Pubblicazioni recenti - cardiac stem
-
Cardiac differentiation of human pluripotent stem cells using defined extracellular matrix proteins reveals essential role of fibronectin.
Elife2022 Jun;11():. doi: 10.7554/eLife.69028.
Zhang Jianhua, Gregorich Zachery R, Tao Ran, Kim Gina C, Lalit Pratik A, Carvalho Juliana L, Markandeya Yogananda, Mosher Deane F, Palecek Sean P, Kamp Timothy J,
Abstract
Research and therapeutic applications using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) require robust differentiation strategies. Efforts to improve hPSC-CM differentiation have largely overlooked the role of extracellular matrix (ECM). The present study investigates the ability of defined ECM proteins to promote hPSC cardiac differentiation. Fibronectin (FN), laminin-111, and laminin-521 enabled hPSCs to attach and expand. However, only addition of FN promoted cardiac differentiation in response to growth factors Activin A, BMP4, and bFGF in contrast to the inhibition produced by laminin-111 or laminin-521. hPSCs in culture produced endogenous FN which accumulated in the ECM to a critical level necessary for effective cardiac differentiation. Inducible shRNA knockdown of FN prevented Brachyury mesoderm formation and subsequent hPSC-CM generation. Antibodies blocking FN binding integrins ?4?1 or ?V?1, but not ?5?1, inhibited cardiac differentiation. Furthermore, inhibition of integrin-linked kinase led to a decrease in phosphorylated AKT, which was associated with increased apoptosis and inhibition of cardiac differentiation. These results provide new insights into defined matrices for culture of hPSCs that enable production of FN-enriched ECM which is essential for mesoderm formation and efficient cardiac differentiation.
© 2022, Zhang et al.
Guarda su PubMed -
Adult Mesenchymal Stem Cells and Derivatives in Improved Elastin Homeostasis in a Rat Model of Abdominal Aortic Aneurysms.
Stem Cells Transl Med2022 Jun;():. doi: szac043.
Dahal Shataakshi, Dayal Simran, Androjna Charlie, Peterson John, Ramamurthi Anand,
Abstract
Abdominal aortic aneurysms (AAAs) are localized rupture-prone expansions of the aorta with limited reversibility that develop due to proteolysis of the elastic matrix. Natural regenerative repair of an elastic matrix is difficult due to the intrinsically poor elastogenicity of adult vascular smooth muscle cells (VSMCs). This justifies the need to provide external, pro-elastin regenerative- and anti-proteolytic stimuli to VSMCs in the AAA wall towards reinstating matrix structure in the aorta wall. Introducing alternative phenotypes of highly elastogenic and contractile cells into the AAA wall capable of providing such cues, proffers attractive prospects for AAA treatment. In this regard, we have previously demonstrated the superior elastogenicity of bone marrow mesenchymal stem cell (BM-MSC)-derived SMCs (cBM-SMCs) and their ability to provide pro-elastogenic and anti-proteolytic stimuli to aneurysmal SMCs in vitro. However, the major issues associated with cell therapy, such as their natural ability to home into the AAA tissue, their in vivo biodistribution and retention in the AAA wall, and possible paracrine effects on AAA tissue repair processes in the event of localization in remote tissues remain uncertain. Therefore, in this study we focused on assessing the fate, safety, and AAA reparative effects of BM-MSC-derived cBM-SMCs in vivo. Our results indicate that the cBM-SMCs (a) possess natural homing abilities similar to the undifferentiated BM-MSCs, (b) exhibit higher retention upon localization in the aneurysmal aorta than BM-MSCs, (c) downregulate the expression of several inflammatory and pro-apoptotic cytokines that are upregulated in the AAA wall contributing to accelerated elastic matrix breakdown and suppression of elastic fiber neo-assembly, repair, and crosslinking, and (d) improve elastic matrix content and structure in the AAA wall toward slowing the growth of AAAs. Our study provides initial evidence of the in vivo elastic matrix reparative benefits of cBM-SMCs and their utility in cell therapy to reverse the pathophysiology of AAAs.
© The Author(s) 2022. Published by Oxford University Press.
Guarda su PubMed -
Mesenchymal Stem Cell-derived Extracellular Vesicles Prevent Experimental Bronchopulmonary Dysplasia Complicated By Pulmonary Hypertension.
Stem Cells Transl Med2022 Jun;():. doi: szac041.
Sharma Mayank, Bellio Michael A, Benny Merline, Kulandavelu Shathiyah, Chen Pingping, Janjindamai Chawisa, Han Chenxu, Chang Liming, Sterling Shanique, Williams Kevin, Damianos Andreas, Batlahally Sunil, Kelly Kaitlyn, Aguilar-Caballero Daniela, Zambrano Ronald, Chen Shaoyi, Huang Jian, Wu Shu, Hare Joshua M, Schmidt Augusto, Khan Aisha, Young Karen,
Abstract
Mesenchymal stem cell (MSC) extracellular vesicles (EVs) have beneficial effects in preclinical bronchopulmonary dysplasia and pulmonary hypertension (BPD-PH) models. The optimal source, dosing, route, and duration of effects are however unknown. The objectives of this study were to (a) compare the efficacy of GMP-grade EVs obtained from Wharton's Jelly MSCs (WJ-MSCs) and bone marrow (BM-MSCs), (b) determine the optimal dosing and route of administration, (c) evaluate its long-term effects, and (d) determine how MSC EVs alter the lung transcriptome. Newborn rats exposed to normoxia or hyperoxia (85% O2) from postnatal day (P)1-P14 were given (a) intra-tracheal (IT) BM or WJ-MSC EVs or placebo, (b) varying doses of IT WJ-MSC EVs, or (c) IT or intravenous (IV) WJ-MSC EVs on P3. Rats were evaluated at P14 or 3 months. Early administration of IT BM-MSC or WJ-MSC EVs had similar beneficial effects on lung structure and PH in hyperoxia-exposed rats. WJ-MSC EVs however had superior effects on cardiac remodeling. Low, medium, and high dose WJ-MSC EVs had similar cardiopulmonary regenerative effects. IT and IV WJ-MSC EVs similarly improved vascular density and reduced PH in hyperoxic rats. Gene-set enrichment analysis of transcripts differentially expressed in WJ-MSC EV-treated rats showed that induced transcripts were associated with angiogenesis. Long-term studies demonstrated that a single early MSC EV dose has pulmonary vascular protective effects 3 months after administration. Together, our findings have significant translational implications as it provides critical insight into the optimal source, dosing, route, mechanisms of action, and duration of effects of MSC-EVs for BPD-PH.
© The Author(s) 2022. Published by Oxford University Press.
Guarda su PubMed -
Investigating the Paracrine Role of Perinatal Derivatives: Human Amniotic Fluid Stem Cell-Extracellular Vesicles Show Promising Transient Potential for Cardiomyocyte Renewal.
Front Bioeng Biotechnol2022 ;10():902038. doi: 10.3389/fbioe.2022.902038.
Costa Ambra, Balbi Carolina, Garbati Patrizia, Palamà Maria Elisabetta Federica, Reverberi Daniele, De Palma Antonella, Rossi Rossana, Paladini Dario, Coviello Domenico, De Biasio Pierangela, Ceresa Davide, Malatesta Paolo, Mauri Pierluigi, Quarto Rodolfo, Gentili Chiara, Barile Lucio, Bollini Sveva,
Abstract
Cardiomyocyte renewal represents an unmet clinical need for cardiac regeneration. Stem cell paracrine therapy has attracted increasing attention to resurge rescue mechanisms within the heart. We previously characterized the paracrine effects that human amniotic fluid-derived stem cells (hAFSC) can exert to provide cardioprotection and enhance cardiac repair in preclinical models of myocardial ischemia and cardiotoxicity. Here, we analyze whether hAFSC secretome formulations, namely, hAFSC conditioned medium (hAFSC-CM) over extracellular vesicles (hAFSC-EVs) separated from it, can induce cardiomyocyte renewal. c-KIT+ hAFSC were obtained by leftover samples of II trimester prenatal amniocentesis (fetal hAFSC) and from clinical waste III trimester amniotic fluid during scheduled C-section procedures (perinatal hAFSC). hAFSC were primed under 1% O to enrich hAFSC-CM and EVs with cardioactive factors. Neonatal mouse ventricular cardiomyocytes (mNVCM) were isolated from cardiac tissue of R26pFUCCI2 mice with cell cycle fluorescent tagging by mutually exclusive nuclear signal. mNVCM were stimulated by fetal versus perinatal hAFSC-CM and hAFSC-EVs to identify the most promising formulation for assessment in a R26pFUCCI2 neonatal mouse model of myocardial infarction (MI) intraperitoneal delivery. While the perinatal hAFSC secretome did not provide any significant cardiogenic effect, fetal hAFSC-EVs significantly sustained mNVCM transition from S to M phase by 2-fold, while triggering cytokinesis by 4.5-fold over vehicle-treated cells. Treated mNVCM showed disorganized expression of cardiac alpha-actinin, suggesting cytoskeletal re-arrangements prior to cell renewal, with a 40% significant downregulation of and a positive trend of polymerized F-Actin. Fetal hAFSC-EVs increased cardiomyocyte cell cycle progression by 1.8-fold in the 4-day-old neonatal left ventricle myocardium short term after MI; however, such effect was lost at the later stage. Fetal hAFSC-EVs were enriched with a short isoform of Agrin, a mediator of neonatal heart regeneration acting by YAP-related signaling; yet application of YAP inhibitor verteporfin partially affected EV paracrine stimulation on mNVCM. EVs secreted by developmentally juvenile fetal hAFSC can support cardiomyocyte renewal to some extension, intercellular conveyance of candidates possibly involving Agrin in combination with other factors. These perinatal derivative promising cardiogenic effects need further investigation to define their specific mechanism of action and enhance their potential translation into therapeutic opportunity.
Copyright © 2022 Costa, Balbi, Garbati, Palamà, Reverberi, De Palma, Rossi, Paladini, Coviello, De Biasio, Ceresa, Malatesta, Mauri, Quarto, Gentili, Barile and Bollini.
Guarda su PubMed -
Effects of high glucose and insulin on the electrophysiological properties of cardiomyocytes derived from human-induced pluripotent stem cells.
Zhong Nan Da Xue Xue Bao Yi Xue Ban2022 May;47(5):610-618. doi: 1672-7347(2022)05-0610-09.
Wei Feng, Zhang Yushun, Wang Xingye, Huo Jianhua,
Abstract
OBJECTIVES:
The risk of arrhythmia increases in diabetic patients. However, the effects of hyperglycemia and insulin therapy on the electrophysiological properties of human cardiomyocytes remain unclear. This study is to explore the effects of high glucose and insulin on the electrophysiological properties and arrhythmias of cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs).
METHODS:
Immunofluorescent staining and flow cytometry were used to analyze the purity of hiPSC-CMs generated from human skin fibroblasts of a healthy donor. The hiPSC-CMs were divided into 3 group (treated with normal medium, high glucose and insulin for 4 days): a control group (NM group, containing 5 mmol/L glucose), a high glucose group (HG group, containing 15 mmol/L glucose), and a high glucose combined with insulin (HG+INS group, containing 15 mmol/L glucose+100 mg/L insulin). Electrophysiological changes of hiPSC-CMs were detected by microelectrode array (MEA) before or after treatment with glucose and insulin, including beating rate (BR), field potential duration (FPD) (similar to QT interval in ECG), FPDc (FPD corrected by BR), spike amplitude and conduction velocity (CV). Effects of sotalol on electrophysiological properties and arrhythmias of hiPSC-CMs were also evaluated.
RESULTS:
The expression of cardiac-specific marker of cardiac troponin T was high in the hiPSC-CMs. The purity of hiPSC-CMs was 99.06%. Compared with the NM group, BR was increased by (9.14±0.8)% in the HG group (0.05). Ten µmol/L of sotalol can induce significant arrhythmias from all wells in the HG group. After treatment with insulin and high glucose, compared with the HG group, BR was increased by (8.3±0.5)% in the HG+INS group (0.05). The induction experiment showed that 10 ?mol/L of sotalol could prolong the FPDc of hiPSC-CMs by (78.9±11.6)% in the HG+INS group, but no arrhythmia was induced in each well.
CONCLUSIONS:
High glucose can induce FPD/FPDc of hiPSC-CMs prolongation and increase the risk of arrhythmia induced by drugs. Insulin can reduce the FPD/FPDc prolongation and the risk of induced arrhythmia by high glucose.These results are important to understand the electrophysiological changes of the myocardium in diabetic patients and the impact of insulin therapy on its electrophysiology. Further study on the mechanism may provide new ideas and methods for the treatment of acquired and even inherited long QT syndrome.
Guarda su PubMed -
Wharton's jelly mesenchymal stem cell-derived small extracellular vesicles as natural nanoparticles to attenuate cartilage injury via microRNA regulation.
Int J Pharm2022 Jun;():121952. doi: S0378-5173(22)00507-5.
Chen Penghong, Tang Shijie, Gao Hangqi, Zhang Haoruo, Chen Caixiang, Fang Zhuoqun, Peng Guohao, Weng Haiyan, Chen Aizhen, Zhang Chaoyu, Qiu Zhihuang, Li Shirong, Chen Jinghua, Chen Liangwan, Chen Xiaosong,
Abstract
The main strategy of tissue repair and regeneration focuses on the application of mesenchymal stem cells and cell-based nanoparticles, but there are still multiple challenges that may have negative impacts on human safety and therapeutic efficacy. Cell-free nanotechnology can effectively overcome these obstacles and limitations. Mesenchymal stem cell (MSC)-derived natural small extracellular vesicles (sEVs) represent ideal nanotherapeutics due to their low immunogenicity and lack of tumorigenicity. Here, sEVs harvested from Wharton's jelly mesenchymal stem cells (WJMSCs) were identified. In vitro results showed that WJMSC-sEVs efficiently entered chondrocytes in the osteoarthritis (OA) model, further promoted chondrocyte migration and proliferation and modulated immune reactivity. In vivo, WJMSC-sEVs notably promoted chondrogenesis, which was consistent with the effect of WJMSCs. RNA sequencing results revealed that sEV-microRNA-regulated biocircuits can significantly contribute to the treatment of OA, such as by promoting the activation of the calcium signaling pathway, ECM-receptor interaction pathway and NOTCH signaling pathway. In particular, let-7e-5p, which is found in WJMSC-sEVs, was shown to be a potential core molecule for promoting cartilage regeneration by regulating the levels of STAT3 and IGF1R. Our findings suggest that WJMSC-sEV-induced chondrogenesis is a promising innovative and feasible cell-free nanotherapy for OA treatment.
Copyright © 2022. Published by Elsevier B.V.
Guarda su PubMed -
The road to physiological maturation of stem cell-derived cardiac muscle runs through the sarcomere.
J Mol Cell Cardiol2022 Jun;():. doi: S0022-2828(22)00123-7.
Metzger Joseph M,
Abstract
Recent advances the cardiac biomedical sciences have been propelled forward by the development and implementation of human iPSC-derived cardiac muscle. These notable successes notwithstanding, it is well recognized in the field that a major roadblock persists in the lack of full "adult cardiac muscle-like" maturation of hiPSC-CMs. This Perspective centers focus on maturation roadblocks in the essential physiological unit of muscle, the sarcomere. Stalled sarcomere maturation must be addressed and overcome before this elegant experimental platform can reach the mountaintop of making impactful contributions in disease pathogenesis, drug discovery, and in clinical regenerative medicine.
Copyright © 2022. Published by Elsevier Ltd.
Guarda su PubMed -
Targeting miR-30d reverses pathological cardiac hypertrophy.
EBioMedicine2022 Jun;81():104108. doi: S2352-3964(22)00289-4.
Li Jin, Sha Zhao, Zhu Xiaolan, Xu Wanru, Yuan Weilin, Yang Tingting, Jin Bing, Yan Yuwei, Chen Rui, Wang Siqi, Yao Jianhua, Xu Jiahong, Wang Zitong, Li Guoping, Das Saumya, Yang Liming, Xiao Junjie,
Abstract
BACKGROUND:
Pathological cardiac hypertrophy occurs in response to numerous stimuli and precedes heart failure (HF). Therapies that ameliorate pathological cardiac hypertrophy are highly needed.
METHODS:
The expression level of miR-30d was analyzed in hypertrophy models and serum of patients with chronic heart failure by qRT-PCR. Gain and loss-of-function experiments of miR-30d were performed in vitro. miR-30d gain of function were performed in vivo. Bioinformatics, western blot, luciferase assay, qRT-PCR, and immunofluorescence were performed to examine the molecular mechanisms of miR-30d.
FINDINGS:
miR-30d was decreased in both murine and neonatal rat cardiomyocytes (NRCMs) models of hypertrophy. miR-30d overexpression ameliorated phenylephrine (PE) and angiotensin II (Ang II) induced hypertrophy in NRCMs, whereas the opposite phenotype was observed when miR-30d was downregulated. Consistently, the miR-30d transgenic rat was found to protect against isoproterenol (ISO)-induced pathological hypertrophy. Mechanistically, methyltransferase EZH2 could promote H3K27me3 methylation in the promotor region of miR-30d and suppress its expression during the pathological cardiac hypertrophy. miR-30d prevented pathological cardiac hypertrophy via negatively regulating its target genes MAP4K4 and GRP78 and inhibiting pro-hypertrophic nuclear factor of activated T cells (NFAT). Adeno-associated virus (AAV) serotype 9 mediated-miR-30d overexpression exhibited beneficial effects in murine hypertrophic model. Notably, miR-30d was reduced in serum of patients with chronic heart failure and miR-30d overexpression could significantly ameliorate pathological hypertrophy in human embryonic stem cell-derived cardiomyocytes.
INTERPRETATION:
Overexpression of miR-30d may be a potential approach to treat pathological cardiac hypertrophy.
FUNDING:
This work was supported by the grants from National Key Research and Development Project (2018YFE0113500 to J Xiao), National Natural Science Foundation of China (82020108002 to J Xiao, 81900359 to J Li), the grant from Science and Technology Commission of Shanghai Municipality (20DZ2255400 and 21XD1421300 to J Xiao, 22010500200 to J Li), Shanghai Sailing Program (19YF1416400 to J Li), the "Dawn" Program of Shanghai Education Commission (19SG34 to J Xiao), the "Chen Guang" project supported by the Shanghai Municipal Education Commission and Shanghai Education Development Foundation (19CG45 to J Li).
Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.
Guarda su PubMed -
Translating bone marrow rejuvenation from the bench to beside.
Aging (Albany NY)2022 Jun;undefined(undefined):. doi: 10.18632/aging.204142.
Alibhai Faisal J, Li Ren-Ke,
Guarda su PubMed -
Modeling the human heart ex vivo-current possibilities and strive for future applications.
J Tissue Eng Regen Med2022 Jun;():. doi: 10.1002/term.3335.
Ka?u?na Ewelina, Nadel Agnieszka, Zimna Agnieszka, Rozwadowska Natalia, Kolanowski Tomasz,
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
© 2022 The Authors. Journal of Tissue Engineering and Regenerative Medicine published by John Wiley & Sons Ltd.
Guarda su PubMed -
The Clinical Characteristics and Prognosis of Chinese Patients with Light-Chain Amyloidosis: A Retrospective Multicenter Analysis.
Indian J Hematol Blood Transfus2022 Jul;38(3):444-453. doi: 10.1007/s12288-021-01469-y.
He Donghua, Guan Fangshu, Hu Minli, Zheng Gaofeng, He Jingsong, Han Xiaoyan, Yang Yang, Hong Pan, Wang Gang, Zhao Yi, Wu Wenjun, Cai Zhen,
Abstract
To retrospectively identify the critical characteristics and prognostic factors of light-chain amyloidosis.
PATIENTS AND METHODS:
Data were collected and compared from 91 patients who were diagnosed with light-chain amyloidosis at four hospitals between January 2010 and November 2018. We analyzed the clinical characteristics and performed an overall survival (OS) analysis.
RESULTS:
Patients (median age, 60 years) were diagnosed with organ involvement of the kidney (91.2%), heart (56%), liver (14.3%), soft tissue (18.7%), or gastrointestinal tract (15.4%), and 68.1% of patients had more than two organs involved. Patients were most treated with bortezomib-based regimens (56%), and only one patient had autologous stem cell transplantation (auto-ASCT). The median OS was 36.33 months and was influenced by the ECOG score, renal involvement, cardiac involvement, hepatic involvement, and persistence of positive immunofixation. Patients who received bortezomib-based treatment had a trend of favorable OS compared to those who received non-bortezomib-based treatments, but the difference was not statistically significant. Although the overall number of organs involved was not related to OS, the number of organs involved in the heart, liver and kidney was related. Multivariate analysis indicated that cardiac involvement and negative hematologic response with persistence of positive immunofixation were independent prognostic factors for OS.
CONCLUSION:
Cardiac involvement and the hematologic response to treatment were independent prognostic factors for OS in light-chain amyloidosis patients.
© The Author(s) 2021.
Guarda su PubMed -
Bone marrow endothelial dysfunction promotes myeloid cell expansion in cardiovascular disease.
Nat Cardiovasc Res2022 Jan;1(1):28-44. doi: 10.1038/s44161-021-00002-8.
Rohde David, Vandoorne Katrien, Lee I-Hsiu, Grune Jana, Zhang Shuang, McAlpine Cameron S, Schloss Maximilian J, Nayar Ribhu, Courties Gabriel, Frodermann Vanessa, Wojtkiewicz Gregory, Honold Lisa, Chen Qi, Schmidt Stephen, Iwamoto Yoshiko, Sun Yuan, Cremer Sebastian, Hoyer Friedrich F, Iborra-Egea Oriol, Muñoz-Guijosa Christian, Ji Fei, Zhou Bin, Adams Ralf H, Wythe Joshua D, Hidalgo Juan, Watanabe Hideto, Jung Yookyung, van der Laan Anja M, Piek Jan J, Kfoury Youmna, Désogère Pauline A, Vinegoni Claudio, Dutta Partha, Sadreyev Ruslan I, Caravan Peter, Bayes-Genis Antoni, Libby Peter, Scadden David T, Lin Charles P, Naxerova Kamila, Swirski Filip K, Nahrendorf Matthias,
Abstract
Abnormal hematopoiesis advances cardiovascular disease by generating excess inflammatory leukocytes that attack the arteries and the heart. The bone marrow niche regulates hematopoietic stem cell proliferation and hence the systemic leukocyte pool, but whether cardiovascular disease affects the hematopoietic organ's microvasculature is unknown. Here we show that hypertension, atherosclerosis and myocardial infarction (MI) instigate endothelial dysfunction, leakage, vascular fibrosis and angiogenesis in the bone marrow, altogether leading to overproduction of inflammatory myeloid cells and systemic leukocytosis. Limiting angiogenesis with endothelial deletion of (encoding vascular endothelial growth factor (VEGF) receptor 2) curbed emergency hematopoiesis after MI. We noted that bone marrow endothelial cells assumed inflammatory transcriptional phenotypes in all examined stages of cardiovascular disease. Endothelial deletion of or (encoding versican), genes shown to be highly expressed in mice with atherosclerosis or MI, reduced hematopoiesis and systemic myeloid cell numbers in these conditions. Our findings establish that cardiovascular disease remodels the vascular bone marrow niche, stimulating hematopoiesis and production of inflammatory leukocytes.
Guarda su PubMed -
Mice on Balb/C Background Have Less Severe Cardiorespiratory Phenotype and SGLT2 Over-Expression Compared to 129x1/SvJ and C57Bl/6 Backgrounds.
Int J Mol Sci2022 Jun;23(12):. doi: 6674.
Irion Camila I, Williams Monique, Capcha Jose Condor, Eisenberg Trevor, Lambert Guerline, Takeuchi Lauro M, Seo Grace, Yousefi Keyvan, Kanashiro-Takeuchi Rosemeire, Webster Keith A, Young Karen C, Hare Joshua M, Shehadeh Lina A,
Abstract
Alport syndrome (AS) is a hereditary renal disorder with no etiological therapy. In the preclinical model of AS, disease progression and severity vary depending on mouse strain. The sodium-glucose cotransporter 2 (SGLT2) is emerging as an attractive therapeutic target in cardiac/renal pathologies, but its application to AS remains untested. This study investigates cardiorespiratory function and SGLT2 renal expression in mice from three different genetic backgrounds, 129x1/SvJ, C57Bl/6 and Balb/C. male 129x1/SvJ mice displayed alterations consistent with heart failure with preserved ejection fraction (HFpEF). Female, but not male, C57Bl/6 and Balb/C mice exhibited mild changes in systolic and diastolic function of the heart by echocardiography. Male C57Bl/6 mice presented systolic dysfunction by invasive hemodynamic analysis. All strains except Balb/C males demonstrated alterations in respiratory function. SGLT2 expression was significantly increased in AS compared to WT mice from all strains. However, cardiorespiratory abnormalities and SGLT2 over-expression were significantly less in AS Balb/C mice compared to the other two strains. Systolic blood pressure was significantly elevated only in mutant 129x1/SvJ mice. The results provide further evidence for strain-dependent cardiorespiratory and hypertensive phenotype variations in mouse AS models, corroborated by renal SGLT2 expression, and support ongoing initiatives to develop SGLT2 inhibitors for the treatment of AS.
Guarda su PubMed -
Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine.
Int J Mol Sci2022 Jun;23(12):. doi: 6480.
Matsuzaka Yasunari, Yashiro Ryu,
Abstract
Extracellular vesicles (EVs) are lipid bilayer membrane particles that play critical roles in intracellular communication through EV-encapsulated informative content, including proteins, lipids, and nucleic acids. Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal ability derived from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc., which they use to induce tissue regeneration by their direct recruitment into injured tissues, including the heart, liver, lung, kidney, etc., or secreting factors, such as or insulin-like growth factor. Recently, MSC-derived EVs have been shown to have regenerative effects against various diseases, partially due to the post-transcriptional regulation of target genes by miRNAs. Furthermore, EVs have garnered attention as novel drug delivery systems, because they can specially encapsulate various target molecules. In this review, we summarize the regenerative effects and molecular mechanisms of MSC-derived EVs.
Guarda su PubMed -
Effects of Sex, Training, and Maturity Status on the Cardiopulmonary and Muscle Deoxygenation Responses during Incremental Ramp Exercise.
Int J Environ Res Public Health2022 Jun;19(12):. doi: 7410.
Runacres Adam, Mackintosh Kelly, Evans Tim, McNarry Melitta A,
Abstract
Whilst participation in regular exercise and sport has generally increased over recent decades globally, fundamental questions remain regarding the influence of growth, maturation, and sex on the magnitude of training response throughout adolescence. Trained (108 participants, 43 girls; age: 14.3 ± 1.8 years) and untrained (108 participants, 43 girls; age: 14.7 ± 1.7 years) adolescents completed an incremental ramp test to exhaustion during which breath by gas exchange, beat-by-beat heart rate (HR), stroke volume (SV) and cardiac output (Q·) and muscle deoxygenation were assessed. Device-based physical activity was also assessed over seven consecutive days. Boys, irrespective of training status, had a significantly higher absolute (2.65 ± 0.70 L min vs. 2.01 ± 0.45 L min, < 0.01) and allometrically scaled (183.8 ± 31.4 mL·kg min vs. 146.5 ± 28.5 mL·kg min, < 0.01) peak oxygen uptake (V·O2) than girls. There were no sex differences in peak HR, SV or Q· but boys had a higher muscle deoxygenation plateau when expressed against absolute work rate and V·O2 ( < 0.05). Muscle deoxygenation appears to be more important in determining the sex differences in peak V·O2 in youth. Future research should examine the effects of sex on the response to different training methodologies in youth.
Guarda su PubMed -
Short-Chained Alcohols Make Membrane Surfaces Conducive for Melittin Action: Implication for the Physiological Role of Alcohols in Cells.
Cells2022 Jun;11(12):. doi: 1928.
Wang Haoyu, Qin Hao, Garab Gy?z?, Gasanoff Edward S,
Abstract
Alcohols are a part of cellular metabolism, but their physiological roles are not well understood. We investigated the effects of short-chain alcohols on and model membranes mimicking the lipid composition of eukaryotic inner mitochondrial membranes. We also studied the synergistic effects of alcohols with the bee venom membrane-active peptide, melittin, which is structurally similar to endogenous membrane-active peptides. The alcohols, from ethanol to octanol, gradually decreased the heart rate and the mitochondrial ATP synthesis of daphnia; in contrast, in combination with melittin, which exerted no sizeable effect, they gradually increased both the heart rate and the ATP synthesis. Lipid packing and the order parameter of oriented films, monitored by EPR spectroscopy of the spin-labeled probe 5-doxylstrearic acid, revealed gradual alcohol-assisted bilayer to non-bilayer transitions in the presence of melittin; further, while the alcohols decreased, in combination with melittin they increased the order parameter of the film, which is attributed to the alcohol-facilitated association of melittin with the membrane. A H-NMR spectroscopy of the liposomes confirmed the enhanced induction of a non-bilayer lipid phase that formed around the melittin, without the permeabilization of the liposomal membrane. Our data suggest that short-chain alcohols, in combination with endogenous peptides, regulate protein functions via modulating the lipid polymorphism of membranes.
Guarda su PubMed -
Acetyl-CoA metabolism drives epigenome change and contributes to carcinogenesis risk in fatty liver disease.
Genome Med2022 Jun;14(1):67. doi: 10.1186/s13073-022-01071-5.
Assante Gabriella, Chandrasekaran Sriram, Ng Stanley, Tourna Aikaterini, Chung Carolina H, Isse Kowsar A, Banks Jasmine L, Soffientini Ugo, Filippi Celine, Dhawan Anil, Liu Mo, Rozen Steven G, Hoare Matthew, Campbell Peter, Ballard J William O, Turner Nigel, Morris Margaret J, Chokshi Shilpa, Youngson Neil A,
Abstract
BACKGROUND:
The incidence of non-alcoholic fatty liver disease (NAFLD)-associated hepatocellular carcinoma (HCC) is increasing worldwide, but the steps in precancerous hepatocytes which lead to HCC driver mutations are not well understood. Here we provide evidence that metabolically driven histone hyperacetylation in steatotic hepatocytes can increase DNA damage to initiate carcinogenesis.
METHODS:
Global epigenetic state was assessed in liver samples from high-fat diet or high-fructose diet rodent models, as well as in cultured immortalized human hepatocytes (IHH cells). The mechanisms linking steatosis, histone acetylation and DNA damage were investigated by computational metabolic modelling as well as through manipulation of IHH cells with metabolic and epigenetic inhibitors. Chromatin immunoprecipitation and next-generation sequencing (ChIP-seq) and transcriptome (RNA-seq) analyses were performed on IHH cells. Mutation locations and patterns were compared between the IHH cell model and genome sequence data from preneoplastic fatty liver samples from patients with alcohol-related liver disease and NAFLD.
RESULTS:
Genome-wide histone acetylation was increased in steatotic livers of rodents fed high-fructose or high-fat diet. In vitro, steatosis relaxed chromatin and increased DNA damage marker ?H2AX, which was reversed by inhibiting acetyl-CoA production. Steatosis-associated acetylation and ?H2AX were enriched at gene clusters in telomere-proximal regions which contained HCC tumour suppressors in hepatocytes and human fatty livers. Regions of metabolically driven epigenetic change also had increased levels of DNA mutation in non-cancerous tissue from NAFLD and alcohol-related liver disease patients. Finally, genome-scale network modelling indicated that redox balance could be a key contributor to this mechanism.
CONCLUSIONS:
Abnormal histone hyperacetylation facilitates DNA damage in steatotic hepatocytes and is a potential initiating event in hepatocellular carcinogenesis.
© 2022. The Author(s).
Guarda su PubMed -
Optimizing haematopoietic stem and progenitor cell apheresis collection from plerixafor-mobilized patients with sickle cell disease.
Br J Haematol2022 Jun;():. doi: 10.1111/bjh.18311.
Sharma Akshay, Leonard Alexis, West Kamille, Gossett Jeffrey M, Uchida Naoya, Panch Sandhya, Stroncek David, Poston Leigh, Akel Salem, Hankins Jane S, Fitzhugh Courtney, Hsieh Matthew M, Kang Guolian, Tisdale John F, Weiss Mitchell J, Zheng Yan,
Abstract
We adjusted haematopoietic stem and progenitor cell (HSPC) apheresis collection from patients with sickle cell disease (SCD) by targeting deep buffy coat collection using medium or low collection preference (CP), and by increasing anticoagulant-citrate-dextrose-solution A dosage. In 43 HSPC collections from plerixafor-mobilized adult patients with SCD, we increased the collection efficiency to 35.79% using medium CP and 82.23% using low CP. Deep buffy coat collection increased red blood cell contamination of the HSPC product, the product haematocrit was 4.7% with medium CP and 6.4% with low CP. These adjustments were well-tolerated and allowed efficient HSPC collection from SCD patients.
© 2022 British Society for Haematology and John Wiley & Sons Ltd.
Guarda su PubMed -
Reduction of A-to-I RNA editing in the failing human heart regulates formation of circular RNAs.
Basic Res Cardiol2022 Jun;117(1):32. doi: 10.1007/s00395-022-00940-9.
Kokot Karoline E, Kneuer Jasmin M, John David, Rebs Sabine, Möbius-Winkler Maximilian N, Erbe Stephan, Müller Marion, Andritschke Michael, Gaul Susanne, Sheikh Bilal N, Haas Jan, Thiele Holger, Müller Oliver J, Hille Susanne, Leuschner Florian, Dimmeler Stefanie, Streckfuss-Bömeke Katrin, Meder Benjamin, Laufs Ulrich, Boeckel Jes-Niels,
Abstract
Alterations of RNA editing that affect the secondary structure of RNAs can cause human diseases. We therefore studied RNA editing in failing human hearts. Transcriptome sequencing showed that adenosine-to-inosine (A-to-I) RNA editing was responsible for 80% of the editing events in the myocardium. Failing human hearts were characterized by reduced RNA editing. This was primarily attributable to Alu elements in introns of protein-coding genes. In the failing left ventricle, 166 circRNAs were upregulated and 7 circRNAs were downregulated compared to non-failing controls. Most of the upregulated circRNAs were associated with reduced RNA editing in the host gene. ADAR2, which binds to RNA regions that are edited from A-to-I, was decreased in failing human hearts. In vitro, reduction of ADAR2 increased circRNA levels suggesting a causal effect of reduced ADAR2 levels on increased circRNAs in the failing human heart. To gain mechanistic insight, one of the identified upregulated circRNAs with a high reduction of editing in heart failure, AKAP13, was further characterized. ADAR2 reduced the formation of double-stranded structures in AKAP13 pre-mRNA, thereby reducing the stability of Alu elements and the circularization of the resulting circRNA. Overexpression of circAKAP13 impaired the sarcomere regularity of human induced pluripotent stem cell-derived cardiomyocytes. These data show that ADAR2 mediates A-to-I RNA editing in the human heart. A-to-I RNA editing represses the formation of dsRNA structures of Alu elements favoring canonical linear mRNA splicing and inhibiting the formation of circRNAs. The findings are relevant to diseases with reduced RNA editing and increased circRNA levels and provide insights into the human-specific regulation of circRNA formation.
© 2022. The Author(s).
Guarda su PubMed -
Application of Three-Dimensional Culture Method in the Cardiac Conduction System Research.
Methods Protoc2022 Jun;5(3):. doi: 50.
Mishra Abhishek, Pasumarthi Kishore B S,
Abstract
Congenital heart defects (CHD) are the most common type of birth defects. Several human case studies and genetically altered animal models have identified abnormalities in the development of ventricular conduction system (VCS) in the heart. While cell-based therapies hold promise for treating CHDs, translational efforts are limited by the lack of suitable in vitro models for feasibility and safety studies. A better understanding of cell differentiation pathways can lead to development of cell-based therapies for individuals living with CHD/VCS disorders. Here, we describe a new and reproducible 3-D cell culture method for studying cardiac cell lineage differentiation in vitro. We used primary ventricular cells isolated from embryonic day 11.5 (E11.5) mouse embryos, which can differentiate into multiple cardiac cell types including VCS cells. We compared 3-D cultures with three types of basement membrane extracts (BME) for their abilities to support E11.5 ventricular cell differentiation. In addition, the effects of atrial natriuretic peptide (ANP) and an inhibitor for its high affinity receptor were tested on cell differentiation in 3-D cultures. Following the cell culture, protocols for immunofluorescence imaging, cell extraction and protein isolation from the 3-D culture matrix and in-cell western methods are described. Further, these approaches can be used to study the effects of various ligands and genetic interventions on VCS cell development. We propose that these methodologies may also be extended for differentiation studies using other sources of stem cells such as induced pluripotent stem cells.
Guarda su PubMed
