51
|
The functional mechanism of bone marrow-derived mesenchymal stem cells in the treatment of animal models with Alzheimer's disease: crosstalk between autophagy and apoptosis. Stem Cell Res Ther 2022; 13:90. [PMID: 35241159 PMCID: PMC8895531 DOI: 10.1186/s13287-022-02765-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
The transplantation of bone marrow-derived mesenchymal stem cells (BMMSCs) alleviates neuropathology and improves cognitive deficits in animal models with Alzheimer's disease. However, the underlying mechanism remains undefined. Based on meta-analysis and comprehensive review, high-profile studies support the theory that transplanted BMMSCs activate autophagy, as evidenced by the expression levels of signal molecules such as Beclin-1, Atg5, LC3-II, and mTOR. Functional autophagy mitigates neuronal apoptosis, which is reflected by the alterations of IAPs, Bcl-2, caspase-3, and so forth. Moreover, the transplantation of BMMSCs can decrease aberrant amyloid-beta peptides as well as tau aggregates, inhibit neuroinflammation, and stimulate synaptogenesis. There is a signal crosstalk between autophagy and apoptosis, which may be regulated to produce synergistic effect on the preconditioning of stem cells. Forasmuch, the therapeutic effect of transplanted BMMSCs can be enhanced by autophagy and/or apoptosis modulators.
Collapse
|
52
|
Hula N, Vu J, Quon A, Kirschenman R, Spaans F, Liu R, Cooke CLM, Davidge ST. Sex-Specific Effects of Prenatal Hypoxia on the Cardiac Endothelin System in Adult Offspring. Am J Physiol Heart Circ Physiol 2022; 322:H442-H450. [PMID: 35119336 DOI: 10.1152/ajpheart.00636.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal hypoxia, a major consequence of complicated pregnancies, impairs offspring cardiac tolerance to ischemia/reperfusion (I/R) insult, however, the mechanisms remain unknown. Endothelin-1 (ET-1) signaling through the endothelin A receptors (ETA) is associated with cardiac dysfunction. We hypothesized that prenatal hypoxia exacerbates cardiac susceptibility to I/R via increased ET-1 and ETA levels, while ETA inhibition ameliorates this. Pregnant Sprague-Dawley rats were exposed to normoxia (21% O2) or hypoxia (11% O2) on gestational days 15-21. Offspring were aged to 4 months, and hearts were aerobically perfused or subjected to ex vivo I/R, with or without pre-infusion with an ETA antagonist (ABT-627). ET-1 levels were assessed with ELISA in aerobically perfused and post-I/R left ventricles (LV). ETA and ETB levels were assessed by Western blotting in non-perfused LV. As hypothesized, ABT-627 infusion tended to improve post-I/R recovery in hypoxic females (p=0.0528), however, surprisingly, ABT-627 prevented post-I/R recovery only in the hypoxic males (p<0.001). ET-1 levels were increased in post-I/R LV in both sexes regardless of the prenatal exposure (p<0.01). ETA expression was similar among all groups, while ETB (isoform C) levels were decreased in prenatally hypoxic females (p<0.05). In prenatally hypoxic males, ETA signaling may be essential for tolerance to I/R, while in prenatally hypoxic females, ETA may contribute to cardiac dysfunction. Our data illustrate that understanding the prenatal history has critical implications for treatment strategies in adult chronic diseases.
Collapse
Affiliation(s)
- Nataliia Hula
- Department of Physiology, University of Alberta, Edmonton, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jennie Vu
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Ricky Liu
- Department of Physiology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Sandra T Davidge
- Department of Physiology, University of Alberta, Edmonton, Canada.,Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
53
|
Salehi MS, Safari A, Pandamooz S, Jurek B, Hooshmandi E, Owjfard M, Bayat M, Zafarmand SS, Miyan JA, Borhani-Haghighi A. The Beneficial Potential of Genetically Modified Stem Cells in the Treatment of Stroke: a Review. Stem Cell Rev Rep 2022; 18:412-440. [PMID: 34033001 PMCID: PMC8144279 DOI: 10.1007/s12015-021-10175-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 12/16/2022]
Abstract
The last two decades have witnessed a surge in investigations proposing stem cells as a promising strategy to treat stroke. Since growth factor release is considered as one of the most important aspects of cell-based therapy, stem cells over-expressing growth factors are hypothesized to yield higher levels of therapeutic efficiency. In pre-clinical studies of the last 15 years that were investigating the efficiency of stem cell therapy for stroke, a variety of stem cell types were genetically modified to over-express various factors. In this review we summarize the current knowledge on the therapeutic efficiency of stem cell-derived growth factors, encompassing techniques employed and time points to evaluate. In addition, we discuss several types of stem cells, including the recently developed model of epidermal neural crest stem cells, and genetically modified stem cells over-expressing specific factors, which could elevate the restorative potential of naive stem cells. The restorative potential is based on enhanced survival/differentiation potential of transplanted cells, apoptosis inhibition, infarct volume reduction, neovascularization or functional improvement. Since the majority of studies have focused on the short-term curative effects of genetically engineered stem cells, we emphasize the need to address their long-term impact.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Institute of Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Jaleel A Miyan
- Faculty of Biology, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| | | |
Collapse
|
54
|
Role of ranolazine in heart failure: From cellular to clinic perspective. Eur J Pharmacol 2022; 919:174787. [PMID: 35114190 DOI: 10.1016/j.ejphar.2022.174787] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/25/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022]
Abstract
Ranolazine was approved by the US Food and Drug Administration as an antianginal drug in 2006, and has been used since in certain groups of patients with stable angina. The therapeutic action of ranolazine was initially attributed to inhibitory effects on fatty acids metabolism. As investigations went on, however, it developed that the main beneficial effects of ranolazine arise from its action on the late sodium current in the heart. Since late sodium currents were discovered to be involved in various heart pathologies such as ischemia, arrhythmias, systolic and diastolic dysfunctions, and all these conditions are associated with heart failure, ranolazine has in some way been tested either directly or indirectly on heart failure in numerous experimental and clinical studies. As the heart continuously remodels following any sort of severe injury, the inhibition by ranolazine of the underlying mechanisms of cardiac remodeling including ion disturbances, oxidative stress, inflammation, apoptosis, fibrosis, metabolic dysregulation, and neurohormonal impairment are discussed, along with unresolved issues. A projection of pathologies targeted by ranolazine from cellular level to clinical is provided in this review.
Collapse
|
55
|
Lee YS, Javan H, Reems JA, Li L, Lusty Beech J, Schaaf CI, Pierce J, Phillips JD, Selzman CH. Acellular human amniotic fluid protects the ischemic/reperfused rat myocardium. Am J Physiol Heart Circ Physiol 2022; 322:H406-H416. [DOI: 10.1152/ajpheart.00331.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amniotic products are potent immunomodulators utilized clinically to repair tissue injury. Little information exists regarding the potential of cell-free human amniotic fluid (hAF) to treat cardiovascular disease. Herein, we sought to determine the influence and efficacy of acellular hAF on myocardial ischemia/reperfusion injury. Processed hAF was obtained from volunteer donors at the time of elective caesarean section and manufactured using proprietary methods. Left anterior descending coronary artery ligation was performed on rats for 60 minutes. Thirty minutes after release and reperfusion, either saline or hAF was injected intramyocardially. Serial echocardiography revealed that compared to saline injected rats, hAF animals maintained their ejection fraction and did not adversely remodel through the 4-week period. This preserved ventricular function correlated with decreased infarct size, less fibrosis, and reduced expression of cytokines and infiltrating inflammatory cells. Comparative arrays of different donor hAF lots confirmed the presence of a wide array of immunomodulatory and host-defense proteins. The observed functional cardioprotection was furthermore evident when given intravenously and across multiple hAF donors. In conclusion, our data demonstrate, for the first time, the cardioprotective effect of acellular hAF on myocardial injury. These observations spanned across diverse donors and likely result from the mixture of a plethora of naturally produced cytokines, chemokines, and immune-modulating proteins rather than a single, defined mechanistic culprit. The ubiquitous availability of hAF as a cell-free solution further suggests its potential for widespread adoption as a therapy for myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Young Sook Lee
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Hadi Javan
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jo-Anna Reems
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Ling Li
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jessica Lusty Beech
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Christine I. Schaaf
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jan Pierce
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - John D. Phillips
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Craig H. Selzman
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
56
|
Zhao D, Ji J, Li S, Wu A. Skullcapflavone II protects neuronal damage in cerebral ischemic rats via inhibiting NF-ĸB and promoting angiogenesis. Microvasc Res 2022; 141:104318. [PMID: 35026288 DOI: 10.1016/j.mvr.2022.104318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Cerebral ischemia (CI) is considered as a main cause of cerebral stroke (CS) and poses significant risk to the mankind across the world. In the present study, we intended to investigate the protective effect of Skullcapflavone II (SCP) a flavonoid isolated from S. baicalensis on cerebral ischemia/reperfusion (I/R) injury. METHODS The middle cerebral artery occlusion (MCAO) and reperfusion was used to create ischemic stroke rat model. The rats were treated with (5, 10, and 15 mg/kg) SCP and after the end of the experiment the rats were sacrificed and various biochemical parameters were assed to determine the pharmacological action of SCP. RESULTS SCP dramatically decreases cerebral edema, infarct volume, and improves neurological manifestation as confirmed by reduced neurological deficit. SCP also improves the survivability of neurons as evidenced by H and E and Nissl staining. The level of oxidative stress in the cerebral cortex of the rats was found reduced after treatment with SCP, as confirmed by increase in GSH and SOD activity with reduction in MDA content. In addition, SCP attenuated inflammation via reducing the level of TNF-α, IL-1β and IL-6 in brain tissues of rats. SCP increases the expression of Bcl2, cleaved caspase-3 and -9, while decreasing Bax, and NF-ĸB/TLR4. It causes induction of angiogenesis as suggested by increased expression of VEGF, Ang-1 and Tie-2 in cerebral cortex of rat. CONCLUSIONS Our data determined that SCP may provide protective effect on the I/R-induced cerebral ischemia.
Collapse
Affiliation(s)
- Danpeng Zhao
- Department of Neurology, Ward 6, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou City 450000, China
| | - Jinming Ji
- Department of Neurology, Binzhou People's Hospital, Binzhou, Shandong Province 256610, China
| | - Shanshan Li
- Department of Neurology, Binzhou People's Hospital, Binzhou, Shandong Province 256610, China
| | - Aimei Wu
- Department of Neurology, Xi'an Fengcheng Hospital, No.9 Fengcheng 3(rd) Road, Economic and Technological Development Zone, Xi'an, Shaanxi 710000, China.
| |
Collapse
|
57
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Ma H, Zhang Y, Prasad NR, Singh N, Fu F, Pei JM, Sarybaev A, Sydykov A. The effect of adaptation to hypoxia on cardiac tolerance to ischemia/reperfusion. J Biomed Res 2022:1-25. [PMID: 37183617 PMCID: PMC10387748 DOI: 10.7555/jbr.36.20220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The acute myocardial infarction (AMI) and sudden cardiac death (SCD), both associated with acute cardiac ischemia, are one of the leading causes of adult death in economically developed countries. The development of new approaches for the treatment and prevention of AMI and SCD remains the highest priority for medicine. A study on the cardiovascular effects of chronic hypoxia (CH) may contribute to the development of these methods. Chronic hypoxia exerts both positive and adverse effects. The positive effects are the infarct-reducing, vasoprotective, and antiarrhythmic effects, which can lead to the improvement of cardiac contractility in reperfusion. The adverse effects are pulmonary hypertension and right ventricular hypertrophy. This review presents a comprehensive overview of how CH enhances cardiac tolerance to ischemia/reperfusion. It is an in-depth analysis of the published data on the underlying mechanisms, which can lead to future development of the cardioprotective effect of CH. A better understanding of the CH-activated protective signaling pathways may contribute to new therapeutic approaches in an increase of cardiac tolerance to ischemia/reperfusion.
Collapse
|
58
|
Kessler EL, Wang JW, Kok B, Brans MA, Nederlof A, van Stuijvenberg L, Huang C, Vink A, Arslan F, Efimov IR, Lam CSP, Vos MA, de Kleijn DPV, Fontes MSC, van Veen TAB. Ventricular TLR4 Levels Abrogate TLR2-Mediated Adverse Cardiac Remodeling upon Pressure Overload in Mice. Int J Mol Sci 2021; 22:ijms222111823. [PMID: 34769252 PMCID: PMC8583975 DOI: 10.3390/ijms222111823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Involvement of the Toll-like receptor 4 (TLR4) in maladaptive cardiac remodeling and heart failure (HF) upon pressure overload has been studied extensively, but less is known about the role of TLR2. Interplay and redundancy of TLR4 with TLR2 have been reported in other organs but were not investigated during cardiac dysfunction. We explored whether TLR2 deficiency leads to less adverse cardiac remodeling upon chronic pressure overload and whether TLR2 and TLR4 additively contribute to this. We subjected 35 male C57BL/6J mice (wildtype (WT) or TLR2 knockout (KO)) to sham or transverse aortic constriction (TAC) surgery. After 12 weeks, echocardiography and electrocardiography were performed, and hearts were extracted for molecular and histological analysis. TLR2 deficiency (n = 14) was confirmed in all KO mice by PCR and resulted in less hypertrophy (heart weight to tibia length ratio (HW/TL), smaller cross-sectional cardiomyocyte area and decreased brain natriuretic peptide (BNP) mRNA expression, p < 0.05), increased contractility (QRS and QTc, p < 0.05), and less inflammation (e.g., interleukins 6 and 1β, p < 0.05) after TAC compared to WT animals (n = 11). Even though TLR2 KO TAC animals presented with lower levels of ventricular TLR4 mRNA than WT TAC animals (13.2 ± 0.8 vs. 16.6 ± 0.7 mg/mm, p < 0.01), TLR4 mRNA expression was increased in animals with the largest ventricular mass, highest hypertrophy, and lowest ejection fraction, leading to two distinct groups of TLR2 KO TAC animals with variations in cardiac remodeling. This variation, however, was not seen in WT TAC animals even though heart weight/tibia length correlated with expression of TLR4 in these animals (r = 0.078, p = 0.005). Our data suggest that TLR2 deficiency ameliorates adverse cardiac remodeling and that ventricular TLR2 and TLR4 additively contribute to adverse cardiac remodeling during chronic pressure overload. Therefore, both TLRs may be therapeutic targets to prevent or interfere in the underlying molecular processes.
Collapse
Affiliation(s)
- Elise L. Kessler
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- Correspondence: ; Tel.: +31-628706156
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore; (J.-W.W.); (C.H.)
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore 117599, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Bart Kok
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Maike A. Brans
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
| | - Angelique Nederlof
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Leonie van Stuijvenberg
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore; (J.-W.W.); (C.H.)
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, 3508GA Utrecht, The Netherlands;
| | - Fatih Arslan
- Laboratory Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands
| | - Igor R. Efimov
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA;
| | - Carolyn S. P. Lam
- National Heart Centre Singapore and Duke-National University of Singapore, 5 Hospital Dr, Singapore 169609, Singapore;
- UMC Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Marc A. Vos
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Dominique P. V. de Kleijn
- Department of Vascular Surgery, The Netherlands & Netherlands Heart Institute, University Medical Center Utrecht, Utrecht University, 3508GA Utrecht, The Netherlands;
- The Netherlands Heart Institute, Moreelsepark 1, 3511EP Utrecht, The Netherlands
| | - Magda S. C. Fontes
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3584CM Utrecht, The Netherlands; (B.K.); (M.A.B.); (A.N.); (L.v.S.); (M.A.V.); (M.S.C.F.); (T.A.B.v.V.)
| |
Collapse
|
59
|
Loss of exosomal LncRNA HCG15 prevents acute myocardial ischemic injury through the NF-κB/p65 and p38 pathways. Cell Death Dis 2021; 12:1007. [PMID: 34707098 PMCID: PMC8551195 DOI: 10.1038/s41419-021-04281-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
Exosomes are nanosized bilayer membrane vesicles that may mediate intercellular communication by transporting bioactive molecules, including noncoding RNAs, mRNAs, and proteins. Research has shown that exosomes play an important role in acute myocardial infarction (AMI), but the function and regulation of exosomal long noncoding RNAs (lncRNAs) in AMI are unclear. Thus, RNA sequencing (RNA-Seq) was conducted to investigate the exosomal lncRNA transcriptome from MI patients and identified 65 differentially expressed lncRNAs between the MI and control groups. HCG15, one of the differentially expressed lncRNAs, was verified to have the highest correlation with cTnT by qRT-PCR, and it also contributed to the diagnosis of AMI by receiver operating characteristic (ROC) analysis. Upregulation of HCG15 expression facilitated cardiomyocyte apoptosis and inflammatory cytokine production and inhibited cell proliferation. We also confirmed that HCG15 was mainly wrapped in exosomes from AC16 cardiomyocytes under hypoxia, which contributed to cardiomyocyte apoptosis, the release of inflammatory factors, and inhibition of cell proliferation via the activation of the NF-κB/p65 and p38 pathways, whereas suppressing the expression of HCG15exerted opposite effects, In addition, Overexpression of HCG15 aggravated cardiac IR injury in C57BL/6J mice. This study not only helps elucidate exosomal lncRNA function in AMI pathogenesis but also contributes to the development of novel therapeutic strategies.
Collapse
|
60
|
Sugai S, Nonaka T, Tamegai K, Sato T, Haino K, Enomoto T, Nishijima K. Successful repeated uterine artery embolization in postpartum hemorrhage with disseminated intravascular coagulation: a case report and literature review. BMC Pregnancy Childbirth 2021; 21:710. [PMID: 34686156 PMCID: PMC8532337 DOI: 10.1186/s12884-021-04191-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 01/24/2023] Open
Abstract
Background Postpartum hemorrhage (PPH) is a potentially fatal condition requiring urgent and appropriate intervention. Uterine artery embolization (UAE) has a high hemostatic capacity for PPH, but it may fail. Disseminated intravascular coagulation (DIC) has been reported as a risk factor associated with the failure of UAE. Case presentation A 37-year-old primigravida with dichorionic diamniotic twins and placenta previa underwent cesarean section. The blood loss during surgery was 4950 mL. Hemostasis was achieved using an intrauterine balloon tamponade device. However, she lost a further 2400 mL of blood 5 h after surgery. We embolized both uterine arteries using gelatin sponges and confirmed hemostasis. She was suffering from DIC and received ample blood transfusions. However, a further 1300 mL of blood was lost 18 h after surgery and we performed repeated UAE, with complete recanalization of the uterine arteries on both sides and re-embolization with gelatin sponges. Her DIC was treated successfully by blood transfusions at this time, and she showed no further bleeding after the repeated UAE. Conclusions DIC is a risk factor for the failure of UAE. Repeated UAE may be effective after sufficient improvement of the hematological status in patients with PPH and DIC.
Collapse
Affiliation(s)
- Shunya Sugai
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan.
| | - Taro Nonaka
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan
| | - Kana Tamegai
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan
| | - Tatsuhiko Sato
- Radiology and Radiation Oncology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Kazufumi Haino
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan
| | - Takayuki Enomoto
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan
| | - Koji Nishijima
- Departments of Obstetrics and Gynecology, Niigata University Medical and Dental Hospital, 1-757 Asahimachi-dori, Chuo-ku, 951-8510, Niigata, Japan
| |
Collapse
|
61
|
Qin C, Li Y, Wang K. Functional Mechanism of Bone Marrow-Derived Mesenchymal Stem Cells in the Treatment of Animal Models with Alzheimer's Disease: Inhibition of Neuroinflammation. J Inflamm Res 2021; 14:4761-4775. [PMID: 34566422 PMCID: PMC8456430 DOI: 10.2147/jir.s327538] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/18/2021] [Indexed: 12/27/2022] Open
Abstract
The transplantation of bone marrow-derived mesenchymal stem cells (BMMSCs) alleviates neuropathology and improves cognitive deficits in animal models with Alzheimer’s disease. However, the underlying mechanisms remain to be determined. Available data demonstrate transplanted BMMSCs can inhibit neuroinflammation, which may be related to microglial M1/M2 polarization and is regulated by the secretion of autocrine and paracrine cytokines. BMMSCs also mitigate Aβ plaques and Tau tangles in the brain, which may be associated with the recruitment of peripheral blood monocytes and the subsequent comprehensive effects. The therapeutic effects of stem cells involve potential mechanisms such as immunomodulation, apoptosis, and proliferation. BMMSC-mediated functional reconstruction through dynamic remodeling develops a novel balance. Herein, present review recapitulates the molecular basis of BMMSC-assisted biological processes and summarizes the possible mechanisms related to the interaction between BMMSCs and microglia. The transplanted BMMSCs can suppress neuroinflammation that plays a key role in the pathogenesis of Alzheimer’s disease.
Collapse
Affiliation(s)
- Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yongning Li
- Department of International Medical Service & Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Kewei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
62
|
The Efficacy of Transarterial Embolization for Postpartum Hemorrhage Complicated with Disseminated Intravascular Coagulation: A Single-Center Experience. J Clin Med 2021; 10:jcm10184082. [PMID: 34575193 PMCID: PMC8468128 DOI: 10.3390/jcm10184082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Indications for the use of transarterial embolization (TAE) for postpartum hemorrhage (PPH) have been established. However, the efficacy of TAE for PPH complicated by disseminated intravascular coagulation (DIC) remains controversial. In this study, we investigated the efficacy of TAE for PPH complicated by DIC. A database review was conducted to identify patients who were treated with TAE for PPH at our hospital. TAE was performed in 41 patients during the study period. Effective hemostasis was achieved in all cases, but additional procedures, such as re-embolization or hysterectomy, were required in five patients (12.2%). The typical causes of PPH included uterine atony (18 cases), placenta previa (15 cases), amniotic fluid embolism (DIC-type) (11 cases), and placenta accreta spectrum (10 cases). The mean blood loss was 3836 mL. The mean obstetrical DIC and the International Society on Thrombosis and Hemostasis DIC scores were 7.9 and 2.6, respectively. The efficacy of hemostasis was comparable between patients with and without DIC. However, the complete success rate of TAE was lower in patients with DIC as the condition worsened than that in non-DIC patients. Overall, TAE is effective as a minimally invasive treatment for PPH complicated by DIC.
Collapse
|
63
|
Interleukin 1 receptor antagonism abrogates acute pressure-overload induced murine heart failure. Ann Thorac Surg 2021; 114:98-107. [PMID: 34419440 DOI: 10.1016/j.athoracsur.2021.07.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent clinical trials have suggested that blockade of interleukin-1 can favorably impact patients with myocardial infarction and heart failure. However, the mechanism of how antagonism of this specific cytokine in mediating cardiac disease remains unclear. Hence, we sought to determine the influence of IL-1 blockade on acute hypertensive remodeling. METHODS Transverse aortic constriction (TAC) was performed in C57BL mice with or without intraperitoneal administration of interleukin 1 receptor antagonism (IL-1ra). Function, structure, and molecular diagnostics were subsequently performed and analyzed. RESULTS Six weeks after TAC, a progressive decline of ejection fraction and increases in LV mass and dimensions was effectively mitigated with IL-1ra. TAC resulted in an expected profile of hypertrophic markers including myosin heavy chain, atrial natriuretic peptide, and skeletal muscle actin which were all significantly lower in IL-1ra treated mice. While trichrome staining 2-weeks post TAC demonstrated similar levels of fibrosis, IL-1ra reduced expression of collagen-1, TIMP1, and periostin. Investigating the angiogenic response to pressure overload, similar levels of VEGF were observed, but IL-1ra was associated with more SDF-1. Immune cell infiltration (macrophages and lymphocytes) was also decreased in IL-1ra treated mice. Similarly, cytokine concentrations of IL-1, IL-18, and IL-6 were all reduced in IL-1ra-treated animals. CONCLUSIONS IL-1ra prevents the progression towards heart failure associated with acute pressure overload. This functional response was associated with reductions in mediators of fibrosis, cellular infiltration, and cytokine production. These results provide mechanistic insight into recent clinical trials and could springboard future investigations in patients with pressure-overload based cardiomyopathies.
Collapse
|
64
|
Savira F, Magaye R, Scullino CV, Flynn BL, Pitson SM, Anderson D, Creek DJ, Hua Y, Xiong X, Huang L, Liew D, Reid C, Kaye D, Kompa AR, Wang BH. Sphingolipid imbalance and inflammatory effects induced by uremic toxins in heart and kidney cells are reversed by dihydroceramide desaturase 1 inhibition. Toxicol Lett 2021; 350:133-142. [PMID: 34303789 DOI: 10.1016/j.toxlet.2021.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Non-dialysable protein-bound uremic toxins (PBUTs) contribute to the development of cardiovascular disease (CVD) in chronic kidney disease (CKD) and vice versa. PBUTs have been shown to alter sphingolipid imbalance. Dihydroceramide desaturase 1 (Des1) is an important gatekeeper enzyme which controls the non-reversible conversion of sphingolipids, dihydroceramide, into ceramide. The present study assessed the effect of Des1 inhibition on PBUT-induced cardiac and renal effects in vitro, using a selective Des1 inhibitor (CIN038). Des1 inhibition attenuated hypertrophy in neonatal rat cardiac myocytes and collagen synthesis in neonatal rat cardiac fibroblasts and renal mesangial cells induced by the PBUTs, indoxyl sulfate and p-cresol sulfate. This is at least attributable to modulation of NF-κB signalling and reductions in β-MHC, Collagen I and TNF-α gene expression. Lipidomic analyses revealed Des1 inhibition restored C16-dihydroceramide levels reduced by indoxyl sulfate. In conclusion, PBUTs play a critical role in mediating sphingolipid imbalance and inflammatory responses in heart and kidney cells, and these effects were attenuated by Des1 inhibition. Therefore, sphingolipid modifying agents may have therapeutic potential for the treatment of CVD and CKD and warrant further investigation.
Collapse
Affiliation(s)
- Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Ruth Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Carmen V Scullino
- Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia
| | - Yue Hua
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew R Kompa
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Bing Hui Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| |
Collapse
|
65
|
Pluijmert NJ, Atsma DE, Quax PHA. Post-ischemic Myocardial Inflammatory Response: A Complex and Dynamic Process Susceptible to Immunomodulatory Therapies. Front Cardiovasc Med 2021; 8:647785. [PMID: 33996944 PMCID: PMC8113407 DOI: 10.3389/fcvm.2021.647785] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 01/04/2023] Open
Abstract
Following acute occlusion of a coronary artery causing myocardial ischemia and implementing first-line treatment involving rapid reperfusion, a dynamic and balanced inflammatory response is initiated to repair and remove damaged cells. Paradoxically, restoration of myocardial blood flow exacerbates cell damage as a result of myocardial ischemia-reperfusion (MI-R) injury, which eventually provokes accelerated apoptosis. In the end, the infarct size still corresponds to the subsequent risk of developing heart failure. Therefore, true understanding of the mechanisms regarding MI-R injury, and its contribution to cell damage and cell death, are of the utmost importance in the search for successful therapeutic interventions to finally prevent the onset of heart failure. This review focuses on the role of innate immunity, chemokines, cytokines, and inflammatory cells in all three overlapping phases following experimental, mainly murine, MI-R injury known as the inflammatory, reparative, and maturation phase. It provides a complete state-of-the-art overview including most current research of all post-ischemic processes and phases and additionally summarizes the use of immunomodulatory therapies translated into clinical practice.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
66
|
Deng X, Yang P, Gao T, Liu M, Li X. Allicin attenuates myocardial apoptosis, inflammation and mitochondrial injury during hypoxia-reoxygenation: an in vitro study. BMC Cardiovasc Disord 2021; 21:200. [PMID: 33882833 PMCID: PMC8059159 DOI: 10.1186/s12872-021-01918-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/11/2021] [Indexed: 01/19/2023] Open
Abstract
Background Myocardial ischemia–reperfusion (IR) injury is a damage due to an initial reduction in blood flow to the heart, preventing it from receiving enough oxygen, and subsequent restoration of blood flow through the opening of an occluded coronary artery producing paradoxical harmful effects. The finding of new therapies to prevent IR is of utmost importance. Allicin is a compound isolated from garlic having the ability to prevent and cure different diseases, and a protective effect on the myocardium was also demonstrated. Therefore, the aim of this study was to evaluate the in vitro protective effect of Allicin against myocardial IR injury on cardiomyocytes. Methods We established an in vitro hypoxia-reoxygenation (HR) model of primary porcine cardiomyocytes to simulate myocardial IR injury. Primary porcine cardiomyocytes were extracted from Mini-musk swines (1 day old). After a period of adaptation of at least 2–3 days, cardiomyocytes in good condition were selected and randomly divided into control group (normal oxygen for 5 h), HR group (2 h of hypoxia/3 h of reoxygenation), and HR + Allicin group (hypoxia/reoxygenation + Allicin treatment). Results After the induction of hypoxia/reoxygenation, Allicin treatment enhanced the cell viability. Moreover, Allicin treatment resulted in a reduction of apoptosis from 13.5 ± 1.2% to 6.11 ± 0.15% compared with the HR group (p < 0.05), and the apoptosis related proteins were regulated as well, with a decreased expression of Bax, cleaved caspase-3 and cytosolic cytochrome C and an increase in Bcl-2 expression in the HR + Allicin group (all p < 0.01). Pro-inflammatory cytokines, such as interleukin-6 and tumor necrosis factor alpha were down-regulated by the treatment with Allicin (both p < 0.01). In addition, it significantly decreased intracellular reactive oxygen species generation (p < 0.01) and reduced the loss of mitochondrial membrane potential (p < 0.01). Furthermore, the expression of PPARγ coactivator-1α and endothelial nitric oxide synthase was up-regulated (both p < 0.01), while the expression of Endothelin-1, hypoxia inducing factor-1α and transforming growth factor beta was down-regulated (all p < 0.01) by Allicin treatment. Conclusions These results suggested that Allicin protected the cardiomyocytes against HR damage by reducing apoptosis, inflammation and mitochondrial injury, thus providing a basis for its potential use in the treatment of myocardial IR. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-01918-6.
Collapse
Affiliation(s)
- Xinyi Deng
- Peking University China-Japan Friendship School of Clinical Medicine, 2 East Yinghuayuan Street, Hepingli, Beijing, 100029, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Peng Yang
- Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Tong Gao
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Mengru Liu
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Xianlun Li
- Peking University China-Japan Friendship School of Clinical Medicine, 2 East Yinghuayuan Street, Hepingli, Beijing, 100029, China. .,Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China. .,Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
67
|
Liu L, Zhao Q, Lin L, Yang G, Yu L, Zhuo L, Yang Y, Xu Y. Myeloid MKL1 Disseminates Cues to Promote Cardiac Hypertrophy in Mice. Front Cell Dev Biol 2021; 9:583492. [PMID: 33898415 PMCID: PMC8063155 DOI: 10.3389/fcell.2021.583492] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac hypertrophy is a key pathophysiological process in the heart in response to stress cues. Although taking place in cardiomyocytes, the hypertrophic response is influenced by other cell types, both within the heart and derived from circulation. In the present study we investigated the myeloid-specific role of megakaryocytic leukemia 1 (MKL1) in cardiac hypertrophy. Following transverse aortic constriction (TAC), myeloid MKL1 conditional knockout (MFCKO) mice exhibit an attenuated phenotype of cardiac hypertrophy compared to the WT mice. In accordance, the MFCKO mice were protected from excessive cardiac inflammation and fibrosis as opposed to the WT mice. Conditioned media collected from macrophages enhanced the pro-hypertrophic response in cardiomyocytes exposed to endothelin in an MKL1-dependent manner. Of interest, expression levels of macrophage derived miR-155, known to promote cardiac hypertrophy, were down-regulated in the MFCKO mice compared to the WT mice. MKL1 depletion or inhibition repressed miR-155 expression in macrophages. Mechanistically, MKL1 interacted with NF-κB to activate miR-155 transcription in macrophages. In conclusion, our data suggest that MKL1 may contribute to pathological hypertrophy via regulating macrophage-derived miR-155 transcription.
Collapse
Affiliation(s)
- Li Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Qianwen Zhao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Lin Lin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Guang Yang
- Department of Pathology, Suzhou Municipal Hospital Affiliated with Nanjing Medical University, Suzhou, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Lili Zhuo
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
68
|
Wang YM, Khederzadeh S, Li SR, Otecko NO, Irwin DM, Thakur M, Ren XD, Wang MS, Wu DD, Zhang YP. Integrating Genomic and Transcriptomic Data to Reveal Genetic Mechanisms Underlying Piao Chicken Rumpless Trait. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:787-799. [PMID: 33631431 PMCID: PMC9170765 DOI: 10.1016/j.gpb.2020.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/14/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022]
Abstract
Piao chicken, a rare Chinese native poultry breed, lacks primary tail structures, such as pygostyle, caudal vertebra, uropygial gland, and tail feathers. So far, the molecular mechanisms underlying tail absence in this breed remain unclear. In this study, we comprehensively employed comparative transcriptomic and genomic analyses to unravel potential genetic underpinnings of rumplessness in Piao chicken. Our results reveal many biological factors involved in tail development and several genomic regions under strong positive selection in this breed. These regions contain candidate genes associated with rumplessness, including Irx4, Il18, Hspb2, and Cryab. Retrieval of quantitative trait loci (QTL) and gene functions implies that rumplessness might be consciously or unconsciously selected along with the high-yield traits in Piao chicken. We hypothesize that strong selection pressures on regulatory elements might lead to changes in gene activity in mesenchymal stem cells of the tail bud. The ectopic activity could eventually result in tail truncation by impeding differentiation and proliferation of the stem cells. Our study provides fundamental insights into early initiation and genetic basis of the rumpless phenotype in Piao chicken.
Collapse
Affiliation(s)
- Yun-Mei Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China; Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow 143026, Russia
| | - Saber Khederzadeh
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Shi-Rong Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Newton Otieno Otecko
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - David M Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada
| | - Mukesh Thakur
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Zoological Survey of India, Kolkata 700053, India
| | - Xiao-Die Ren
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Ming-Shan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
69
|
Fiegle DJ, Schöber M, Dittrich S, Cesnjevar R, Klingel K, Volk T, Alkassar M, Seidel T. Severe T-System Remodeling in Pediatric Viral Myocarditis. Front Cardiovasc Med 2021; 7:624776. [PMID: 33537349 PMCID: PMC7848076 DOI: 10.3389/fcvm.2020.624776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic heart failure (HF) in adults causes remodeling of the cardiomyocyte transverse tubular system (t-system), which contributes to disease progression by impairing excitation-contraction (EC) coupling. However, it is unknown if t-system remodeling occurs in pediatric heart failure. This study investigated the t-system in pediatric viral myocarditis. The t-system and integrity of EC coupling junctions (co-localization of L-type Ca2+ channels with ryanodine receptors and junctophilin-2) were analyzed by 3D confocal microscopy in left-ventricular (LV) samples from 5 children with myocarditis (age 14 ± 3 months), undergoing ventricular assist device (VAD) implantation, and 5 children with atrioventricular septum defect (AVSD, age 17 ± 3 months), undergoing corrective surgery. LV ejection fraction (EF) was 58.4 ± 2.3% in AVSD and 12.2 ± 2.4% in acute myocarditis. Cardiomyocytes from myocarditis samples showed increased t-tubule distance (1.27 ± 0.05 μm, n = 34 cells) and dilation of t-tubules (volume-length ratio: 0.64 ± 0.02 μm2) when compared with AVSD (0.90 ± 0.02 μm, p < 0.001; 0.52 ± 0.02 μm2, n = 61, p < 0.01). Intriguingly, 4 out of 5 myocarditis samples exhibited sheet-like t-tubules (t-sheets), a characteristic feature of adult chronic heart failure. The fraction of extracellular matrix was slightly higher in myocarditis (26.6 ± 1.4%) than in AVSD samples (24.4 ± 0.8%, p < 0.05). In one case of myocarditis, a second biopsy was taken and analyzed at VAD explantation after extensive cardiac recovery (EF from 7 to 56%) and clinical remission. When compared with pre-VAD, t-tubule distance and density were unchanged, as well as volume-length ratio (0.67 ± 0.04 μm2 vs. 0.72 ± 0.05 μm2, p = 0.5), reflecting extant t-sheets. However, junctophilin-2 cluster density was considerably higher (0.12 ± 0.02 μm−3 vs. 0.05 ± 0.01 μm−3, n = 9/10, p < 0.001), approaching values of AVSD (0.13 ± 0.05 μm−3, n = 56), and the measure of intact EC coupling junctions showed a distinct increase (20.2 ± 5.0% vs. 6.8 ± 2.2%, p < 0.001). Severe t-system loss and remodeling to t-sheets can occur in acute HF in young children, resembling the structural changes of chronically failing adult hearts. T-system remodeling might contribute to cardiac dysfunction in viral myocarditis. Although t-system recovery remains elusive, recovery of EC coupling junctions may be possible and deserves further investigation.
Collapse
Affiliation(s)
- Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Schöber
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sven Dittrich
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Karin Klingel
- Cardiopathology, University Hospital Tuebingen, Tübingen, Germany
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Muhannad Alkassar
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
70
|
Zhao C, Yang Y, An Y, Yang B, Li P. Cardioprotective role of phyllanthin against myocardial ischemia-reperfusion injury by alleviating oxidative stress and inflammation with increased adenosine triphosphate levels in the mice model. ENVIRONMENTAL TOXICOLOGY 2021; 36:33-44. [PMID: 32798296 DOI: 10.1002/tox.23008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/01/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ischemic heart disease is an imperative cause of high morbidity and mortality globally. The cardiac ischemia/reperfusion damage occur in both reperfusion and ischemia. OBJECTIVE In this exploration, we have planned to examine the cardio-protective action of phyllanthin against the myocardial ischemic-reperfusion injury in mice. MATERIALS AND METHODS The myocardial ischemic reperfusion injury (MI-RI) stimulated via coronary artery occlusion, followed by the 10 mg/kg of phyllanthin treatment. The serum cardiac markers and pro-inflammatory markers level was investigated by using the assay kits. The expressions of oxidative stress and inflammatory markers level were investigated by immunohistochemical analysis. Lipid peroxidation, antioxidant enzymes, and ATPase levels level was examined by standard methods. The expression of oxidative stress markers were inspected by the reverse transcription polymerase chain reaction technique. The heart histology was investigated microscopically. RESULTS The phyllanthin treatment increased the body weight, and heart weight also diminished the infarct size in the MI/RI mice. Cardiac markers status was diminished and the blood pressure markers were augmented by the phyllanthin. Histological analysis revealed the protective role of phyllanthin. Suppressed lipid peroxidation and enhanced antioxidant enzymes were noted in the phyllanthin treated mice MI-RI mice. Phyllanthin appreciably suppressed the pro-inflammatory regulators that is, NF-αB p65, IL-6, IL-1β, and TNF-α and enhanced the antioxidant marker expressions. ATPase levels were improved by the phyllanthin in the MI-RI mice. CONCLUSION These novel findings were confirmed the therapeutic role of phyllanthin against the MI-RI in mice. Hence, it can be a promising agent to treat the MI-RI induced cardiac dysfunction.
Collapse
Affiliation(s)
- Cong Zhao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yufei Yang
- College of Basic Medicine, Qingdao Binhai University, Qingdao, China
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
71
|
Silvis MJM, Kaffka genaamd Dengler SE, Odille CA, Mishra M, van der Kaaij NP, Doevendans PA, Sluijter JPG, de Kleijn DPV, de Jager SCA, Bosch L, van Hout GPJ. Damage-Associated Molecular Patterns in Myocardial Infarction and Heart Transplantation: The Road to Translational Success. Front Immunol 2020; 11:599511. [PMID: 33363540 PMCID: PMC7752942 DOI: 10.3389/fimmu.2020.599511] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022] Open
Abstract
In the setting of myocardial infarction (MI), ischemia reperfusion injury (IRI) occurs due to occlusion (ischemia) and subsequent re-establishment of blood flow (reperfusion) of a coronary artery. A similar phenomenon is observed in heart transplantation (HTx) when, after cold storage, the donor heart is connected to the recipient's circulation. Although reperfusion is essential for the survival of cardiomyocytes, it paradoxically leads to additional myocardial damage in experimental MI and HTx models. Damage (or danger)-associated molecular patterns (DAMPs) are endogenous molecules released after cellular damage or stress such as myocardial IRI. DAMPs activate pattern recognition receptors (PRRs), and set in motion a complex signaling cascade resulting in the release of cytokines and a profound inflammatory reaction. This inflammatory response is thought to function as a double-edged sword. Although it enables removal of cell debris and promotes wound healing, DAMP mediated signalling can also exacerbate the inflammatory state in a disproportional matter, thereby leading to additional tissue damage. Upon MI, this leads to expansion of the infarcted area and deterioration of cardiac function in preclinical models. Eventually this culminates in adverse myocardial remodeling; a process that leads to increased myocardial fibrosis, gradual further loss of cardiomyocytes, left ventricular dilation and heart failure. Upon HTx, DAMPs aggravate ischemic damage, which results in more pronounced reperfusion injury that impacts cardiac function and increases the occurrence of primary graft dysfunction and graft rejection via cytokine release, cardiac edema, enhanced myocardial/endothelial damage and allograft fibrosis. Therapies targeting DAMPs or PRRs have predominantly been investigated in experimental models and are potentially cardioprotective. To date, however, none of these interventions have reached the clinical arena. In this review we summarize the current evidence of involvement of DAMPs and PRRs in the inflammatory response after MI and HTx. Furthermore, we will discuss various current therapeutic approaches targeting this complex interplay and provide possible reasons why clinical translation still fails.
Collapse
Affiliation(s)
- Max J. M. Silvis
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Clémence A. Odille
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mudit Mishra
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niels P. van der Kaaij
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Central Military Hospital, Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Joost P. G. Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Saskia C. A. de Jager
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Netherlands
| | - Lena Bosch
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Gerardus P. J. van Hout
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
72
|
Han X, Chen X, Han J, Zhong Y, Li Q, An Y. MiR-324/SOCS3 Axis Protects Against Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury and Regulates Myocardial Ischemia via TNF/NF-κB Signaling Pathway. Int Heart J 2020; 61:1258-1269. [PMID: 33191336 DOI: 10.1536/ihj.19-687] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We aimed at exploring the function of microRNA-324/cytokine signaling 3 (miR-324/SOCS3) axis in hypoxia/reoxygenation (H/R) -induced cardiomyocyte injury and its underlying mechanism. The differential expression genes were analyzed based on the GSE83500 and GSE48060 datasets from the Gene Expression Omnibus (GEO) database. Then, to conduct the function enrichment analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used. The upstream regulatory microRNAs (miRNAs) of the identified genes were predicted by miRanda, miRWalk, and TargetScan websites. MiR-324 expression was measured with quantitative real-time polymerase chain reaction (qRT-PCR). The target binding of miR-324 and SOCS3 was established by dual-luciferase reporter assay. Cardiomyocyte proliferation was analyzed by cell counting kit-8 (CCK-8) assay, whereas the apoptosis was investigated via flow cytometry. The expression of TNF pathway-related proteins was detected by western blot analysis. SOCS3 was upregulated in patients with myocardial infarction (MI), and function enrichment analyses proved that SOCS3 was enriched in TNF signaling pathway. Moreover, we found that miR-324 was the upstream regulatory miRNA of SOCS3 and negatively regulated SOCS3 expression. MiR-324 was downregulated in cardiomyocytes with H/R-induced injury, inhibiting cell proliferation. In the H/R model, SOCS3 suppresses cardiomyocyte proliferation, which was recovered by miR-324, and induces cell apoptosis, which was repressed by miR-324 via regulating the expression of cleaved caspase-3 and p P38-MAPK. MiR-324 upregulation decreased the protein levels of TNF-α, p-P65, and p-IκBα in cardiomyocytes that suffered from H/R, which was reversed with SOCS3 overexpression. MiR-324/SOCS3 axis could improve the H/R-induced injury of cardiomyocytes via regulating TNF/NF-κB signaling pathway, and this might provide a new therapy strategy for myocardial ischemia.
Collapse
Affiliation(s)
- Xuefu Han
- Department of medicine, Qingdao University.,Department of Cardiology, Weifang People's Hospital
| | - Xi Chen
- Department of Stomatology, Weifang Maternal and Child Health Hospital
| | - Jiaqi Han
- Department of medicine, Qingdao University
| | - Yu Zhong
- Department of Personnel, Weifang Maternal and Child Health Hospital
| | - Qinghua Li
- School of Public Health, Weifang Medical University
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Qingdao University.,Qingdao University
| |
Collapse
|
73
|
Liu C, Cai Z, Hu T, Yao Q, Zhang L. Cathepsin B aggravated doxorubicin‑induced myocardial injury via NF‑κB signalling. Mol Med Rep 2020; 22:4848-4856. [PMID: 33173960 PMCID: PMC7646931 DOI: 10.3892/mmr.2020.11583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/25/2020] [Indexed: 11/09/2022] Open
Abstract
Myocyte apoptosis and oxidative stress key critical roles in the process of doxorubicin (DOX)-induced cardiotoxicity. However, how apoptosis and oxidative stress arise in DOX-induced heart injury remains largely unknown. Cathepsin B (CTSB) is a typical lysosomal cysteine protease that is associated with apoptosis, inflammatory responses, oxidative stress and autophagy. The present study aimed to investigate the role of CTSB in DOX-induced heart injury and its potential mechanism. H9C2 cells were infected with adenovirus or transfected with small interfering RNA to overexpress or knock down CTSB, respectively, and then stimulated with DOX. DOX induced increased CTSB expression levels in H9C2 cells. DOX-induced cardiomyocyte apoptosis and oxidative stress were attenuated by CTSB knockdown but aggravated by CTSB overexpression in vitro. Mechanistically, the present study showed that CTSB activated the NF-κB pathway in response to DOX. In summary, CTSB aggravated DOX-induced H9C2 cell apoptosis and oxidative stress via NF-κB signalling. CTSB constitutes a potential therapeutic target for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhulan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lijun Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
74
|
Guo X, Hong S, He H, Zeng Y, Chen Y, Mo X, Li J, Li L, Steinmetz R, Liu Q. NFκB promotes oxidative stress-induced necrosis and ischemia/reperfusion injury by inhibiting Nrf2-ARE pathway. Free Radic Biol Med 2020; 159:125-135. [PMID: 32745764 PMCID: PMC7530060 DOI: 10.1016/j.freeradbiomed.2020.07.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 01/06/2023]
Abstract
In this study, we identified an unexpected pro-cell death role for NFκB in mediating oxidative stress-induced necrosis, and provide new mechanistic evidence that NFκB, in cooperation with HDAC3, negatively regulates Nrf2-ARE anti-oxidative signaling through transcriptional silencing. We showed that genetic inactivation of NFκB-p65 inhibited, whereas activation of NFκB promoted, oxidative stress-induced cell death and HMGB1 release, a biomarker of necrosis. Moreover, NFκB-luciferase activity was elevated in cardiomyocytes after simulated ischemia/reperfusion (sI/R) or doxorubicin (DOX) treatment, and inhibition of NFκB with Ad-p65-shRNA or Ad-IκBαM diminished sI/R- and DOX-induced cell death and HMGB1 release. Importantly, NFκB negatively regulated Nrf2-ARE activity and the expression of antioxidant proteins. Mechanistically, co-immunoprecipitation revealed that p65 was required for Nrf2-HDAC3 interaction and transcriptional silencing of Nrf2-ARE activity. Further, the ability of HDAC3 to repress Nrf2-ARE activity was lost in p65 deficient cells. Pharmacologic inhibition of HADCs or NFκB with trichostatin A (TSA) or BMS-345541, respectively, increased Nrf2-ARE activity and promoted cell survival after sI/R. In vivo, NFκB transcriptional activity in the mouse heart was significantly elevated after ischemia/reperfusion (I/R) injury, which was abolished by cardiomyocyte-specific deletion of p65 using p65fl/flNkx2.5-Cre mice. Moreover, genetic ablation of p65 in the mouse heart attenuated myocardial infarct size after acute I/R injury and improved cardiac remodeling and functional recovery after chronic myocardial infarction. Thus, our results identified NFκB as a key regulator of oxidative stress-induced necrosis by suppressing the Nrf2-ARE antioxidant pathway through an HDAC3-dependent mechanism. This study also revealed a new pathogenic role of NFκB in cardiac ischemic injury and pathological remodeling.
Collapse
Affiliation(s)
- Xiaoyun Guo
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Siqi Hong
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Hui He
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Yachang Zeng
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Yi Chen
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Xiaoliang Mo
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Jing Li
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Lei Li
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Rachel Steinmetz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
75
|
Ischemic postconditioning reduced myocardial ischemia-reperfusion injury: The roles of melatonin and uncoupling protein 3. Anatol J Cardiol 2020; 23:19-27. [PMID: 31911566 PMCID: PMC7141427 DOI: 10.14744/anatoljcardiol.2019.72609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Protective effects of ischemic postconditioning (PostC) decrease/disappear with age and chronic heart diseases. Similarly, low serum melatonin levels have been reported in the same risk groups. The aims of this study were to investigate the effects of melatonin on the protection of PostC in ischemia–reperfusion (I/R)-induced infarct size and roles of uncoupling protein (UCP) 3, irisin, and nuclear factor kappa B (NFkB) levels. Methods: Rats were pinealectomized (Px) or sham operated (non-Px) 2 months before the I/R studies. The left main coronary artery was occluded for 30 min followed by 120 min reperfusion. PostC was induced with three cycles of R/I (10 s each) after ischemia. Results: The infarct size was found to be significantly higher in Px rats (54.68±1.5%) than in the control group (35.1±2.5%). PostC and melatonin administrations to non-Px rats significantly reduced the infarct size. On the other hand, PostC did not create a significant effect in Px rats, but protection was provided when PostC was co-administrated with melatonin. While significant decreases were detected in the UCP3 levels, irisin and NFkB levels increased with I/R and Px. Treatment with PostC and melatonin in non-Px groups and their co-administration in Px groups were found to return all the genes close to normal levels. Conclusion: The physiological and pharmacological concentrations of melatonin may play a role in the protection of PostC. In cases when physiological melatonin is reduced, such as aging and heart diseases, this protection may decrease, and this effect may be restored by melatonin replacement. PostC and melatonin may regulate energy metabolism and inflammatory mediators and protect mitochondria by affecting the UCP3, irisin, and NFkB levels.
Collapse
|
76
|
Yang K, Xu J, Fan M, Tu F, Wang X, Ha T, Williams DL, Li C. Lactate Suppresses Macrophage Pro-Inflammatory Response to LPS Stimulation by Inhibition of YAP and NF-κB Activation via GPR81-Mediated Signaling. Front Immunol 2020; 11:587913. [PMID: 33123172 PMCID: PMC7573489 DOI: 10.3389/fimmu.2020.587913] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
Recent evidence from cancer research indicates that lactate exerts a suppressive effect on innate immune responses in cancer. This study investigated the mechanisms by which lactate suppresses macrophage pro-inflammatory responses. Macrophages [Raw 264.7 and bone marrow derived macrophages (BMDMs)] were treated with LPS in the presence or absence of lactate. Pro-inflammatory cytokines, NF-κB and YAP activation and nuclear translocation were examined. Our results show that lactate significantly attenuates LPS stimulated macrophage TNF-α and IL-6 production. Lactate also suppresses LPS stimulated macrophage NF-κB and YAP activation and nuclear translocation in macrophages. Interestingly, YAP activation and nuclear translocation are required for LPS stimulated macrophage NF-κB activation and TNFα production. Importantly, lactate suppressed YAP activation and nuclear translocation is mediated by GPR81 dependent AMKP and LATS activation which phosphorylates YAP, resulting in YAP inactivation. Finally, we demonstrated that LPS stimulation induces an interaction between YAP and NF-κB subunit p65, while lactate decreases the interaction of YAP and NF-κB, thus suppressing LPS induced pro-inflammatory cytokine production. Our study demonstrates that lactate exerts a previously unknown role in the suppression of macrophage pro-inflammatory cytokine production via GPR81 mediated YAP inactivation, resulting in disruption of YAP and NF-κB interaction and nuclear translocation in macrophages.
Collapse
Affiliation(s)
- Kun Yang
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jingjing Xu
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Fei Tu
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Xiaohui Wang
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
77
|
Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape. Metabolites 2020; 10:metabo10100394. [PMID: 33023034 PMCID: PMC7600384 DOI: 10.3390/metabo10100394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. Molecular chaperones belong to the heat shock protein (HSP) family. They have been identified as important participants in immune functions including antigen presentation, immunostimulation and immunomodulation, and play crucial roles in metabolic rewiring and epigenetic circuits. Growing evidence has accumulated to indicate that metabolic pathways and their metabolites influence the function of immune cells and can alter transcriptional activity through epigenetic modification of (de)methylation and (de)acetylation. However, whether molecular chaperones can regulate metabolic programs to influence immune activity is still largely unclear. In this review, we discuss the available data on the biological function of molecular chaperones to immune responses during inflammation, with a specific focus on the interplay between molecular chaperones and metabolic pathways that drive immune cell fate and function.
Collapse
|
78
|
Sun F, Du J, Li H, Hao S, Zhao G, Lu F. FABP4 inhibitor BMS309403 protects against hypoxia-induced H9c2 cardiomyocyte apoptosis through attenuating endoplasmic reticulum stress. J Cell Mol Med 2020; 24:11188-11197. [PMID: 32896039 PMCID: PMC7576298 DOI: 10.1111/jcmm.15666] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/11/2020] [Accepted: 07/03/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myocardial infarction is characterized by ischaemia-induced cardiomyocyte apoptosis, in which the endoplasmic reticulum (ER) stress plays an important role. The fatty acid-binding protein-4 (FABP4) has been implicated in regulating ER stress and apoptosis. Yet, whether FABP4 is involved in modulating cardiomyocyte apoptosis remains unclarified. By applying an in vitro model of hypoxia-induced apoptosis of H9c2 cardiomyocytes, we found that FABP4 expression was elevated upon hypoxia stimulation, which was further demonstrated to be transcriptionally activated by the hypoxia-inducible factor 1a (HIF-1α). In addition, the pharmacological inhibition of FABP4 with BMS309403 protected against hypoxia-induced apoptosis in cardiomyocytes, indicating that FABP4 induction is detrimental for cardiomyocyte survival under hypoxic condition. Moreover, BMS309403 attenuated ER stress in cardiomyocytes exposed to hypoxia, which, however, was reversed by tunicamycin, an ER stress activator. More importantly, the protective effect of BMS309403 on cardiomyocytes vanished in the presence of tunicamycin. Thus, these observations establish that FABP4 inhibitor BMS309403 reduces hypoxia-induced cardiomyocyte apoptosis through attenuating excessive ER stress, implying that FABP4 inhibition may be of clinical benefit for MI treatment.
Collapse
Affiliation(s)
- Fuqiang Sun
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiangchuan Du
- Department of UltrasoundThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hongbin Li
- Department of Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shuang Hao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guochang Zhao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fanfan Lu
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
79
|
Han J, Su GH, Wang YH, Lu YX, Zhao HL, Shuai XX. 18β-Glycyrrhetinic Acid Improves Cardiac Diastolic Function by Attenuating Intracellular Calcium Overload. Curr Med Sci 2020; 40:654-661. [PMID: 32862375 DOI: 10.1007/s11596-020-2232-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Ranolazine, a late sodium current inhibitor, has been demonstrated to be effective on heart failure. 18β-glycyrrhetinic acid (18β-GA) has the similar inhibitory effect on late sodium currents. However, its effect on diastolic function is still unknown. This study aimed to determine whether 18β-GA can improve the diastolic function and to explore the underlying mechanisms. Eighty male Sprague Dawley (SD) rats of Langendorff model were randomly divided into the following groups: group A, normal cardiac perfusion group; group B, ischemia-reperfusion group; group C, ischemia-reperfusion with anemoniasulcata toxin II (ATX-II); group D, ranolazine group; and group E, 18β-GA group with four different concentrations. Furthermore, a pressure-overloaded rat model induced by trans-aortic constriction (TAC) was established. Echocardiography and hemodynamics were used to evaluate diastolic function at 14th day after TAC. Changes of free intracellular calcium (Ca2+) concentration was indirectly detected by laser scanning confocal microscope to confirm the inhibition of late sodium currents. With the intervention of ATX-II on ischemia reperfusion injury group, 5 µmol/L ranolazine, and 5, 10, 20, 40 µmol/L 18β-GA could improve ATX-II-induced cardiac diastolic dysfunction. 630 mg/kg glycyrrhizin tablets could improve cardiac diastolic function in the pressure-overloaded rats. 18β-GA and ranolazine had similar effects on reducing the free calcium in cardiomyocytes. The study demonstrates that 18β-GA and glycyrrhizin could improve diastolic dysfunction induced by ischemia-reperfusion injury in Langendorff-perfused rat hearts and pressure-overloaded rats. The mechanism may be attributed to the inhibition of enhanced late sodium currents.
Collapse
Affiliation(s)
- Jun Han
- Department of Cardiology, Wuhan Fourth Hospital Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China
| | - Guan-Hua Su
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Hui Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yong-Xin Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hong-Liang Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin-Xin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
80
|
Huang Q, Lou T, Wang M, Xue L, Lu J, Zhang H, Zhang Z, Wang H, Jing C, Zhao D, Sun L, Li X. Compound K inhibits autophagy-mediated apoptosis induced by oxygen and glucose deprivation/reperfusion via regulating AMPK-mTOR pathway in neurons. Life Sci 2020; 254:117793. [PMID: 32416164 DOI: 10.1016/j.lfs.2020.117793] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 12/27/2022]
Abstract
AIMS Oxygen and glucose deprivation and reperfusion (OGD/R) injury contributes to the pathophysiology after ischemic stroke, which needs to urgently develop treatment strategies. Previous studies have demonstrated that autophagy in reperfusion period exerted adverse effects on the cerebral ischemic injury. Ginsenoside monomer compound K (CK) is the main intestinal metabolite of ginseng that exerts the pharmacological activities and has a protective effect against cerebral OGD/R injury. However, the specific molecular mechanism of CK protects against OGD/R injury in neurons is still unclear. MATERIALS AND METHODS In this study, cell viability, reactive oxygen species (ROS) generation, Ca2+ overload, mitochondrial membrane potential depolarization, autophagy and apoptosis were investigated in OGD/R-induced neuronal cells injury after pretreatment with CK and in combination with BML-275 or rapamycin. KEY FINDINGS Our study found that pretreatment with CK protected neurons against OGD/R injury by increasing cell viability and decreasing the ROS generation, mitochondrial damage, and Ca2+ overload. Moreover, CK cut down autophagy-mediated apoptosis via promoting the process of forming autophagosomes into phagocytic precursors. Furthermore, our study clarified the neuroprotective of CK against OGD/R-induced neural autophagy and apoptosis through the regulation of the AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) pathway. SIGNIFICANCE Taken together, our study provides credible experimental evidence and explains the potential molecular mechanism of CK as one of the main bioactive ingredients of ginseng for the treatment of cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Qingxia Huang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China; Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tingting Lou
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China; Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Manying Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China; Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Linyuan Xue
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - He Zhang
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hui Wang
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Chenxu Jing
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, Jilin, China.
| | - Xiangyan Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun University of Chinese Medicine, Changchun, Jilin, China; Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China.
| |
Collapse
|
81
|
Hatami S, White CW, Qi X, Buchko M, Ondrus M, Kinnear A, Himmat S, Sergi C, Nagendran J, Freed DH. Immunity and Stress Responses Are Induced During Ex Situ Heart Perfusion. Circ Heart Fail 2020; 13:e006552. [PMID: 32498623 DOI: 10.1161/circheartfailure.119.006552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ex situ heart perfusion (ESHP) preserves the donated heart in a perfused, beating condition preventing cold storage-related ischemia and provides a platform to evaluate myocardial viability during preservation. However, myocardial function declines gradually during ESHP. Extracorporeal circulation systems are associated with the induction of systemic inflammatory and stress responses. Our aim was to evaluate the incidence of inflammation and induction of endoplasmic reticulum stress responses during an extended period of ESHP. METHODS Cardiac function, myocardial tissue injury, markers of inflammation, oxidative stress, and endoplasmic reticulum stress were assessed in healthy pig hearts, perfused for 12 hours either in nonworking mode (non-WM=7) or working mode (WM, n=6). RESULTS Cardiac function declined during ESHP but was significantly better preserved in the hearts perfused in WM (median 11-hour cardiac index/1-hour cardiac index: WM=27% versus non-WM=9.5%, P=0.022). Myocardial markers of endoplasmic reticulum stress were expressed higher in ESHP hearts compared with in vivo samples. The proinflammatory cytokines and oxidized low-density lipoprotein significantly increased in the perfusate throughout the perfusion in both perfusion groups. The left ventricular expression of the cytokines and malondialdehyde was induced in non-WM, whereas it was not different between WM and in vivo. CONCLUSIONS Myocardial function declines during ESHP regardless of perfusion mode. However, ESHP in WM may lead to superior preservation of myocardial function and viability. Both inflammation and endoplasmic reticulum stress responses are significantly induced during ESHP and may contribute to the myocardial functional decline, representing a potential therapeutic target to improve the clinical donor heart preservation.
Collapse
Affiliation(s)
- Sanaz Hatami
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | | | - Xiao Qi
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Max Buchko
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Martin Ondrus
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Alexandra Kinnear
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Sayed Himmat
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Consolato Sergi
- Laboratory Medicine and Pathology (C.S.), University of Alberta, Edmonton, Canada
| | - Jayan Nagendran
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Alberta Transplant Institute, Edmonton, Canada (J.N., D.N.F.)
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Darren H Freed
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Physiology (D.H.F.), University of Alberta, Edmonton, Canada
- Biomedical Engineering (D.H.F.), University of Alberta, Edmonton, Canada
- Alberta Transplant Institute, Edmonton, Canada (J.N., D.N.F.)
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| |
Collapse
|
82
|
Qin C, Lu Y, Wang K, Bai L, Shi G, Huang Y, Li Y. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer's disease: a meta-analytic review on potential mechanisms. Transl Neurodegener 2020; 9:20. [PMID: 32460886 PMCID: PMC7251864 DOI: 10.1186/s40035-020-00199-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative disorder. Therapeutically, a transplantation of bone marrow mesenchymal stem cells (BMMSCs) can play a beneficial role in animal models of Alzheimer's disease. However, the relevant mechanism remains to be fully elucidated. MAIN BODY Subsequent to the transplantation of BMMSCs, memory loss and cognitive impairment were significantly improved in animal models with Alzheimer's disease (AD). Potential mechanisms involved neurogenesis, apoptosis, angiogenesis, inflammation, immunomodulation, etc. The above mechanisms might play different roles at certain stages. It was revealed that the transplantation of BMMSCs could alter some gene levels. Moreover, the differential expression of representative genes was responsible for neuropathological phenotypes in Alzheimer's disease, which could be used to construct gene-specific patterns. CONCLUSIONS Multiple signal pathways involve therapeutic mechanisms by which the transplantation of BMMSCs improves cognitive and behavioral deficits in AD models. Gene expression profile can be utilized to establish statistical regression model for the evaluation of therapeutic effect. The transplantation of autologous BMMSCs maybe a prospective therapy for patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China.
| | - Yalan Lu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Kewei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Lin Bai
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Guiying Shi
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yiying Huang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yongning Li
- Department of International Medical Service & Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan 1, Dong Cheng District, Beijing, 100730, China
| |
Collapse
|
83
|
Herwig M, Kolijn D, Lódi M, Hölper S, Kovács Á, Papp Z, Jaquet K, Haldenwang P, Dos Remedios C, Reusch PH, Mügge A, Krüger M, Fielitz J, Linke WA, Hamdani N. Modulation of Titin-Based Stiffness in Hypertrophic Cardiomyopathy via Protein Kinase D. Front Physiol 2020; 11:240. [PMID: 32351396 PMCID: PMC7174613 DOI: 10.3389/fphys.2020.00240] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
The giant protein titin performs structure-preserving functions in the sarcomere and is important for the passive stiffness (Fpassive) of cardiomyocytes. Protein kinase D (PKD) enzymes play crucial roles in regulating myocardial contraction, hypertrophy, and remodeling. PKD phosphorylates myofilament proteins, but it is not known whether the giant protein titin is also a PKD substrate. Here, we aimed to determine whether PKD phosphorylates titin and thereby modulates cardiomyocyte Fpassive in normal and failing myocardium. The phosphorylation of titin was assessed in cardiomyocyte-specific PKD knock-out mice (cKO) and human hearts using immunoblotting with a phosphoserine/threonine and a phosphosite-specific titin antibody. PKD-dependent site-specific titin phosphorylation in vivo was quantified by mass spectrometry using stable isotope labeling by amino acids in cell culture (SILAC) of SILAC-labeled mouse heart protein lysates that were mixed with lysates isolated from hearts of either wild-type control (WT) or cKO mice. Fpassive of single permeabilized cardiomyocytes was recorded before and after PKD and HSP27 administration. All-titin phosphorylation was reduced in cKO compared to WT hearts. Multiple conserved PKD-dependent phosphosites were identified within the Z-disk, A-band and M-band regions of titin by quantitative mass spectrometry, and many PKD-dependent phosphosites detected in the elastic titin I-band region were significantly decreased in cKO. Analysis of titin site-specific phosphorylation showed unaltered or upregulated phosphorylation in cKO compared to matched WT hearts. Fpassive was elevated in cKO compared to WT cardiomyocytes and PKD administration lowered Fpassive of WT and cKO cardiomyocytes. Cardiomyocytes from hypertrophic cardiomyopathy (HCM) patients showed higher Fpassive compared to control hearts and significantly lower Fpassive after PKD treatment. In addition, we found higher phosphorylation at CaMKII-dependent titin sites in HCM compared to control hearts. Expression and phosphorylation of HSP27, a substrate of PKD, were elevated in HCM hearts, which was associated with increased PKD expression and phosphorylation. The relocalization of HSP27 in HCM away from the sarcomeric Z-disk and I-band suggested that HSP27 failed to exert its protective action on titin extensibility. This protection could, however, be restored by administration of HSP27, which significantly reduced Fpassive in HCM cardiomyocytes. These findings establish a previously unknown role for PKDin regulating diastolic passive properties of healthy and diseased hearts.
Collapse
Affiliation(s)
- Melissa Herwig
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Detmar Kolijn
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Soraya Hölper
- Sanofi-Aventis Deutschland GmbH Industriepark Höchst, Frankfurt, Germany
| | - Árpád Kovács
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kornelia Jaquet
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Cris Dos Remedios
- School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, NSW, Australia
| | - Peter H Reusch
- Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Mügge
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Jens Fielitz
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, University of Münster, Münster, Germany
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
84
|
NGF mediates protection of mesenchymal stem cells-conditioned medium against 2,5-hexanedione-induced apoptosis of VSC4.1 cells via Akt/Bad pathway. Mol Cell Biochem 2020; 469:53-64. [PMID: 32279149 DOI: 10.1007/s11010-020-03727-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/03/2020] [Indexed: 12/25/2022]
Abstract
It has been shown that the conditioned medium of bone mesenchymal stem cells (BMSC-CM) can inhibit apoptosis of neural cells exposed to 2,5-hexanedione (HD), but its protective mechanism remains unclear. To investigate the underlying mechanism, VSC4.1 cells were given HD and 5, 10 and 15% BMSC-CM (v/v) in the current experiment. Our data showed that BMSC-CM concentration-dependently attenuated HD-induced cell apoptosis. Moreover, BMSC-CM remarkably decreased the mitochondrial cytochrome c (Cyt C) release and the caspase-3 activity in HD-given VSC4.1 cells. Given a relatively high expression of NGF in BMSCs and BMSC-CM, we hypothesized that NGF might be an important mediator of the protection of BMSC-CM against apoptosis induced by HD. To verify our hypothesis, the VSC4.1 cells were administrated with NGF and anti-NGF antibody in addition to HD. As expected, NGF could perfectly mimic BMSC-CM's protective role and these beneficial effects were abolished by anti-NGF antibody intervention. To further explore its mechanism, inhibitors of TrkA and Akt were given to the VSC4.1 cells and NGF/Akt/Bad pathway turned out to be involved in anti-apoptotic role of BMSC-CM. Based on these findings, it was revealed that BMSC-CM beneficial role was mediated by NGF and relied on the Akt/Bad pathway.
Collapse
|
85
|
Du X, Yang J, Liu C, Wang S, Zhang C, Zhao H, Du H, Geng X. Hypoxia-Inducible Factor 1α and 2α Have Beneficial Effects in Remote Ischemic Preconditioning Against Stroke by Modulating Inflammatory Responses in Aged Rats. Front Aging Neurosci 2020; 12:54. [PMID: 32210788 PMCID: PMC7076079 DOI: 10.3389/fnagi.2020.00054] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/18/2020] [Indexed: 11/29/2022] Open
Abstract
Limb remote ischemic preconditioning (RIPC) has been proven to alleviate stroke injury in young rats, but its protective effect and its mechanism in aged rats are still unclear. Hypoxia-inducible factor (HIF) is one of the important markers of stroke, and its high expression plays an important role in the pathogenesis of stroke. In this study, we tested the hypothesis that RIPC could regulate the expression of HIF, leading to reduced inflammatory responses in aged rats. Stroke was induced by transient middle cerebral artery occlusion (MCAo) in aged rats, and RIPC was conducted in both hind limbs. The HIF-1α and HIF-2α mRNA and protein were examined by real-time RT-PCR and western blotting (WB). Inflammatory cytokines in the peripheral blood and brain were measured using AimPlex multiplex immunoassays. The protein levels of p-Akt, Akt, p-ERK, and ERK were examined by WB. We investigated that RIPC reduced the infarct size, improved neurological functions, and decreased the expression of HIF-1α and HIF-2α in the ischemic brain. RIPC reduced the levels of IL-1β, IL-6 and IFN-γ in the peripheral blood and the levels of IL-1β and IFN-γ in the ischemic brain 48 h post-stroke. Moreover, intraperitoneal injection of the HIF inhibitor, acriflavine hydrochloride (ACF), abolished the protection of RIPC with respect to infarct size and neurological functions and neutralized the downregulation of pro-inflammatory IL-1β, IL-6 and IFN-γ. ACF also reversed the activation of the Akt signaling pathway induced by RIPC following stroke. HIF may play a key role in RIPC, which was likely mediated by the Akt signaling pathway and systemic modulation of the inflammatory response in aged rats.
Collapse
Affiliation(s)
- Xiangnan Du
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jian Yang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Cuiying Liu
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Sainan Wang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chencheng Zhang
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Heng Zhao
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Huishan Du
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
86
|
Guo K, Yao X, Wu W, Yu Z, Li Z, Ma Z, Liu D. HIF-1α/SDF-1/CXCR4 axis reduces neuronal apoptosis via enhancing the bone marrow-derived mesenchymal stromal cell migration in rats with traumatic brain injury. Exp Mol Pathol 2020; 114:104416. [PMID: 32165091 DOI: 10.1016/j.yexmp.2020.104416] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/21/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal injection is a promising therapy for traumatic brain injury (TBI). The aim of this study was to explore the effects of the HIF-1α/SDF-1/CXCR4 axis on neuron repair in TBI rats through improving the bone marrow-derived mesenchymalstromal cells (BMSCs) migration. TBI rat models were established. The rats were treated with exogenous SDF-1, and then the neuronal apoptosis in TBI rats was measured. BMSCs from rats were collected, and the roles of NF-κB p65 expression in nuclei, overexpression of SDF-1 and HIF-1α, as well as downregulation of CXCR4 in BMSC migration were identified. HIF-1α- and SDF-1- treated BMSCs were transplanted into TBI rats, after which the neuronal apoptosis and activity of the HIF-1α/SDF-1/CXCR4 axis were detected. Consequently, we found SDF-1 elevated the HIF-1α/SDF-1/CXCR4 activity and presented protective roles in TBI rat hippocampal neurons with reduced neuronal apoptosis. SDF-1 promoted BMSC migration in vitro, and co-effects of SDF-1 and HIF-1α showed strong promotion, while CXCR4 inhibition suppressed BMSC migration. BMSC transplantation activated the HIF-1α/SDF-1/CXCR4 axis and reduced neuronal apoptosis in TBI rats. To conclude, our study demonstrated that the HIF-1α/SDF-1/CXCR4 axis could enhance BMSC migration and alleviate neuronal damage and apoptosis in TBI rats. This study provided novel options for TBI therapy.
Collapse
Affiliation(s)
- Kai Guo
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Xinyu Yao
- Department of Anesthesia, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Weijing Wu
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Ziyi Yu
- Department of Intensive Care Unit,Tangshan Worker Hospital, Tangshan 063000, Hebei, PR China
| | - Zhenzhong Li
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Zenglu Ma
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Dengxiang Liu
- Department of Radiotherapy, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China.
| |
Collapse
|
87
|
Lebek S, Pichler K, Reuthner K, Trum M, Tafelmeier M, Mustroph J, Camboni D, Rupprecht L, Schmid C, Maier LS, Arzt M, Wagner S. Enhanced CaMKII-Dependent Late I
Na
Induces Atrial Proarrhythmic Activity in Patients With Sleep-Disordered Breathing. Circ Res 2020; 126:603-615. [DOI: 10.1161/circresaha.119.315755] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rationale:
Sleep-disordered breathing (SDB) is frequently associated with atrial arrhythmias. Increased CaMKII (Ca/calmodulin-dependent protein kinase II) activity has been previously implicated in atrial arrhythmogenesis.
Objective:
We hypothesized that CaMKII-dependent dysregulation of Na current (I
Na
) may contribute to atrial proarrhythmic activity in patients with SDB.
Methods and Results:
We prospectively enrolled 113 patients undergoing elective coronary artery bypass grafting for cross-sectional study and collected right atrial appendage biopsies. The presence of SDB (defined as apnea-hypopnea index ≥15/h) was assessed with a portable SDB monitor the night before surgery. Compared with 56 patients without SDB, patients with SDB (57) showed a significantly increased level of activated CaMKII. Patch clamp was used to measure I
Na
. There was a significantly enhanced late I
Na
, but reduced peak I
Na
due to enhanced steady-state inactivation in atrial myocytes of patients with SDB consistent with significantly increased CaMKII-dependent cardiac Na channel phosphorylation (Na
V
1.5, at serine 571, Western blotting). These gating changes could be fully reversed by acute CaMKII inhibition (AIP [autocamtide-2 related inhibitory peptide]). As a consequence, we observed significantly more cellular afterdepolarizations and more severe premature atrial contractions in atrial trabeculae of patients with SDB, which could be blocked by either AIP or KN93 (N-[2-[[[(E)-3-(4-chlorophenyl)prop-2-enyl]-methylamino]methyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide). In multivariable linear regression models incorporating age, sex, body mass index, existing atrial fibrillation, existing heart failure, diabetes mellitus, and creatinine levels, apnea-hypopnea index was independently associated with increased CaMKII activity, enhanced late I
Na
and correlated with premature atrial contraction severity.
Conclusions:
In atrial myocardium of patients with SDB, increased CaMKII-dependent phosphorylation of Na
V
1.5 results in dysregulation of I
Na
with proarrhythmic activity that was independent from preexisting comorbidities. Inhibition of CaMKII may be useful for prevention or treatment of arrhythmias in SDB.
Clinical Trial Registration:
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT02877745.
Visual Overview:
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Simon Lebek
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Konstantin Pichler
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Kathrin Reuthner
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Maximillian Trum
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Maria Tafelmeier
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Julian Mustroph
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Daniele Camboni
- Department of Cardiothoracic Surgery (D.C., L.R., C.S.), University Hospital Regensburg, Germany
| | - Leopold Rupprecht
- Department of Cardiothoracic Surgery (D.C., L.R., C.S.), University Hospital Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery (D.C., L.R., C.S.), University Hospital Regensburg, Germany
| | - Lars S. Maier
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Michael Arzt
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| | - Stefan Wagner
- From the Department of Internal Medicine II (S.L., K.P., K.R., M. Trum, M. Tafelmeier, J.M., L.S.M., M.A., S.W.), University Hospital Regensburg, Germany
| |
Collapse
|
88
|
Neuromuscular Diseases Due to Chaperone Mutations: A Review and Some New Results. Int J Mol Sci 2020; 21:ijms21041409. [PMID: 32093037 PMCID: PMC7073051 DOI: 10.3390/ijms21041409] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle and the nervous system depend on efficient protein quality control, and they express chaperones and cochaperones at high levels to maintain protein homeostasis. Mutations in many of these proteins cause neuromuscular diseases, myopathies, and hereditary motor and sensorimotor neuropathies. In this review, we cover mutations in DNAJB6, DNAJB2, αB-crystallin (CRYAB, HSPB5), HSPB1, HSPB3, HSPB8, and BAG3, and discuss the molecular mechanisms by which they cause neuromuscular disease. In addition, previously unpublished results are presented, showing downstream effects of BAG3 p.P209L on DNAJB6 turnover and localization.
Collapse
|
89
|
Jiang T, You H, You D, Zhang L, Ding M, Yang B. A miR-1275 mimic protects myocardiocyte apoptosis by regulating the Wnt/NF-κB pathway in a rat model of myocardial ischemia-reperfusion-induced myocardial injury. Mol Cell Biochem 2020; 466:129-137. [PMID: 32056105 DOI: 10.1007/s11010-020-03695-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
This study evaluated the cardioprotective effects of a miR-1275 mimic in a rat model of myocardial ischemia-reperfusion (I/R)-induced myocardial injury (MI). Three groups of rats were established: a sham-operated group, a MI group and a MI+miR-1275 group pretreated for 1 week i.p. with a miR-1275 mimic at a concentration of 30 pmol/mL. MI was induced by I/R. The levels of myocardial enzymes in serum were estimated in all rats, together with haemodynamic functions. The effects of the miR-1275 mimic were determined based on the serum concentrations of inflammatory mediators in the treated vs. sham and MI rats. In addition, western blot assay and immunohistochemical analyses were performed to examine the effect of the miR-1275 mimic on the Wnt/NF-kB signalling pathway in MI rats. Treatment with the miR-1275 mimic attenuated the altered levels of myocardial enzymes and haemodynamic functions seen in MI rats. The myocardial infarct was smaller in rats treated with the miR-1275 mimic than in MI rats. The miR-1275 mimic also reduced myocardiocyte apoptosis and ameliorated the altered Wnt/KF-kB pathway. These results demonstrate the efficacy of the miR-1275 mimic in preventing myocardial I/R-induced MI in rats, by regulating the Wnt/NF-κB pathway.
Collapse
Affiliation(s)
- Tiechao Jiang
- Department of Cardiovascular Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, 130033, China
| | - Hong You
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Dong You
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lirong Zhang
- Depatment of Pathology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Mei Ding
- Department of Cardiovascular Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China. .,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, 130033, China.
| | - Bin Yang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| |
Collapse
|
90
|
Engelland RE, Hemingway HW, Tomasco OG, Olivencia-Yurvati AH, Romero SA. Acute lower leg hot water immersion protects macrovascular dilator function following ischaemia-reperfusion injury in humans. Exp Physiol 2019; 105:302-311. [PMID: 31707732 DOI: 10.1113/ep088154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS • What is the central question of this study? What is the effect of lower leg hot water immersion on vascular ischaemia-reperfusion injury induced in the arm of young healthy humans? • What is the main finding and its importance? Lower leg hot water immersion successfully protects against vascular ischaemia-reperfusion injury in humans. This raises the possibility that targeted heating of the lower legs may be an alternative therapeutic approach to whole-body heating that is equally efficacious at protecting against vascular ischaemia-reperfusion injury. ABSTRACT Reperfusion that follows a period of ischaemia paradoxically reduces vasodilator function in humans and contributes to the tissue damage associated with an ischaemic event. Acute whole-body hot water immersion protects against vascular ischaemia-reperfusion (I-R) injury in young healthy humans. However, the effect of acute lower leg heating on I-R injury is unclear. Therefore, the purpose of this study was to test the hypothesis that, compared with thermoneutral control immersion, acute lower leg hot water immersion would prevent the decrease in macro- and microvascular dilator functions following I-R injury in young healthy humans. Ten young healthy subjects (5 female) immersed their lower legs into a circulated water bath for 60 min under two randomized conditions: (1) thermoneutral control immersion (∼33°C) and (2) hot water immersion (∼42°C). Macrovascular (brachial artery flow-mediated dilatation) and microvascular (forearm reactive hyperaemia) dilator functions were assessed using Doppler ultrasound at three time points: (1) pre-immersion, (2) 60 min post-immersion, and (3) post-I/R (20 min of arm ischaemia followed by 20 min of reperfusion). Ischaemia-reperfusion injury reduced macrovascular dilator function following control immersion (pre-immersion 6.0 ± 2.1% vs. post-I/R 3.6 ± 2.1%; P < 0.05), but was well-maintained with prior hot water immersion (pre-immersion 5.8 ± 2.1% vs. post-I/R 5.3 ± 2.1%; P = 0.8). Microvascular dilator function did not differ between conditions or across time. Taken together, acute lower leg hot water immersion prevents the decrease in macrovascular dilator function that occurs following I-R injury in young healthy humans.
Collapse
Affiliation(s)
- Rachel E Engelland
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Holden W Hemingway
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Olivia G Tomasco
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Albert H Olivencia-Yurvati
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Surgery, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Steven A Romero
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
91
|
Deng L, Zhang F, Wu Y, Luo J, Mao X, Long L, Gou M, Yang L, Deng DYB. RGD-Modified Nanocarrier-Mediated Targeted Delivery of HIF-1α-AA Plasmid DNA to Cerebrovascular Endothelial Cells for Ischemic Stroke Treatment. ACS Biomater Sci Eng 2019; 5:6254-6264. [PMID: 33405532 DOI: 10.1021/acsbiomaterials.9b01362] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Studies have shown that the use of proangiogenic genes can improve the prognosis of ischemic stroke by promoting angiogenesis at the injury site. For example, within this study, hypoxia-inducible factor 1-α (HIF-1α) has exhibited an angiogenic effect. Our previous study reported a more stable HIF-1α mutant form (HIF-1α-AA), which was transfected into mesenchymal stem cells to provide neuroprotective effects against ischemic stroke. The safety of nonviral gene vectors has attracted researchers' attention. This study encapsulated the HIF-1α-AA plasmid DNA into a newly synthesized effective nonviral gene vector, a hyperbranched cationic amylopectin derivative (DMAPA-Amyp) nanocarrier. In addition, a targeting strategy was applied to select the RGD peptides and bind to the designed nanocarrier as a molecule targeting endothelial cells. The targeting strategy is used to directly deliver the nanocarriers to the vascular endothelial cells of the brain peri-infarct site. This study emphasizes the targeting ability of nanocarrier and its therapeutic effect on cerebral ischemia. The results showed that RGD-DMAPA-Amyp had good biocompatibility and a high cell uptake rate, indicating that it is a safe nonviral gene vector that can be endocytosed by human cells. In rat models of ischemic stroke, compared with the nontargeted nanocarrier group, more RGD-DMAPA-Amyp nanoparticles aggregated in vascular endothelial cells of the peri-infarct region and significantly improved the recovery of neurological function. It is indicated that the RGD-modified nanomedicine promotes the recovery of nerve function more efficiently. Further study on the mechanism of RGD-DMAPA-Amyp/HIF-1α-AA in the treatment of cerebral ischemia displayed potential to significantly promote the formation of new blood vessels in vivo. Our findings suggest that the RGD-modified nonviral gene vector containing HIF-1α-AA appears to be a safe and promising therapeutic strategy for ischemic stroke gene therapy.
Collapse
Affiliation(s)
- Lingna Deng
- Scientific Research Center and Department of Orthopedic, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | | | | | | | - Lingli Long
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | | | - David Y B Deng
- Scientific Research Center and Department of Orthopedic, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
92
|
Al-Huseini I, Harada M, Nishi K, Nguyen-Tien D, Kimura T, Ashida N. Improvement of insulin signalling rescues inflammatory cardiac dysfunction. Sci Rep 2019; 9:14801. [PMID: 31616027 PMCID: PMC6794250 DOI: 10.1038/s41598-019-51304-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation resulting from virus infection is the cause of myocarditis; however, the precise mechanism by which inflammation induces cardiac dysfunction is still unclear. In this study, we investigated the contribution of insulin signalling to inflammatory cardiac dysfunction induced by the activation of signalling by NF-κB, a major transcriptional factor regulating inflammation. We generated mice constitutively overexpressing kinase-active IKK-β, an essential kinase for NF-κB activation, in cardiomyocytes (KA mice). KA mice demonstrated poor survival and significant cardiac dysfunction with remarkable dilation. Histologically, KA hearts revealed increased cardiac apoptosis and fibrosis and the enhanced recruitment of immune cells. By molecular analysis, we observed the increased phosphorylation of IRS-1, indicating the suppression of insulin signalling in KA hearts. To evaluate the contribution of insulin signalling to cardiac dysfunction in KA hearts, we generated mice with cardiac-specific suppression of phosphatase and tensin homologue 10 (PTEN), a negative regulator of insulin signalling, in the KA mouse background (KA-PTEN). The suppression of PTEN successfully improved insulin signalling in KA-PTEN hearts, and interestingly, KA-PTEN mice showed significantly improved cardiac function and survival. These results indicated that impaired insulin signalling underlies the mechanism involved in inflammation-induced cardiac dysfunction, which suggests that it may be a target for the treatment of myocarditis.
Collapse
Affiliation(s)
- Isehaq Al-Huseini
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Masayuki Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiyoto Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA98109, USA
| | - Dat Nguyen-Tien
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Molecular Immunology and Inflammation, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noboru Ashida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
93
|
Pradhan D, Tambe V, Raval N, Gondalia P, Bhattacharya P, Kalia K, Tekade RK. Dendrimer grafted albumin nanoparticles for the treatment of post cerebral stroke damages: A proof of concept study. Colloids Surf B Biointerfaces 2019; 184:110488. [PMID: 31541894 DOI: 10.1016/j.colsurfb.2019.110488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/23/2019] [Accepted: 09/01/2019] [Indexed: 01/08/2023]
Abstract
Stroke is the second largest disease of mortality. The biggest hurdle in designing effective brain drug delivery systems is offered by the blood-brain barrier (BBB), which is highly impermeable to many drugs. Albumin nanoparticles (NP) have gained attention due to their multiple ligand binding sites and long circulatory half-life. Citicoline (CIT) is reported to enhance the acetylcholine secretion in the brain and also helps in membrane repair and regeneration. However, the poor BBB permeation of CIT results in lower levels of CIT in the brain. This demands the development of a suitable delivery platform to completely realize the therapeutic benefit of CIT in stroke therapy. This investigation reports the synthesis and characterization of second generation (2.0 G) dendrimer Amplified Albumin (dAA) biopolymer by FTIR, MALDI-TOF, and surface charge (mV). Further, the synthesized biopolymer has been utilized to develop a CIT nanoformulation using a commercially translatable one-pot process. Release of CIT from biopolymer was performed within an acetate buffer at pH 5 and Phosphate buffer at pH 7.4. Further, we investigated the ability of biopolymer to permeate BBB by in vitro permeability assay in bEnd.3 cells. MTT assay of CIT-dAA-NP, CIT-ANP, and 2.0 G PAMAM dendrimers was performed in bEnd.3 cells. Therapeutic efficacy of the synthesized biopolymer was determined by VEGF gene expression within an in vitro hypoxia model in PC12 cells. Thus, this investigation resulted in biopolymers that can be used to deliver any therapeutic agent by altering the permeability of the BBB. Also, cationization by dendrimer grafting is one such strategy that may be used to cationize any other negatively charged polymer, such as albumin. The synthesized biopolymer is not limited to deliver molecules to the brain, but can also be used to increase the loading of negatively-charged drug molecules, siRNA, or any other oligonucleotide.
Collapse
Affiliation(s)
- Deepak Pradhan
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Vishakha Tambe
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Nidhi Raval
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Piyush Gondalia
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Pallab Bhattacharya
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Palaj, Opp. Air Force Station, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
94
|
Recent Advances in Pharmacological and Non-Pharmacological Strategies of Cardioprotection. Int J Mol Sci 2019; 20:ijms20164002. [PMID: 31426434 PMCID: PMC6720817 DOI: 10.3390/ijms20164002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemic heart diseases (IHD) are the leading cause of death worldwide. Although the principal form of treatment of IHD is myocardial reperfusion, the recovery of coronary blood flow after ischemia can cause severe and fatal cardiac dysfunctions, mainly due to the abrupt entry of oxygen and ionic deregulation in cardiac cells. The ability of these cells to protect themselves against injury including ischemia and reperfusion (I/R), has been termed “cardioprotection”. This protective response can be stimulated by pharmacological agents (adenosine, catecholamines and others) and non-pharmacological procedures (conditioning, hypoxia and others). Several intracellular signaling pathways mediated by chemical messengers (enzymes, protein kinases, transcription factors and others) and cytoplasmic organelles (mitochondria, sarcoplasmic reticulum, nucleus and sarcolemma) are involved in cardioprotective responses. Therefore, advancement in understanding the cellular and molecular mechanisms involved in the cardioprotective response can lead to the development of new pharmacological and non-pharmacological strategies for cardioprotection, thus contributing to increasing the efficacy of IHD treatment. In this work, we analyze the recent advances in pharmacological and non-pharmacological strategies of cardioprotection.
Collapse
|
95
|
Roles of forkhead box O (FoxO) transcription factors in neurodegenerative diseases: A panoramic view. Prog Neurobiol 2019; 181:101645. [PMID: 31229499 DOI: 10.1016/j.pneurobio.2019.101645] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/03/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDDs), which are among the most important aging-related diseases, are typically characterized by neuronal damage and a progressive impairment in neurological function during aging. Few effective therapeutic targets for NDDs have been revealed; thus, an understanding of the pathogenesis of NDDs is important. Forkhead box O (FoxO) transcription factors have been implicated in the mechanisms regulating aging and longevity. The functions of FoxOs are regulated by diverse post-translational modifications (e.g., phosphorylation, acetylation, ubiquitination, methylation and glycosylation). FoxOs exert both detrimental and protective effects on NDDs. Therefore, an understanding of the precise function of FoxOs in NDDs will be helpful for developing appropriate treatment strategies. In this review, we first introduce the post-translational modifications of FoxOs. Next, the regulation of FoxO expression and post-translational modifications in the central nervous system (CNS) is described. Afterwards, we analyze and address the important roles of FoxOs in NDDs. Finally, novel potential directions of future FoxO research in NDDs are discussed. This review recapitulates essential facts and questions about the promise of FoxOs in treating NDDs, and it will likely be important for the design of further basic studies and to realize the potential for FoxOs as therapeutic targets in NDDs.
Collapse
|
96
|
Hu J, Huang CX, Rao PP, Cao GQ, Zhang Y, Zhou JP, Zhu LY, Liu MX, Zhang GG. MicroRNA-155 inhibition attenuates endoplasmic reticulum stress-induced cardiomyocyte apoptosis following myocardial infarction via reducing macrophage inflammation. Eur J Pharmacol 2019; 857:172449. [PMID: 31207208 DOI: 10.1016/j.ejphar.2019.172449] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 01/05/2023]
Abstract
Endoplasmic reticulum stress (ERS)-induced cardiomyocyte apoptosis plays an important role in the pathological process following myocardial infarction (MI). Macrophages that express microRNA-155 (miR-155) mediate cardiac inflammation, fibrosis, and hypertrophy. Therefore, we investigated if miR-155 regulates ERS-induced cardiomyocyte apoptosis after MI using a mouse model, lipopolysaccharide (LPS)-induced rat bone marrow derived macrophages (BMDMs)and hypoxia-induced neonatal rat cardiomyocytes (NRCMs). In vivo, miR-155 levelswere significantly higher in the MI group compared to the sham group. MI increasedmacrophage infiltration, nuclear factor-κB (NF-κB) activation, ERS induced-apoptosis, and SOCS1 expression, all of which were attenuated by the miR-155 antagomir, with the exception of SOCS1 expression. Additionally, post-MI cardiac dysfunction was significantly improved by miR-155 inhibition. In vitro, LPS upregulated miR-155 expression in BMDMs, and the miR-155 antagomir decreased LPS-induced macrophage inflammation and NF-κB pathway activation, but increased expression of SOCS1. Hypoxia increased NF-κB pathway activation, ERS marker expression, and apoptosis in NRCMs. Interestingly, conditioned medium from LPS-induced macrophages in combination with the miR-155 antagomir decreased, while the miR-155 agomir increased, the hypoxia-induced effects in NRCM's. The miR-155 agomir effects were reversed by inhibiting the NF-κB pathway in cardiomyocytes. Moreover, SOCS1 knockdown in LPS-induced macrophages promoted NF-κB pathway activation and ERS-induced cardiomyocyte apoptosis in the hypoxia-induced NRCMs, but the SOCS1-siRNA-induced effects were markedly decreased by miR-155 antagomir treatment. These data suggest that miR-155 inhibition attenuates ERS-induced cardiomyocyte apoptosis after MI via reducing macrophage inflammation through the SOCS1/NF-κB pathway.
Collapse
Affiliation(s)
- Juan Hu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; Institute of Hypertension, Central South University, Changsha, Hunan, China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, PR China
| | - Pan-Pan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, PR China
| | - Gui-Qiu Cao
- Department of Cardiovascular Medicine, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, PR China
| | - Yin Zhang
- Department of Cardiovascular Medicine, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, PR China
| | - Ji-Peng Zhou
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; Institute of Hypertension, Central South University, Changsha, Hunan, China
| | - Ling-Yan Zhu
- Department of Endocrinology, The First Affiliated Hospital of NanChang University, Nanchang, 330006, China
| | - Ming-Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, PR China
| | - Guo-Gang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; Institute of Hypertension, Central South University, Changsha, Hunan, China.
| |
Collapse
|
97
|
Hadzi‐Petrushev N, Angelovski M, Rebok K, Mitrokhin V, Kamkin A, Mladenov M. Antioxidant and anti‐inflammatory effects of the monocarbonyl curcumin analogs B2BRBC and C66 in monocrotaline‐induced right ventricular hypertrophy. J Biochem Mol Toxicol 2019; 33:e22353. [DOI: 10.1002/jbt.22353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 04/03/2019] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Nikola Hadzi‐Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Marija Angelovski
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Katerina Rebok
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
| | - Vadim Mitrokhin
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| | - Andre Kamkin
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology“Ss. Cyril and Methodius” University in Skopje Skopje Republic of Macedonia
- Department of Fundamental and Applied PhysiologyRussian National Research Medical University Moscow Russia
| |
Collapse
|
98
|
Fender AC, Wakili R, Dobrev D. Straight to the heart: Pleiotropic antiarrhythmic actions of oral anticoagulants. Pharmacol Res 2019; 145:104257. [PMID: 31054953 DOI: 10.1016/j.phrs.2019.104257] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Mechanistic understanding of atrial fibrillation (AF) pathophysiology and the complex bidirectional relationship with thromboembolic risk remains limited. Oral anticoagulation is a mainstay of AF management. An emerging concept is that anticoagulants may themselves have potential pleiotropic disease-modifying effects. We here review the available evidence for hemostasis-independent actions of the oral anticoagulants on electrical and structural remodeling, and the inflammatory component of the vulnerable substrate.
Collapse
Affiliation(s)
- Anke C Fender
- Institute of Pharmacology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Germany.
| | - Reza Wakili
- Clinic for Cardiology and Angiology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Germany
| |
Collapse
|
99
|
Early low-anticoagulant desulfated heparin after traumatic brain injury: Reduced brain edema and leukocyte mobilization is associated with improved watermaze learning ability weeks after injury. J Trauma Acute Care Surg 2019; 84:727-735. [PMID: 29373460 DOI: 10.1097/ta.0000000000001819] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Unfractionated heparin administered immediately after traumatic brain injury (TBI) reduces brain leukocyte (LEU) accumulation, and enhances early cognitive recovery, but may increase bleeding after injury. It is unknown how non-anticoagulant heparins, such as 2,3-O desulfated heparin (ODSH), impact post-TBI cerebral inflammation and long-term recovery. We hypothesized that ODSH after TBI reduces LEU-mediated brain inflammation and improves long-term neurologic recovery. METHODS CD1 male mice (n = 66) underwent either TBI (controlled cortical impact [CCI]) or sham craniotomy. 2,3-O desulfated heparin (25 mg/kg [25ODSH] or 50 mg/kg [50ODSH]) or saline was administered for 48 hours after TBI in 46 animals. At 48 hours, intravital microscopy visualized rolling LEUs and fluorescent albumin leakage in the pial circulation, and the Garcia Neurologic Test assessed neurologic function. Brain edema (wet/dry ratio) was evaluated post mortem. In a separate group of animals (n = 20), learning/memory ability (% time swimming in the Probe platform quadrant) was assessed by the Morris Water Maze 17 days after TBI. Analysis of variance with Bonferroni correction determined significance (p < 0.05). RESULTS Compared with CCI (LEU rolling: 32.3 ± 13.7 LEUs/100 μm per minute, cerebrovascular albumin leakage: 57.4 ± 5.6%), both ODSH doses reduced post-TBI pial LEU rolling (25ODSH: 18.5 ± 9.2 LEUs/100 μm per minute, p = 0.036; 50ODSH: 7.8 ± 3.9 LEUs/100 μm per minute, p < 0.001) and cerebrovascular albumin leakage (25ODSH: 37.9 ± 11.7%, p = 0.001, 50ODSH: 32.3 ± 8.7%, p < 0.001). 50ODSH also reduced injured cerebral hemisphere edema (77.7 ± 0.4%) vs. CCI (78.7 ± 0.4 %, p = 0.003). Compared with CCI, both ODSH doses improved Garcia Neurologic Test at 48 hours. Learning/memory ability (% time swimming in target quadrant) was lowest in CCI (5.9 ± 6.4%) and significantly improved in the 25ODSH group (27.5 ± 8.2%, p = 0.025). CONCLUSION 2,3-O desulfated heparin after TBI reduces cerebral LEU recruitment, microvascular permeability and edema. 2,3-O desulfated heparin may also improve acute neurologic recovery leading to improved learning/memory ability weeks after injury.
Collapse
|
100
|
Zhang L, Jian LL, Li JY, Jin X, Li LZ, Zhang YL, Gong HY, Cui Y. Possible involvement of alpha B-crystallin in the cardioprotective effect of n-butanol extract of Potentilla anserina L. on myocardial ischemia/reperfusion injury in rat. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:320-329. [PMID: 30940361 DOI: 10.1016/j.phymed.2018.10.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND It has been reported that n-butanol extract of Potentilla anserina L (NP) had protective effect against acute myocardial ischemia/reperfusion (I/R) injury in mice. Because of limited phytochemical study on NP, its bioactive compounds and underlying protective mechanisms are largely unclear. PURPOSE The purpose of this study was to investigate the major bioactive compounds and possible mechanism for the cardioprotective effect of NP on rat with I/R injury. METHODS We analyzed the phytochemical isolation of NP and identified the structure of compounds, which was elucidated by a combination of spectroscopic analyses. An I/R model was established by I-30 min/R-2 h in Sprage-Dawley rats. The rats were given intragastric administration of NP (49.3, 98.6, and 197.2 mg•kg-1) continuously for 10 days before I/R operation. The morphological changes and apoptosis of cardiomyocytes were observed by H&E staining, Transmission electron microscope and TUNEL staining respectively. The activities or contents of catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione (GSH) in plasma were detected. Apoptosis related factors were also measured by RT-PCR and western blot. In order to discover the underlying mechanism of NP on I/R, we performed proteomic analysis using two-dimensional gel electrophoresis (2D-DIGE) and matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/MS) to describe differential proteins expression. Potential target protein resulted from 2D-DIGE coupled to MALDI-TOF/MS analysis were further confirmed by immunohistochemical staining, RT-PCR, and western blot. RESULTS We isolated and identified 14 compounds, of which 7 compounds belong to triterpenes. Rats pretreated with NP showed a significant increase on the activities of GSH, SOD and CAT, and remarkable decrease on the content of MDA. NP significantly inhibited the apoptosis of cardiomyocyte and decreased the expression of Cyt C and cleaved-caspase-3. Proteomic analysis revealed that alpha B-crystallin (CryAB) might participate in the NP protective effect against I/R. NP enhanced the level of pCryAB Ser59, whereas the expression of CryAB was decreased. CONCLUSION NP was showed to alleviate I/R injury and inhibit myocardial apoptosis, which might be associated with reduction on oxidative stress and apoptosis. CryAB as a possible target involved in the NP protective effect. This study supplied valuable information to develop novel cardioprotective agents from NP extract.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Le Le Jian
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China; Shanxi Provincial Crops Hospital, Chinese People's Armed Police Forces, Xi'an, Shanxi, China
| | - Jian Yu Li
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Xin Jin
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Ling Zhi Li
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China; Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, China.
| | - Yong Liang Zhang
- Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, China.
| | - Hai Ying Gong
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Ying Cui
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| |
Collapse
|