center, Afula, Israel
| | - Michal Werbner
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Inbar Levi
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Yishay Shoval
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Galit Tal
- Metabolic Disease Unit, Rambam Health Care Campus, Haifa, Israel.,Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Stavit Shalev
- Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,The Genetic Institute, Ha'emek Medical Center, Afula, Israel
| | - Eli Reuveni
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | | | - Eugene Vlodavsky
- Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Liat Appl-Sarid
- Department of Pathology, Galilee Medical Center, Nahariya, Israel
| | - Dorit Goldsher
- Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Diagnostic Imaging, Rambam Health Care Campus, Haifa, Israel
| | - Reuven Bergman
- Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Dermatology, Rambam Health Care Campus, Haifa, Israel
| | - Zvi Segal
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel.,Department of Ophthalmology, Galilee Medical Center, Nahariya, Israel
| | - Ora Bitterman-Deutsch
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel.,Dermatology Clinic, Galilee Medical Center, Nahariya, Israel
| | - Orly Avni
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| |
Collapse
|
132
|
Tavakoli R, Nemska S, Jamshidi P, Gassmann M, Frossard N. Technique of Minimally Invasive Transverse Aortic Constriction in Mice for Induction of Left Ventricular Hypertrophy. J Vis Exp 2017:56231. [PMID: 28994784 PMCID: PMC5752328 DOI: 10.3791/56231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Transverse aortic constriction (TAC) in mice is one of the most commonly used surgical techniques for experimental investigation of pressure overload-induced left ventricular hypertrophy (LVH) and its progression to heart failure. In the majority of the reported investigations, this procedure is performed with intubation and ventilation of the animal which renders it demanding and time-consuming and adds to the surgical burden to the animal. The aim of this protocol is to describe a simplified technique of minimally invasive TAC without intubation and ventilation of mice. Critical steps of the technique are emphasized in order to achieve low mortality and high efficiency in inducing LVH. Male C57BL/6 mice (10-week-old, 25-30 g, n=60) were anesthetized with a single intraperitoneal injection of a mixture of ketamine and xylazine. In a spontaneously breathing animal following a 3-4 mm upper partial sternotomy, a segment of 6/0 silk suture threaded through the eye of a ligation aid was passed under the aortic arch and tied over a blunted 27-gauge needle. Sham-operated animals underwent the same surgical preparation but without aortic constriction. The efficacy of the procedure in inducing LVH is attested by a significant increase in the heart/body weight ratio. This ratio is obtained at days 3, 7, 14 and 28 after surgery (n = 6 - 10 in each group and each time point). Using our technique, LVH is observed in TAC compared to sham animals from day 7 through day 28. Operative and late (over 28 days) mortalities are both very low at 1.7%. In conclusion, our cost-effective technique of minimally invasive TAC in mice carries very low operative and post-operative mortalities and is highly efficient in inducing LVH. It simplifies the operative procedure and reduces the strain put on the animal. It can be easily performed by following the critical steps described in this protocol.
Collapse
Affiliation(s)
- Reza Tavakoli
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich;
| | - Simona Nemska
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich; Laboratoire d'Innovation Thérapeutique, UMR7200, Université de Strasbourg - CNRS
| | | | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, UMR7200, Université de Strasbourg - CNRS
| |
Collapse
|
133
|
Luan Y, Zhang X, Zhang Y, Dong Y. MicroRNA-210 Protects PC-12 Cells Against Hypoxia-Induced Injury by Targeting BNIP3. Front Cell Neurosci 2017; 11:285. [PMID: 29018330 PMCID: PMC5614931 DOI: 10.3389/fncel.2017.00285] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/01/2017] [Indexed: 11/30/2022] Open
Abstract
MicroRNA (miR)-210 is the most consistently and predominantly up-regulated miR in response to hypoxia in multiple cancer cells. The roles of miR-210 in rat adrenal gland pheochromocytoma (PC-12) cells remain unknown. We aimed to explore the possible effect of miR-210 in neonatal brain injury. We explored the potential molecular mechanism by using PC-12 cells under hypoxia. Scramble miRs, miR-210 mimic, miR-210 inhibitor or its negative control were respectively transfected into PC-12 cells. Cell viability, migration, invasion and apoptosis were also assessed to evaluate hypoxia-induced cell injury. The expression level of miR-210 was identified by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. Apoptosis-related protein expression as well as key kinases in the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signal pathway was studied by Western blot analysis. Hypoxia suppressed cell viability, migration and invasion, but promoted apoptosis through activation of mitochondrial- and caspase-dependent pathways. Hypoxia markedly induced up-regulation of miR-210 in PC-12 cells. Overexpression of miR-210 protected PC-12 cells against hypoxia-induced injury. Bcl-2 adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) was proven to be a target gene of miR-210 in PC-12 cells. miR-210 overexpression ameliorated the hypoxia-induced injury in PC-12 cells by down-regulating BNIP3. Hypoxia-induced alterations of key kinases in the PI3K/AKT/mTOR signal pathway were affected by aberrant expression of BNIP3. These findings suggested that miR-210 protected PC-12 cells against hypoxia-induced injury by targeting BNIP3, involving the PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yonggang Luan
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Xiaoli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yongli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yubin Dong
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| |
Collapse
|
134
|
Zhou J, Li J, Rosenbaum DM, Zhuang J, Poon C, Qin P, Rivera K, Lepore J, Willette RN, Hu E, Barone FC. The prolyl 4-hydroxylase inhibitor GSK360A decreases post-stroke brain injury and sensory, motor, and cognitive behavioral deficits. PLoS One 2017; 12:e0184049. [PMID: 28880966 PMCID: PMC5589177 DOI: 10.1371/journal.pone.0184049] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/17/2017] [Indexed: 12/20/2022] Open
Abstract
There is interest in pharmacologic preconditioning for end-organ protection by targeting the HIF system. This can be accomplished by inhibition of prolyl 4-hydroxylase (PHD). GSK360A is an orally active PHD inhibitor that has been previously shown to protect the failing heart. We hypothesized that PHD inhibition can also protect the brain from injuries and resulting behavioral deficits that can occur as a result of surgery. Thus, our goal was to investigate the effect of pre-stroke surgery brain protection using a verified GSK360A PHD inhibition paradigm on post-stroke surgery outcomes. Vehicle or an established protective dose (30 mg/kg, p.o.) of GSK360A was administered to male Sprague-Dawley rats. Initially, GSK360A pharmacokinetics and organ distribution were determined, and then PHD-HIF pharmacodynamic markers were measured (i.e., to validate the pharmacological effects of the GSK360A administration regimen). Results obtained using this validated PHD dose-regimen indicated significant improvement by GSK360A (30mg/kg); administered at 18 and 5 hours prior to transient middle cerebral artery occlusion (stroke). GSK360A exposure and plasma, kidney and brain HIF-PHD pharmacodynamics endpoints (e.g., erythropoietin; EPO and Vascular Endothelial Growth Factor; VEGF) were measured. GSK360A provided rapid exposure in plasma (7734 ng/ml), kidney (45–52% of plasma level) and brain (1–4% of plasma level), and increased kidney EPO mRNA (80-fold) and brain VEGF mRNA (2-fold). We also observed that GSK360A increased plasma EPO (300-fold) and VEGF (2-fold). Further assessments indicated that GSK360A reduced post-stroke surgery neurological deficits (47–64%), cognitive dysfunction (60–75%) and brain infarction (30%) 4 weeks later. Thus, PHD inhibition using GSK360A pretreatment produced long-term post-stroke brain protection and improved behavioral functioning. These data support PHD inhibition, specifically by GSK360A, as a potential strategy for pre-surgical use to reduce brain injury and functional decline due to surgery-related cerebral injury.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/metabolism
- Brain/pathology
- Brain Injuries/blood
- Brain Injuries/drug therapy
- Brain Injuries/etiology
- Brain Injuries/physiopathology
- Cognition Disorders/drug therapy
- Cognition Disorders/etiology
- Erythropoietin/blood
- Erythropoietin/genetics
- Glycine/administration & dosage
- Glycine/analogs & derivatives
- Glycine/pharmacokinetics
- Glycine/pharmacology
- Glycine/therapeutic use
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Infarction, Middle Cerebral Artery/blood
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Male
- Motor Activity/drug effects
- Organ Specificity/drug effects
- Prolyl Hydroxylases/metabolism
- Prolyl-Hydroxylase Inhibitors/administration & dosage
- Prolyl-Hydroxylase Inhibitors/pharmacology
- Prolyl-Hydroxylase Inhibitors/therapeutic use
- Quinolones/administration & dosage
- Quinolones/pharmacokinetics
- Quinolones/pharmacology
- Quinolones/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Sensation/drug effects
- Stroke/blood
- Stroke/complications
- Stroke/physiopathology
- Vascular Endothelial Growth Factor A/blood
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Jin Zhou
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Jie Li
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Daniel M. Rosenbaum
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
- Robert F. Furchgott Foundation, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Jian Zhuang
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Carrie Poon
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
| | - Pu Qin
- Cardiac Biology, Heart Failure Discovery Performance Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania, United States of America
| | - Katrina Rivera
- Cardiac Biology, Heart Failure Discovery Performance Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania, United States of America
| | - John Lepore
- Cardiac Biology, Heart Failure Discovery Performance Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania, United States of America
| | - Robert N. Willette
- Cardiac Biology, Heart Failure Discovery Performance Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania, United States of America
| | - Erding Hu
- Cardiac Biology, Heart Failure Discovery Performance Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania, United States of America
| | - Frank C. Barone
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
- Robert F. Furchgott Foundation, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, New York, United States of America
- * E-mail:
| |
Collapse
|
135
|
Fung G, Wong J, Berhe F, Mohamud Y, Xue YC, Luo H. Phosphorylation and degradation of αB-crystallin during enterovirus infection facilitates viral replication and induces viral pathogenesis. Oncotarget 2017; 8:74767-74780. [PMID: 29088822 PMCID: PMC5650377 DOI: 10.18632/oncotarget.20366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/25/2017] [Indexed: 01/25/2023] Open
Abstract
Protein quality control (PQC) plays a key role in maintaining cardiomyocyte function and homeostasis, and malfunction in PQC is implicated in various forms of heart diseases. Molecular chaperones serve as the primary checkpoint for PQC; however, their roles in the pathogenesis of viral myocarditis, an inflammation of the myocardium caused by viral infection, are largely unknown. AlphaB-crystallin (CryAB) is the most abundant chaperone protein in the heart. It interacts with desmin and cytoplasmic actin to prevent protein misfolding and aggregation and to help maintain cytoskeletal integrity and cardiac function. Here we showed that coxsackievirus infection induced desminopathy-like phenotype of the myocardium, as characterized by the accumulation of protein aggregates and the disruption of desmin organization. We further demonstrated that CryAB was phosphorylated during early and downregulated at later stages of infection. Moreover, we showed that phosphorylated CryAB had a shorter half-life and was targeted to the ubiquitin-proteasome system for degradation. Lastly, we found that overexpression of CryAB significantly attenuated viral protein production and progeny release, indicating an anti-viral function for CryAB. Together, our results suggest a mechanism by which coxsackieviral infection induces CryAB degradation and loss-of-function, resulting in desmin aggregation, ultimately contributing to compromised cytoskeletal integrity and viral cardiomyopathy.
Collapse
Affiliation(s)
- Gabriel Fung
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jerry Wong
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Feaven Berhe
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuan Chao Xue
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
136
|
Transcriptional downregulation of microRNA-19a by ROS production and NF-κB deactivation governs resistance to oxidative stress-initiated apoptosis. Oncotarget 2017; 8:70967-70981. [PMID: 29050336 PMCID: PMC5642611 DOI: 10.18632/oncotarget.20235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022] Open
Abstract
Cell apoptosis is one of the main pathological alterations during oxidative stress (OS) injury. Previously, we corroborated that nuclear factor-κB (NF-κB) transactivation confers apoptosis resistance against OS in mammalian cells, yet the underlying mechanisms remain enigmatic. Here we report that microRNA-19a (miR-19a) transcriptionally regulated by reactive oxygen species (ROS) production and NF-κB deactivation prevents OS-initiated cell apoptosis through cylindromatosis (CYLD) repression. CYLD contributes to OS-initiated cell apoptosis, for which NF-κB deactivation is essential. MiR-19a directly represses CYLD via targeting 3′ UTR of CYLD, thereby antagonizing OS-initiated apoptosis. CYLD repression by miR-19a restores the IKKβ phosphorylation, RelA disassociation from IκBα, IκBα polyubiquitination and degradation, RelA recruitment at VEGF gene promoter as well as VEGF secretion in the context of OS. Either pharmacological deactivation of NF-κB or genetic upregulation of CYLD compromises the apoptosis-resistant phenotypes of miR-19a. Furthermore, miR-19a is transcriptionally downregulated upon OS in two distinct processes that require ROS production and NF-κB deactivation. VEGF potentiates the ability of miR-19a to activate NF-κB and render apoptosis resistance. Our findings underscore a putative mechanism whereby CYLD repression-mediated and NF-κB transactivation-dependent miR-19a regulatory feedback loop prevents cell apoptosis in response to OS microenvironment.
Collapse
|
137
|
Kaplan A, Abidi E, Ghali R, Booz GW, Kobeissy F, Zouein FA. Functional, Cellular, and Molecular Remodeling of the Heart under Influence of Oxidative Cigarette Tobacco Smoke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3759186. [PMID: 28808498 PMCID: PMC5541812 DOI: 10.1155/2017/3759186] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/01/2017] [Indexed: 01/05/2023]
Abstract
Passive and active chronic cigarette smoking (CS) remains an international epidemic and a key risk factor for cardiovascular disease (CVD) development. CS-induced cardiac damage is divided into two major and interchangeable mechanisms: (1) direct adverse effects on the myocardium causing smoking cardiomyopathy and (2) indirect effects on the myocardium by fueling comorbidities such as atherosclerotic syndromes and hypertension that eventually damage and remodel the heart. To date, our understanding of cardiac remodeling following acute and chronic smoking exposure is not well elucidated. This manuscript presents for the first time the RIMD (oxidative stress (R), inflammation (I), metabolic impairment (M), and cell death (D)) detrimental cycle concept as a major player in CS-induced CVD risks and direct cardiac injury. Breakthroughs and latest findings in the field with respect to structural, functional, cellular, and molecular cardiac remodeling following chronic smoking exposure are summarized. This review also touches the genetics/epigenetics of smoking as well as the smoker's paradox and highlights the most currently prominent pharmacological venues to mitigate CS-induced adverse cardiac remodeling.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Emna Abidi
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Rana Ghali
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - George W. Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center School of Medicine, Jackson, MS, USA
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
138
|
Hu X, Van Marion DMS, Wiersma M, Zhang D, Brundel BJJM. The protective role of small heat shock proteins in cardiac diseases: key role in atrial fibrillation. Cell Stress Chaperones 2017; 22:665-674. [PMID: 28484965 PMCID: PMC5465041 DOI: 10.1007/s12192-017-0799-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 02/06/2023] Open
Abstract
Atrial fibrillation (AF) is the most common tachyarrhythmia which is associated with increased morbidity and mortality. AF usually progresses from a self-terminating paroxysmal to persistent disease. It has been recognized that AF progression is driven by structural remodeling of cardiomyocytes, which results in electrical and contractile dysfunction of the atria. We recently showed that structural remodeling is rooted in derailment of proteostasis, i.e., homeostasis of protein production, function, and degradation. Since heat shock proteins (HSPs) play an important role in maintaining a healthy proteostasis, the role of HSPs was investigated in AF. It was found that especially small heat shock protein (HSPB) levels get exhausted in atrial tissue of patients with persistent AF and that genetic or pharmacological induction of HSPB protects against cardiomyocyte remodeling in experimental models for AF. In this review, we provide an overview of HSPBs as a potential therapeutic target for normalizing proteostasis and suppressing the substrates for AF progression in experimental and clinical AF and discuss HSP activators as a promising therapy to prevent AF onset and progression.
Collapse
Affiliation(s)
- Xu Hu
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Denise M S Van Marion
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Deli Zhang
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
139
|
Franssen C, Kole J, Musters R, Hamdani N, Paulus WJ. α-B Crystallin Reverses High Diastolic Stiffness of Failing Human Cardiomyocytes. Circ Heart Fail 2017; 10:e003626. [PMID: 28242778 DOI: 10.1161/circheartfailure.116.003626] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/23/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cardiomyocytes with a less distensible titin and interstitial collagen contribute to the high diastolic stiffness of failing myocardium. Their relative contributions and mechanisms underlying loss of titin distensibility were assessed in failing human hearts. METHODS AND RESULTS Left ventricular tissue was procured in patients with aortic stenosis (AS, n=9) and dilated cardiomyopathy (DCM, n=6). Explanted donor hearts (n=8) served as controls. Stretches were performed in myocardial strips, and an extraction protocol differentiated between passive tension (Fpassive) attributable to cardiomyocytes or to collagen. Fpassive-cardiomyocytes was higher in AS and DCM at shorter muscle lengths, whereas Fpassive-collagen was higher in AS at longer muscle lengths and in DCM at shorter and longer muscle lengths. Cardiomyocytes were stretched to investigate titin distensibility. Cardiomyocytes were incubated with alkaline phosphatase, subsequently reassessed after a period of prestretch and finally treated with the heat shock protein α-B crystallin. Alkaline phosphatase shifted the Fpassive-sarcomere length relation upward only in donor. Prestretch shifted the Fpassive-sarcomere length relation further upward in donor and upward in AS and DCM. α-B crystallin shifted the Fpassive-sarcomere length relation downward to baseline in donor and to lower than baseline in AS and DCM. In failing myocardium, confocal laser microscopy revealed α-B crystallin in subsarcolemmal aggresomes. CONCLUSIONS High cardiomyocyte stiffness contributed to stiffness of failing human myocardium because of reduced titin distensibility. The latter resulted from an absent stiffness-lowering effect of baseline phosphorylation and from titin aggregation. High cardiomyocyte stiffness was corrected by α-B crystallin probably through relief of titin aggregation.
Collapse
Affiliation(s)
- Constantijn Franssen
- From the Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.F., J.K., R.M., N.H., W.J.P.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (N.H.)
| | - Jeroen Kole
- From the Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.F., J.K., R.M., N.H., W.J.P.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (N.H.)
| | - René Musters
- From the Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.F., J.K., R.M., N.H., W.J.P.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (N.H.)
| | - Nazha Hamdani
- From the Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.F., J.K., R.M., N.H., W.J.P.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (N.H.)
| | - Walter J Paulus
- From the Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (C.F., J.K., R.M., N.H., W.J.P.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (N.H.).
| |
Collapse
|
140
|
Vang A, Clements RT, Chichger H, Kue N, Allawzi A, O'Connell K, Jeong EM, Dudley SC, Sakhatskyy P, Lu Q, Zhang P, Rounds S, Choudhary G. Effect of α7 nicotinic acetylcholine receptor activation on cardiac fibroblasts: a mechanism underlying RV fibrosis associated with cigarette smoke exposure. Am J Physiol Lung Cell Mol Physiol 2017; 312:L748-L759. [PMID: 28258105 PMCID: PMC5451597 DOI: 10.1152/ajplung.00393.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/17/2017] [Accepted: 02/23/2017] [Indexed: 01/03/2023] Open
Abstract
Right ventricular (RV) dysfunction is associated with numerous smoking-related illnesses, including chronic obstructive pulmonary disease (COPD), in which it is present even in the absence of pulmonary hypertension. It is unknown whether exposure to cigarette smoke (CS) has direct effects on RV function and cardiac fibroblast (CF) proliferation or collagen synthesis. In this study, we evaluated cardiac function and fibrosis in mice exposed to CS and determined mechanisms of smoke-induced changes in CF signaling and fibrosis. AKR mice were exposed to CS for 6 wk followed by echocardiography and evaluation of cardiac hypertrophy, collagen content, and pulmonary muscularization. Proliferation and collagen content were evaluated in primary isolated rat CFs exposed to CS extract (CSE) or nicotine. Markers of cell proliferation, fibrosis, and proliferative signaling were determined by immunoblot or Sircol collagen assay. Mice exposed to CS had significantly decreased RV function, as determined by tricuspid annular plane systolic excursion. There were no changes in left ventricular parameters. RV collagen content was significantly elevated, but there was no change in RV hypertrophy or pulmonary vascular muscularization. CSE directly increased CF proliferation and collagen content in CF. Nicotine alone reproduced these effects. CSE and nicotine-induced fibroblast proliferation and collagen content were mediated through α7 nicotinic acetylcholine receptors and were dependent on PKC-α, PKC-δ, and reduced p38-MAPK phosphorylation. CS and nicotine have direct effects on CFs to induce proliferation and fibrosis, which may negatively affect right heart function.
Collapse
MESH Headings
- Animals
- Cell Proliferation/drug effects
- Enzyme Activation/drug effects
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Heart Ventricles/drug effects
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Hemodynamics/drug effects
- Hypertrophy, Right Ventricular/complications
- Hypertrophy, Right Ventricular/diagnostic imaging
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- MAP Kinase Signaling System/drug effects
- Male
- Mice, Inbred AKR
- Myocardium/pathology
- Nicotine/pharmacology
- Phosphorylation/drug effects
- Protein Kinase C-alpha/metabolism
- Protein Kinase C-delta/metabolism
- Rats, Sprague-Dawley
- Smoking/adverse effects
- Vascular Remodeling/drug effects
- Ventricular Dysfunction, Right/complications
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Dysfunction, Right/pathology
- Ventricular Dysfunction, Right/physiopathology
- alpha7 Nicotinic Acetylcholine Receptor/metabolism
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Alexander Vang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Richard T Clements
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Surgery, Rhode Island Hospital, Providence, Rhode Island; and
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
| | - Havovi Chichger
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Nouaying Kue
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Ayed Allawzi
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island
| | - Kelly O'Connell
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Euy-Myoung Jeong
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
| | - Samuel C Dudley
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
| | - Pavlo Sakhatskyy
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Peng Zhang
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island;
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
141
|
Prabhu SD, Frangogiannis NG. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ Res 2017; 119:91-112. [PMID: 27340270 DOI: 10.1161/circresaha.116.303577] [Citation(s) in RCA: 1547] [Impact Index Per Article: 193.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022]
Abstract
In adult mammals, massive sudden loss of cardiomyocytes after infarction overwhelms the limited regenerative capacity of the myocardium, resulting in the formation of a collagen-based scar. Necrotic cells release danger signals, activating innate immune pathways and triggering an intense inflammatory response. Stimulation of toll-like receptor signaling and complement activation induces expression of proinflammatory cytokines (such as interleukin-1 and tumor necrosis factor-α) and chemokines (such as monocyte chemoattractant protein-1/ chemokine (C-C motif) ligand 2 [CCL2]). Inflammatory signals promote adhesive interactions between leukocytes and endothelial cells, leading to extravasation of neutrophils and monocytes. As infiltrating leukocytes clear the infarct from dead cells, mediators repressing inflammation are released, and anti-inflammatory mononuclear cell subsets predominate. Suppression of the inflammatory response is associated with activation of reparative cells. Fibroblasts proliferate, undergo myofibroblast transdifferentiation, and deposit large amounts of extracellular matrix proteins maintaining the structural integrity of the infarcted ventricle. The renin-angiotensin-aldosterone system and members of the transforming growth factor-β family play an important role in activation of infarct myofibroblasts. Maturation of the scar follows, as a network of cross-linked collagenous matrix is formed and granulation tissue cells become apoptotic. This review discusses the cellular effectors and molecular signals regulating the inflammatory and reparative response after myocardial infarction. Dysregulation of immune pathways, impaired suppression of postinfarction inflammation, perturbed spatial containment of the inflammatory response, and overactive fibrosis may cause adverse remodeling in patients with infarction contributing to the pathogenesis of heart failure. Therapeutic modulation of the inflammatory and reparative response may hold promise for the prevention of postinfarction heart failure.
Collapse
Affiliation(s)
- Sumanth D Prabhu
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.)
| | - Nikolaos G Frangogiannis
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.).
| |
Collapse
|
142
|
|
143
|
Xu L, Jia Y, Yang XH, Han F, Zheng Y, Ni Y, Chen X, Hong J, Liu JQ, Li Q, Sun RH, Mo SJ. MicroRNA-130b transcriptionally regulated by histone H3 deacetylation renders Akt ubiquitination and apoptosis resistance to 6-OHDA. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1678-1689. [PMID: 28412322 DOI: 10.1016/j.bbadis.2017.04.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/09/2017] [Accepted: 04/11/2017] [Indexed: 12/12/2022]
Abstract
Apoptosis of DA neurons is a contributing cause of disability and death for Parkinson's disease (PD). Akt may become a potential therapeutic target for PD since Akt has been deactivated during DA neuron apoptosis. We previously demonstrated that Akt confers apoptosis resistance against 6-OHDA in DA neuron-like PC12 cells, yet the underlying mechanisms accounted for this are not fully understood. Here we report that microRNA-130b (miR-130b)-dependent and cylindromatosis (CYLD) repression-mediated Akt ubiquitination renders apoptosis resistance of PC12 cells to 6-OHDA, which elicits histone H3 deacetylation-induced transcriptional downregulation of miR-130b vice versa. CYLD deficiency ubiquitinates Akt at Lys63, thereby phosphorylating Akt and antagonizing 6-OHDA-initiated apoptosis. MiR-130b targetedly represses CYLD and increases apoptosis resistance to 6-OHDA. CYLD repression by miR-130b restores Akt ubiquitination and activation, GSK3β and FoxO3a phosphorylation, FoxO3a removal from Bim promoter as well as Bim downregulation during 6-OHDA administration. CYLD deficiency-mediated Akt activation is instrumental for the apoptosis-resistant phenotypes of miR-130b. In addition, 6-OHDA transcriptionally downregulates miR-130b through recruitment of HDAC3 at the promoter. Furthermore, EPO potentiates the ability of miR-130b to activate Akt and augment apoptosis resistance. Our findings identify the apoptosis-resistant function of miR-130b and suggest that histone H3 deacetylation plays a pivotal role in regulating miR-130b transcription in response to 6-OHDA.
Collapse
Affiliation(s)
- Liang Xu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Yu Jia
- Department of Nephrology, Tongji Hospital, Tongji Medical College of Huanzhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Xiang-Hong Yang
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Fang Han
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Yang Zheng
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Yin Ni
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Xu Chen
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Jun Hong
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Jing-Quan Liu
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Qian Li
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China
| | - Ren-Hua Sun
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China.
| | - Shi-Jing Mo
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, Zhejiang, China.
| |
Collapse
|
144
|
PTPIP51 regulates mouse cardiac ischemia/reperfusion through mediating the mitochondria-SR junction. Sci Rep 2017; 7:45379. [PMID: 28345618 PMCID: PMC5366942 DOI: 10.1038/srep45379] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
Protein tyrosine phosphatase interacting protein 51 (PTPIP51) participates in multiple cellular processes, and dysfunction of PTPIP51 is implicated in diseases such as cancer and neurodegenerative disorders. However, there is no functional evidence showing the physiological or pathological roles of PTPIP51 in the heart. We have therefore investigated the role and mechanisms of PTPIP51 in regulating cardiac function. We found that PTPIP51 was markedly upregulated in ischemia/reperfusion heart. Upregulation of PTPIP51 by adenovirus-mediated overexpression markedly increased the contact of mitochondria-sarcoplasmic reticulum (SR), elevated mitochondrial Ca2+ uptake from SR release through mitochondrial Ca2+uniporter. Inhibition or knockdown of mitochondrial Ca2+uniporter reversed PTPIP51-mediated increase of mitochondrial Ca2+ and protected cardiomyocytes against PTPIP51-mediated apoptosis. More importantly, cardiac specific knockdown of PTPIP51 largely reduced myocardium infarction size and heart injury after ischemia/reperfusion. Our study defines a novel and essential function of PTPIP51 in the cardiac ischemia/reperfusion process by mediating mitochondria-SR contact. Downregulation of PTPIP51 improves heart function after ischemia/reperfusion injury, suggesting PTPIP51 as a therapeutic target for ischemic heart diseases.
Collapse
|
145
|
Erythropoietin Modification Enhances the Protection of Mesenchymal Stem Cells on Diabetic Rat-Derived Schwann Cells: Implications for Diabetic Neuropathy. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6352858. [PMID: 28299330 PMCID: PMC5337339 DOI: 10.1155/2017/6352858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 01/05/2017] [Indexed: 11/18/2022]
Abstract
Diabetes-triggered apoptosis of Schwann cells (SC) contributes to the degradation of diabetic peripheral neuropathy (DNP). In recent years, mesenchymal stem cells (MSC) were applied to DPN repair and it was demonstrated that paracrine secretion played a key role in neuroprotection exerted by MSC. Erythropoietin (EPO) is a potent cytokine capable of reducing apoptosis of SC. However, the expression of EPO in MSC is limited. In this study, we hypothesized that overexpression of EPO in MSC (EPO-MSC) may significantly improve their neuroprotective potentials. The EPO overexpression in MSC was achieved by lentivirus transduction. SC derived from the periphery nerve of diabetic rats were cocultured with MSC or EPO-MSC in normal or high glucose culture condition, respectively. In normal glucose culture condition, the overexpression of EPO in MSC promoted the MSC-induced restoration of SC from diabetic rats, including increases in GSH level and cell viability, decrease in TUNEL apoptosis, upregulation of antiapoptotic proteins, p-Akt, and Bcl-2, and downregulation of proapoptotic proteins, cleaved caspase-3, and Bax. The subsequent results in high glucose culture condition showed similar promotions achieved by EPO-MSC. Thus, it could be concluded that EPO-MSC possessed a potent potential in hampering apoptosis of SC, and the suppression was probably attributed to attenuating oxidative stress and regulating apoptosis related protein factors.
Collapse
|
146
|
Overexpression of Heme Oxygenase-1 in Mesenchymal Stem Cells Augments Their Protection on Retinal Cells In Vitro and Attenuates Retinal Ischemia/Reperfusion Injury In Vivo against Oxidative Stress. Stem Cells Int 2017; 2017:4985323. [PMID: 28255307 PMCID: PMC5309411 DOI: 10.1155/2017/4985323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/26/2016] [Accepted: 12/21/2016] [Indexed: 01/08/2023] Open
Abstract
Retinal ischemia/reperfusion (I/R) injury, involving several ocular diseases, seriously threatens human ocular health, mainly treated by attenuating I/R-induced oxidative stress. Currently, mesenchymal stem cells (MSCs) could restore I/R-injured retina through paracrine secretion. Additionally, heme oxygenase-1 (HO-1) could ameliorate oxidative stress and thus retinal apoptosis, but the expression of HO-1 in MSC is limited. Here, we hypothesized that overexpression of HO-1 in MSC (MSC-HO-1) may significantly improve their retina-protective potentials. The overexpression of HO-1 in MSC was achieved by lentivirus transduction. Then, MSC or MSC-HO-1 was cocultured with retinal ganglion cells (RGC-5) in H2O2-simulated oxidative condition and their protection on RGC-5 was systemically valuated in vitro. Compared with MSC, MSC-HO-1 significantly attenuated H2O2-induced injury of RGC-5, including decrease in cellular ROS level and apoptosis, activation of antiapoptotic proteins p-Akt and Bcl-2, and blockage of proapoptotic proteins cleaved caspase 3 and Bax. In retinal I/R rats model, compared with control MSC, MSC-HO-1-treated retina significantly retrieved its structural thickness, reduced cell apoptosis, markedly attenuated retinal oxidative stress level, and largely regained the activities of typical antioxidant enzymes, SOD and CAT. Therefore, it could be concluded that overexpression of HO-1 provides a promising strategy to enhance the MSC-based therapy for I/R-related retinal injury.
Collapse
|
147
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 635] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
148
|
Ruan L, Wang Y, Chen SC, Zhao T, Huang Q, Hu ZL, Xia NZ, Liu JJ, Chen WJ, Zhang Y, Cheng JL, Gao HC, Yang YJ, Sun HZ. Metabolite changes in the ipsilateral and contralateral cerebral hemispheres in rats with middle cerebral artery occlusion. Neural Regen Res 2017; 12:931-937. [PMID: 28761426 PMCID: PMC5514868 DOI: 10.4103/1673-5374.208575] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cerebral ischemia not only causes pathological changes in the ischemic areas but also induces a series of secondary changes in more distal brain regions (such as the contralateral cerebral hemisphere). The impact of supratentorial lesions, which are the most common type of lesion, on the contralateral cerebellum has been studied in patients by positron emission tomography, single photon emission computed tomography, magnetic resonance imaging and diffusion tensor imaging. In the present study, we investigated metabolite changes in the contralateral cerebral hemisphere after supratentorial unilateral ischemia using nuclear magnetic resonance spectroscopy-based metabonomics. The permanent middle cerebral artery occlusion model of ischemic stroke was established in rats. Rats were randomly divided into the middle cerebral artery occlusion 1-, 3-, 9- and 24-hour groups and the sham group. 1H nuclear magnetic resonance spectroscopy was used to detect metabolites in the left and right cerebral hemispheres. Compared with the sham group, the concentrations of lactate, alanine, γ-aminobutyric acid, choline and glycine in the ischemic cerebral hemisphere were increased in the acute stage, while the concentrations of N-acetyl aspartate, creatinine, glutamate and aspartate were decreased. This demonstrates that there is an upregulation of anaerobic glycolysis (shown by the increase in lactate), a perturbation of choline metabolism (suggested by the increase in choline), neuronal cell damage (shown by the decrease in N-acetyl aspartate) and neurotransmitter imbalance (evidenced by the increase in γ-aminobutyric acid and glycine and by the decrease in glutamate and aspartate) in the acute stage of cerebral ischemia. In the contralateral hemisphere, the concentrations of lactate, alanine, glycine, choline and aspartate were increased, while the concentrations of γ-aminobutyric acid, glutamate and creatinine were decreased. This suggests that there is a difference in the metabolite changes induced by ischemic injury in the contralateral and ipsilateral cerebral hemispheres. Our findings demonstrate the presence of characteristic changes in metabolites in the contralateral hemisphere and suggest that they are most likely caused by metabolic changes in the ischemic hemisphere.
Collapse
Affiliation(s)
- Lei Ruan
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yan Wang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shu-Chao Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tian Zhao
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qun Huang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zi-Long Hu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Neng-Zhi Xia
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jin-Jin Liu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wei-Jian Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yong Zhang
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing-Liang Cheng
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hong-Chang Gao
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yun-Jun Yang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hou-Zhang Sun
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
149
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|
150
|
Lv B, Li F, Fang J, Xu L, Sun C, Han J, Hua T, Zhang Z, Feng Z, Wang Q, Jiang X. Activated Microglia Induce Bone Marrow Mesenchymal Stem Cells to Produce Glial Cell-Derived Neurotrophic Factor and Protect Neurons Against Oxygen-Glucose Deprivation Injury. Front Cell Neurosci 2016; 10:283. [PMID: 28018176 PMCID: PMC5160383 DOI: 10.3389/fncel.2016.00283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/28/2016] [Indexed: 12/16/2022] Open
Abstract
In this study, we investigated interactions among microglia (MG), bone marrow mesenchymal stem cells (BMSCs) and neurons in cerebral ischemia and the potential mechanisms using an in vitro oxygen-glucose deprivation (OGD) model. Rat BMSCs were incubated with conditioned medium (CM) from in vitro cultures of OGD-activated rat MG and murine BV2 MG cells. Effects of glial cell-derived neurotrophic factor (GDNF) on rat neuron viability, apoptosis, lactate dehydrogenase (LDH) leakage and mitochondrial membrane potential (MMP) were analyzed in this model. OGD-activated MG promoted GDNF production by BMSCs (P < 0.01). Tumor necrosis factor-α (TNFα), but not interleukin-6 (IL6) or interleukin 1β (IL1β), promoted GDNF production by BMSCs (P < 0.001). GDNF or CM pre-treated BMSCs elevated neuronal viability and suppressed apoptosis (P < 0.05 or P < 0.01); these effects were inhibited by the RET antibody. GDNF activated MEK/ERK and phosphoinositide-3-kinase (PI3K)/AKT signaling but not JNK/c-JUN. Furthermore, GDNF upregulated B cell lymphoma 2 (BCL2) and heat shock 60 kDa protein 1 (HSP60) levels, suppressed LDH leakage, and promoted MMP. Thus, activated MG produce TNFα to stimulate GDNF production by BMSCs, which prevents and repairs OGD-induced neuronal injury, possibly via regulating MEK/ERK and PI3K/AKT signaling. These findings will facilitate the prevention and treatment of neuronal injury by cerebral ischemia.
Collapse
Affiliation(s)
- Bingke Lv
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Feng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Jie Fang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Limin Xu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Chengmei Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Jianbang Han
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Tian Hua
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Zhongfei Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Zhiming Feng
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Qinghua Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Xiaodan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| |
Collapse
|