Pubblicazioni recenti - cardiac fibroblast
-
Human cardiac fibroblasts produce pro-inflammatory cytokines upon TLRs and RLRs stimulation.
Mol Cell Biochem2021 Apr;():. doi: 10.1007/s11010-021-04157-7.
Li Zhe, Nguyen Tuan T, Valaperti Alan,
Abstract
Heart inflammation is one of the major causes of heart damage that leads to dilated cardiomyopathy and often progresses to end-stage heart failure. In the present study, we aimed to assess whether human cardiac cells could release immune mediators upon stimulation of Toll-like receptors (TLRs) and Retinoic acid-inducible gene (RIG)-I-like receptors (RLRs).Commercially available human cardiac fibroblasts and an immortalized human cardiomyocyte cell line were stimulated in vitro with TLR2, TLR3, and TLR4 agonists. In addition, cytosolic RLRs were activated in cardiac cells after transfection of polyinosinic-polycytidylic acid (PolyIC). Upon stimulation of TLR3, TLR4, MDA5, and RIG-I, but not upon stimulation of TLR2, human cardiac fibroblasts produced high amounts of the pro-inflammatory cytokines IL-6 and IL-8. On the contrary, the immortalized human cardiomyocyte cell line was unresponsive to the tested TLRs agonists. Upon RLRs stimulation, cardiac fibroblasts, and to a lesser extent the cardiomyocyte cell line, induced anti-viral IFN-? expression.These data demonstrate that human cardiac fibroblasts and an immortalized human cardiomyocyte cell line differently respond to various TLRs and RLRs ligands. In particular, human cardiac fibroblasts were able to induce pro-inflammatory and anti-viral cytokines on their own. These aspects will contribute to better understand the immunological function of the different cell populations that make up the cardiac tissue.
Guarda su PubMed -
Multiplexed targeting of miRNA-210 in stem cell-derived extracellular vesicles promotes selective regeneration in ischemic hearts.
Exp Mol Med2021 Apr;():. doi: 10.1038/s12276-021-00584-0.
Song Byeong-Wook, Lee Chang Youn, Kim Ran, Kim Won Jung, Lee Hee Won, Lee Min Young, Kim Jongmin, Jeong Jee-Yeong, Chang Woochul,
Abstract
Extracellular vesicles (EVs) are cell derivatives containing diverse cellular molecules, have various physiological properties and are also present in stem cells used for regenerative therapy. We selected a "multiplexed target" that demonstrates multiple effects on various cardiovascular cells, while functioning as a cargo of EVs. We screened various microRNAs (miRs) and identified miR-210 as a candidate target for survival and angiogenic function. We confirmed the cellular and biological functions of EV-210 (EVs derived from ASC) secreted from adipose-derived stem cells (ASCs) transfected with miR-210 (ASC). Under hypoxic conditions, we observed that ASC inhibits apoptosis by modulating protein tyrosine phosphatase 1B (PTP1B) and death-associated protein kinase 1 (DAPK1). In hypoxic endothelial cells, EV-210 exerted its angiogenic capacity by inhibiting Ephrin A (EFNA3). Furthermore, EV-210 enhanced cell survival under the control of PTP1B and induced antiapoptotic effects in hypoxic H9c2 cells. In cardiac fibroblasts, the fibrotic ratio was reduced after exposure to EV-210, but EVs derived from ASC did not communicate with fibroblasts. Finally, we observed the functional restoration of the ischemia/reperfusion-injured heart by maintaining the intercommunication of EVs and cardiovascular cells derived from ASC. These results suggest that the multiplexed target with ASC is a useful tool for cardiovascular regeneration.
Guarda su PubMed -
Characterisation of cardiac health in the reduced uterine perfusion pressure model and a 3D cardiac spheroid model, of preeclampsia.
Biol Sex Differ2021 Apr;12(1):31. doi: 10.1186/s13293-021-00376-1.
Richards Claire, Sesperez Kimberly, Chhor Michael, Ghorbanpour Sahar, Rennie Claire, Ming Clara Liu Chung, Evenhuis Chris, Nikolic Valentina, Orlic Natasa Karadzov, Mikovic Zeljko, Stefanovic Milan, Cakic Zoran, McGrath Kristine, Gentile Carmine, Bubb Kristen, McClements Lana,
Abstract
BACKGROUND:
Preeclampsia is a dangerous cardiovascular disorder of pregnancy that leads to an increased risk of future cardiovascular and metabolic disorders. Much of the pathogenesis and mechanisms involved in cardiac health in preeclampsia are unknown. A novel anti-angiogenic protein, FKBPL, is emerging as having a potential role in both preeclampsia and cardiovascular disease (CVD). Therefore, in this study we aimed to characterise cardiac health and FKBPL regulation in the rat reduced uterine perfusion pressure (RUPP) and a 3D cardiac spheroid model of preeclampsia.
METHODS:
The RUPP model was induced in pregnant rats and histological analysis performed on the heart, kidney, liver and placenta (n ? 6). Picrosirius red staining was performed to quantify collagen I and III deposition in rat hearts, placentae and livers as an indicator of fibrosis. RT-qPCR was used to determine changes in Fkbpl, Icam1, Vcam1, Flt1 and Vegfa mRNA in hearts and/or placentae and ELISA to evaluate cardiac brain natriuretic peptide (BNP45) and FKBPL secretion. Immunofluorescent staining was also conducted to analyse the expression of cardiac FKBPL. Cardiac spheroids were generated using human cardiac fibroblasts and human coronary artery endothelial cells and treated with patient plasma from normotensive controls, early-onset preeclampsia (EOPE) and late-onset preeclampsia (LOPE); n = 3. FKBPL and CD31 expression was quantified by immunofluorescent labelling.
RESULTS:
The RUPP procedure induced significant increases in blood pressure (p < 0.001), collagen deposition (p < 0.001) and cardiac BNP45 (p < 0.05). It also induced a significant increase in cardiac FKBPL mRNA (p < 0.05) and protein expression (p < 0.01). RUPP placentae also exhibited increased collagen deposition and decreased Flt1 mRNA expression (p < 0.05). RUPP kidneys revealed an increase in average glomerular size (p < 0.05). Cardiac spheroids showed a significant increase in FKBPL expression when treated with LOPE plasma (p < 0.05) and a trend towards increased FKBPL expression following treatment with EOPE plasma (p = 0.06).
CONCLUSIONS:
The rat RUPP model induced cardiac, renal and placental features reflective of preeclampsia. FKBPL was increased in the hearts of RUPP rats and cardiac spheroids treated with plasma from women with preeclampsia, perhaps reflective of restricted angiogenesis and inflammation in this disorder. Elucidation of these novel FKBPL mechanisms in cardiac health in preeclampsia could be key in preventing future CVD.
Guarda su PubMed -
Fibroblast GATA-4 and GATA-6 promote myocardial adaptation to pressure overload by enhancing cardiac angiogenesis.
Basic Res Cardiol2021 Apr;116(1):26. doi: 10.1007/s00395-021-00862-y.
Dittrich Gesine M, Froese Natali, Wang Xue, Kroeger Hannah, Wang Honghui, Szaroszyk Malgorzata, Malek-Mohammadi Mona, Cordero Julio, Keles Merve, Korf-Klingebiel Mortimer, Wollert Kai C, Geffers Robert, Mayr Manuel, Conway Simon J, Dobreva Gergana, Bauersachs Johann, Heineke Joerg,
Abstract
Heart failure due to high blood pressure or ischemic injury remains a major problem for millions of patients worldwide. Despite enormous advances in deciphering the molecular mechanisms underlying heart failure progression, the cell-type specific adaptations and especially intercellular signaling remain poorly understood. Cardiac fibroblasts express high levels of cardiogenic transcription factors such as GATA-4 and GATA-6, but their role in fibroblasts during stress is not known. Here, we show that fibroblast GATA-4 and GATA-6 promote adaptive remodeling in pressure overload induced cardiac hypertrophy. Using a mouse model with specific single or double deletion of Gata4 and Gata6 in stress activated fibroblasts, we found a reduced myocardial capillarization in mice with Gata4/6 double deletion following pressure overload, while single deletion of Gata4 or Gata6 had no effect. Importantly, we confirmed the reduced angiogenic response using an in vitro co-culture system with Gata4/6 deleted cardiac fibroblasts and endothelial cells. A comprehensive RNA-sequencing analysis revealed an upregulation of anti-angiogenic genes upon Gata4/6 deletion in fibroblasts, and siRNA mediated downregulation of these genes restored endothelial cell growth. In conclusion, we identified a novel role for the cardiogenic transcription factors GATA-4 and GATA-6 in heart fibroblasts, where both proteins act in concert to promote myocardial capillarization and heart function by directing intercellular crosstalk.
Guarda su PubMed -
Nuclear mechanosensing drives chromatin remodelling in persistently activated fibroblasts.
Nat Biomed Eng2021 Apr;():. doi: 10.1038/s41551-021-00709-w.
Walker Cierra J, Crocini Claudia, Ramirez Daniel, Killaars Anouk R, Grim Joseph C, Aguado Brian A, Clark Kyle, Allen Mary A, Dowell Robin D, Leinwand Leslie A, Anseth Kristi S,
Abstract
Fibrotic disease is caused by the continuous deposition of extracellular matrix by persistently activated fibroblasts (also known as myofibroblasts), even after the resolution of the injury. Using fibroblasts from porcine aortic valves cultured on hydrogels that can be softened via exposure to ultraviolet light, here we show that increased extracellular stiffness activates the fibroblasts, and that cumulative tension on the nuclear membrane and increases in the activity of histone deacetylases transform transiently activated fibroblasts into myofibroblasts displaying condensed chromatin with genome-wide alterations. The condensed structure of the myofibroblasts is associated with cytoskeletal stability, as indicated by the inhibition of chromatin condensation and myofibroblast persistence after detachment of the nucleus from the cytoskeleton via the displacement of endogenous nesprins from the nuclear envelope. We also show that the chromatin structure of myofibroblasts from patients with aortic valve stenosis is more condensed than that of myofibroblasts from healthy donors. Our findings suggest that nuclear mechanosensing drives distinct chromatin signatures in persistently activated fibroblasts.
Guarda su PubMed -
Fibroblast-Specific Proteo-Transcriptomes Reveal Distinct Fibrotic Signatures of Human Sinoatrial Node in Non-Failing and Failing Hearts.
Circulation2021 Apr;():. doi: 10.1161/CIRCULATIONAHA.120.051583.
Kalyanasundaram Anuradha, Li Ning, Gardner Miranda L, Artiga Esthela J, Hansen Brian J, Webb Amy, Freitas Michael A, Pietrzak Maciej, Whitson Bryan A, Mokadam Nahush A, Janssen Paul M L, Mohler Peter J, Fedorov Vadim V,
Abstract
Up to fifty percent of the adult human sinoatrial node (SAN), is composed of dense connective tissue. Cardiac diseases including heart failure (HF) may further increase fibrosis within the SAN pacemaker complex, leading to impaired automaticity and conduction of electrical activity to the atria. However, unlike the role of cardiac fibroblasts in pathological fibrotic remodeling and tissue repair, nothing is known about fibroblasts that maintain the inherently fibrotic SAN environment. Intact SAN pacemaker complex was dissected from cardioplegically arrested explanted non-failing (non-HF, n=22; 48.7±3.1y.o,) and HF human hearts (n=16; 54.9±2.6y.o.). Connective tissue content was quantified from Masson's trichrome stained head-center and center-tail SAN sections. Expression of extracellular matrix (ECM) proteins, including Collagens 1, 3A1, cartilage intermediate layer protein 1 (CILP1) and periostin, fibroblast and myofibroblast numbers were quantified by in situ and in vitro immunolabeling. Fibroblasts from the central intramural SAN pacemaker compartment (~10x5x2 mm) and right atria (RA) were isolated, cultured, passaged once, and treated ±transforming growth factor beta-1 (TGF?1) and subjected to comprehensive high-throughput next-generation sequencing of whole transcriptome, microRNA and proteomic analyses. Intranodal fibrotic content was significantly higher in SAN pacemaker complex from HF vs non-HF hearts (57.7±2.6% vs 44.0±1.2% <0.0001). Proliferating phosphorylated histone3/vimentin/CD31 fibroblasts were higher in HF SAN. Vimentin/alpha smooth muscle actin/CD31 myofibroblasts along with increased interstitial periostin expression were found only in HF SAN. RNA sequencing and proteomic analyses identified unique differences in mRNA, long non-coding RNA, microRNA and proteomic profiles between non-HF and HF SAN and RA fibroblasts, and TGF?1-induced myofibroblasts. Specifically, proteins and signaling pathways associated with ECM flexibility, stiffness, focal adhesion and metabolism were altered in HF SAN fibroblasts compared to non-HF SAN. This study revealed increased SAN-specific fibrosis with presence of myofibroblasts, CILP1 and periostin-positive interstitial fibrosis only in HF vs non-HF human hearts. Comprehensive proteo-transcriptomic profiles of SAN fibroblasts identified upregulation of genes and proteins promoting stiffer SAN ECM in HF hearts. Fibroblast-specific profiles generated by our proteo-transcriptomic analyses of the human SAN, provide a comprehensive framework for future studies to investigate the role of SAN-specific fibrosis in cardiac rhythm regulation and arrhythmias.
Guarda su PubMed -
Direct Reprogramming of Human Fibroblasts into Myoblasts to Investigate Therapies for Neuromuscular Disorders.
J Vis Exp2021 Apr;(170):. doi: 10.3791/61991.
Almeida Camila F, Frair Emma C, Huang Nianyuan, Neinast Reid, McBride Kim L, Weiss Robert B, Flanigan Kevin M, Wein Nicolas,
Abstract
Investigations into both the pathophysiology and therapeutic targets in muscular dystrophies have been hampered by the limited proliferative capacity of human myoblasts. Several mouse models have been created but they either do not truly represent the human physiopathology of the disease or are not representative of the broad spectrum of mutations found in humans. The immortalization of human primary myoblasts is an alternative to this limitation; however, it is still dependent on muscle biopsies, which are invasive and not easily available. In contrast, skin biopsies are easier to obtain and less invasive to patients. Fibroblasts derived from skin biopsies can be immortalized and transdifferentiated into myoblasts, providing a source of cells with excellent myogenic potential. Here, we describe a fast and direct reprogramming method of fibroblast into a myogenic lineage. Fibroblasts are transduced with two lentiviruses: hTERT to immortalize the primary culture and a tet-inducible MYOD, which upon the addition of doxycycline, induces the conversion of fibroblasts into myoblasts and then mature myotubes, which express late differentiation markers. This quick transdifferentiation protocol represents a powerful tool to investigate pathological mechanisms and to investigate innovative gene-based or pharmacological biotherapies for neuromuscular disorders.
Guarda su PubMed -
Krüppel-Like Factor 15 Modulates Signaling-Mediated Inflammatory Response Contributing to Angiotensin II-Induced Cardiac Remodeling.
Front Cell Dev Biol2021 ;9():644954. doi: 10.3389/fcell.2021.644954.
He Shun, Lu Yuanyuan, Guo Yuetong, Li Shijin, Lu Xiao, Shao Shuai, Zhou Handan, Wang Ruiqi, Wang Jiguang, Gao Pingjin, Li Xiaodong,
Abstract
Inflammation is involved in cardiac remodeling. In response to pathological stimuli, activated cardiac fibroblasts (CFs) secreting inflammatory cytokines and chemokines play an important role in monocyte/macrophage recruitment. However, the precise mechanism of CF-mediated inflammatory response in hypertension-induced cardiac remodeling remains unclear. In the present study, we investigated the role of transcription factor Krüppel-like factor 15 (KLF15) in this process. We found that KLF15 expression decreased while chemokine and its receptor expression increased in the hearts of angiotensin II (Ang II)-infused mice. Compared to the wild-type mice, KLF15 knockout (KO) mice aggravated Ang II-induced cardiac hypertrophy and fibrosis. Deficiency of KLF15 promoted macrophage accumulation, increase of and expression, and mTOR, ERK1/2, NF-?B-p65 signaling activation in the hearts. Mechanistically, Ang II dose- dependently decreased KLF15 expression and increased secretion from cardiac fibroblasts but not cardiac myoblasts. Loss- or gain-of-function studies have shown that KLF15 negatively regulated expression through its transactivation domain (TAD). Intriguingly, the adenovirus-mediated full length of KLF15-but not KLF15 with TAD deletion overexpression-markedly prevented pathological change in Ang II-infused mice. Notably, the administration of inhibitor SB265610 reversed KLF15 knockout-mediated aggravation of cardiac dysfunction, remodeling, and inflammation induced by Ang II. In conclusion, our study identifies that KLF15 in cardiac fibroblasts negatively regulates axis-mediated inflammatory response and subsequent cardiac remodeling in hypertension.
Copyright © 2021 He, Lu, Guo, Li, Lu, Shao, Zhou, Wang, Wang, Gao and Li.
Guarda su PubMed -
Targeting HIF-? for robust prevascularization of human cardiac organoids.
J Tissue Eng Regen Med2021 Feb;15(2):189-202. doi: 10.1002/term.3165.
Coyle Robert C, Barrs Ryan W, Richards Dylan J, Ladd Emma P, Menick Donald R, Mei Ying,
Abstract
Prevascularized 3D microtissues have been shown to be an effective cell delivery vehicle for cardiac repair. To this end, our lab has explored the development of self-organizing, prevascularized human cardiac organoids by co-seeding human cardiomyocytes with cardiac fibroblasts, endothelial cells, and stromal cells into agarose microwells. We hypothesized that this prevascularization process is facilitated by the endogenous upregulation of hypoxia-inducible factor (HIF) pathway in the avascular 3D microtissues. In this study, we used Molidustat, a selective PHD (prolyl hydroxylase domain enzymes) inhibitor that stabilizes HIF-?, to treat human cardiac organoids, which resulted in 150 ± 61% improvement in endothelial expression (CD31) and 220 ± 20% improvement in the number of lumens per organoids. We hypothesized that the improved endothelial expression seen in Molidustat treated human cardiac organoids was dependent upon upregulation of VEGF, a well-known downstream target of HIF pathway. Through the use of immunofluorescent staining and ELISA assays, we determined that Molidustat treatment improved VEGF expression of non-endothelial cells and resulted in improved co-localization of supporting cell types and endothelial structures. We further demonstrated that Molidustat treated human cardiac organoids maintain cardiac functionality. Lastly, we showed that Molidustat treatment improves survival of cardiac organoids when exposed to both hypoxic and ischemic conditions . For the first time, we demonstrate that targeted HIF-? stabilization provides a robust strategy to improve endothelial expression and lumen formation in cardiac microtissues, which will provide a powerful framework for prevascularization of various microtissues in developing successful cell transplantation therapies.
Guarda su PubMed -
Interference of TGF-?1/Smad7 signal pathway affects myocardial fibrosis in hypertension.
Pak J Pharm Sci2020 Nov;33(6):2625-2631.
Xiao Huaping, Li Bingda, Yang Xiaomin, Yin Qiulin,
Abstract
Hypertension can cause myocardial fibrosis, during which tumor growth factor-beta 1 (TGF-?1) can facilitate myocardial cell proliferation and transition towards myofibroblast (MFB). Smad7 is a negative regulator of TGF-?1/Smads signal pathway. This study used hypertension rat model to investigate the regulatory role of TGF-?1/Smad7 signal pathway in myocardial fibrosis. Rat renal hypertension model was established to test collagen volume fraction, myocardial hydroxyl proline content and COL1A1 expression as well as the expression of TGF-?1 and Smad7. The expressions of TGF-?1, Smad7, COLA1A1 and ?-SMA at certain generations (P2, P4 and P6) were measured in cultured human cardiac fibroblast (HCF) during differentiation towards MFB differentiation. P6 generation HCF was transfected with pIRES2-EGFP-Smad7 and pIRES2-EGFP-blank followed by measuring expressions of TGF-?1, Smad7, COLA1A1 and ?-SMA. Hydroxyl-proline content and collagen volume fraction were compared between Ad-NC and Ad-Smad7 injection groups. Hypertensive rats had significantly elevated collagen volume fraction, hydroxyl proline contents, and expression of COLA1 and TGF-?1 than Sham group, whilst Smad7 expression was lower. With increased cell passage, HCF showed gradually increased TGF-?1, COL1A1 and ?-SMA expression, plus decreased Smad7 expression. Over-expression of Smad7 remarkably decreased COLA1 and ?-SMA expression in HCF. Tail vein injection of Ad-Smad7 decreased both hydroxyl proline and collagen volume fraction. Elevated TGF-?1 expression and decreased Smad7 expression are found in fibrotic myocardial tissues of hypertensive rats. Over-expression of Smad7 inhibits differentiation of HCF towards MFB cells, thus decreasing myocardial fibrosis in hypertensive rats.
Guarda su PubMed -
Magnolol, a natural aldehyde dehydrogenase-2 agonist, inhibits the proliferation and collagen synthesis of cardiac fibroblasts.
Bioorg Med Chem Lett2021 Apr;():128045. doi: S0960-894X(21)00271-7.
Chen Ling, Wu Yu-Ting, Gu Xuan-Ye, Xie Ling-Peng, Fan Hui-Jie, Tan Zhang-Bin, Zhang Wen-Tong, Chen Hong-Mei, Li Jun, Huang Gui-Qiong, Liu Bin, Zhou Ying-Chun, Sun Xiao-Min,
Abstract
Inhibiting myocardial fibrosis can help prevent cardiovascular diseases, including heart failure. Magnolol (Mag), a natural component of Magnoliae officinalis, has been reported to inhibit fibrosis. However, the mechanism of Mag activity and its effects on myocardial fibrosis remain unclear. Here, we investigated the involvement of ALDH2, an endogenous protective agent against myocardial fibrosis, in the Mag-mediated inhibition of cardiac fibroblast proliferation and collagen synthesis. We found that Mag significantly inhibited cardiac fibroblast proliferation and collagen synthesis, based on the results of MTT, EdU and western blot assays. Moreover, molecular docking, molecular dynamics simulation and surface plasmon resonance (SPR) assays showed that Mag could bind directly and stably to ALDH2. Further analysis of the mechanism of these effects indicated that treatment with Mag dose-dependently enhanced ALDH2 activity without altering protein expression. Mag could enhance the activity of recombinant human ALDH2 proteins with a half-maximal effective concentration of 5.79 × 10 M. In addition, ALDH2 activation via Alda-1 inhibited cardiac fibroblast proliferation and collagen synthesis, while ALDH2 inhibition via daidzin partially blocked the suppressive effects of Mag. In summary, Mag may act as a natural ALDH2 agonist and inhibit cardiac fibroblast proliferation and collagen synthesis.
Copyright © 2021. Published by Elsevier Ltd.
Guarda su PubMed -
Proteome characterisation of extracellular vesicles isolated from heart.
Proteomics2021 Apr;():e2100026. doi: 10.1002/pmic.202100026.
Claridge Bethany, Rai Alin, Fang Haoyun, Matsumoto Aya, Luo Jieting, McMullen Julie R, Greening David W,
Abstract
Cardiac intercellular communication is critical for heart function and often dysregulated in cardiovascular diseases. While cardiac extracellular vesicles (cEVs) are emerging mediators of signalling, their isolation remains a technical challenge hindering our understanding of cEV protein composition. Here, we utilised Langendorff-collagenase-based enzymatic perfusion and differential centrifugation to isolate cEVs from mouse heart (yield 3-6 ?g/heart). cEVs are ?200 nm, express classical EV markers (Cd63/81/9 , Tsg101 , Pdcd6ip/Alix ), and are depleted of blood (Alb/Fga/Hba) and cardiac damage markers (Mb, Tnnt2, Ldhb). Comparison with mechanically-derived EVs revealed greater detection of EV markers and decreased cardiac damage contaminants. Mass spectrometry-based proteomic profiling revealed 1,721 proteins in cEVs, implicated in proteasomal and autophagic proteostasis, glycolysis and fatty acid metabolism; essential functions often disrupted in cardiac pathologies. There was striking enrichment of 942 proteins in cEVs compared to mouse heart tissue - implicated in EV biogenesis, antioxidant activity, and lipid transport, suggesting active cargo selection and specialised function. Interestingly, cEVs contain marker proteins for cardiomyocytes, cardiac progenitors, B-cells, T-cells, macrophages, smooth muscle cells, endothelial cells, and cardiac fibroblasts, suggesting diverse cellular origin. We present a method of cEV isolation and provide insight into potential functions, enabling future studies into EV roles in cardiac physiology and disease. This article is protected by copyright. All rights reserved.
This article is protected by copyright. All rights reserved.
Guarda su PubMed -
Imaging of cardiac fibroblast activation in a patient after acute myocardial infarction using Ga-FAPI-04.
J Nucl Cardiol2021 Apr;():. doi: 10.1007/s12350-021-02603-z.
Notohamiprodjo Susan, Nekolla Stephan G, Robu Stephanie, Villagran Asiares Alberto, Kupatt Christian, Ibrahim Tareq, Laugwitz Karl-Ludwig, Makowski Marcus R, Schwaiger Markus, Weber Wolfgang A, Varasteh Zohreh,
Abstract
Our previous study has demonstrated the feasibility of noninvasive imaging of fibroblast activation protein (FAP)-expression after myocardial infarction (MI) in MI-territory in a rat model with Ga-FAPI-04-PET. In the current extended clinical case, we sought to delineate cardiac uptake of Ga-FAPI-04 in a patient after MI with clinical indication for the evidence of fibroblast activation. Carcinoma patients without cardiac disease underwent Ga-FAPI-04-PET/CT as control. The patient with one-vessel disease underwent dynamic Ga-FAPI-04-cardiac-PET/CMR for 60 minutes. Correlation of cardiac Ga-FAPI-04 uptake with clinical findings, ECG, echocardiography, coronary-arteriography and enhanced cardiac-MRI with T1 MOLLI and ECV mapping were performed. No uptake was found in normal myocardium and in mature scar. A focal intense Ga-FAPI-04 uptake with continuous wash-out in the infarct territory of coronary occlusion correlating with T1 and ECV mapping was observed. The uptake of Ga-FAPI-04 extends beyond the actual infarcted area and overestimates the infarct size as confirmed by follow-up CMR.
Guarda su PubMed -
Carbon monoxide activates large-conductance calcium-activated potassium channels of human cardiac fibroblasts through various mechanisms.
Korean J Physiol Pharmacol2021 May;25(3):227-237. doi: 10.4196/kjpp.2021.25.3.227.
Bae Hyemi, Kim Taeho, Lim Inja,
Abstract
Carbon monoxide (CO) is a cardioprotectant and potential cardiovascular therapeutic agent. Human cardiac fibroblasts (HCFs) are important determinants of myocardial structure and function. Large-conductance Ca-activated K (BK) channel is a potential therapeutic target for cardiovascular disease. We investigated whether CO modulates BK channels and the signaling pathways in HCFs using whole-cell mode patch-clamp recordings. CO-releasing molecules (CORMs; CORM-2 and CORM-3) significantly increased the amplitudes of BK currents (). The CO-induced stimulating effects on were blocked by pre-treatment with specific nitric oxide synthase (NOS) blockers (L-N-monomethyl arginine citrate and L-N-nitroarginine methyl ester). 8-bromo-cyclic GMP increased . KT5823 (inhibits PKG) or ODQ (inhibits soluble guanylate cyclase) blocked the CO-stimulating effect on . Moreover, 8-bromo-cyclic AMP also increased , and pre-treatment with KT5720 (inhibits PKA) or SQ22536 (inhibits adenylate cyclase) blocked the CO effect. Pre-treatment with Nethylmaleimide (a thiol-alkylating reagent) also blocked the CO effect on , and DLdithiothreitol (a reducing agent) reversed the CO effect. These data suggest that CO activates through NO the NOS and through the PKG, PKA, and -nitrosylation pathways.
Guarda su PubMed -
Aldosterone and Mineralocorticoid Receptor Signaling as Determinants of Cardiovascular and Renal Injury: From Hans Selye to the Present.
Am J Nephrol2021 Apr;():1-8. doi: 10.1159/000515622.
Epstein Murray,
Abstract
BACKGROUND:
A full understanding of the mechanisms of action of aldosterone and its interaction with the mineralocorticoid receptor (MR) allows a theoretical framework to predict the therapeutic potential of MR antagonists (MRAs) in CKD, and heart failure with reduced ejection fraction.
SUMMARY:
The initial focus on the mechanisms of action of aldosterone was directed primarily on its role in modulating renal excretory function. In contrast, many recent studies have demonstrated a wider and expanded role for aldosterone in modulating inflammation, collagen formation, fibrosis, and necrosis. Increasing evidence has accrued that implicates the pathophysiological overactivation of the MR as a major determinant of progression of CKD. By promoting inflammation and fibrosis, MR overactivation constitutes a pivotal determinant of CKD progression and its associated morbidity and mortality. In accord with this mechanism of action, blockade of the MR is currently being investigated as a novel treatment regimen to slow the progression of CKD. The recently reported FIDELIO-DKD (FInerenone in reducing kiDnEy faiLure and dIsease prOgression in Diabetic Kidney Disease) study demonstrated that patients with CKD and type 2 diabetes who were treated with finerenone (a novel nonsteroidal MRA) manifested a lower risk of a composite primary outcome event compared with patients in the placebo arm (defined as kidney failure, or a sustained decrease of ?40% in the estimated glomerular filtration rate from baseline, or death from renal causes). In addition, patients in the finerenone group also manifested a lower risk of a key secondary outcome event (defined as death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure). Key Messages: Based on the success of the FIDELIO-DKD study, future studies should be implemented testing the hypothesis that a wide array of nondiabetic CKD is modulated by overactivation of the MR, and consequently may be amenable to treatment with novel nonsteroidal MRAs. Future studies are encouraged to elucidate the clinical implications of the interplay of nonsteroidal MRAs and the components of the renin-angiotensin cascade. The unique and recently reported interrelationship of fibroblast growth factor (FGF23) and aldosterone may also constitute a propitious subject for future investigation.
© 2021 S. Karger AG, Basel.
Guarda su PubMed -
Orphan nuclear receptor ERR-? regulates hepatic FGF23 production in acute kidney injury.
Proc Natl Acad Sci U S A2021 Apr;118(16):. doi: e2022841118.
Radhakrishnan Kamalakannan, Kim Yong-Hoon, Jung Yoon Seok, Kim Don-Kyu, Na Soon-Young, Lim Daejin, Kim Dong Hun, Kim Jina, Kim Hyung-Seok, Choy Hyon E, Cho Sung Jin, Lee In-Kyu, Ayvaz ?amil, Nittka Stefanie, Fliser Danilo, Schunk Stefan J, Speer Thimoteus, Dooley Steven, Lee Chul-Ho, Choi Hueng-Sik,
Abstract
Fibroblast growth factor 23 (FGF23), a hormone generally derived from bone, is important in phosphate and vitamin D homeostasis. In acute kidney injury (AKI) patients, high-circulating FGF23 levels are associated with disease progression and mortality. However, the organ and cell type of FGF23 production in AKI and the molecular mechanism of its excessive production are still unidentified. For insight, we investigated folic acid (FA)-induced AKI in mice. Interestingly, simultaneous with FGF23, orphan nuclear receptor ERR-? expression is increased in the liver of FA-treated mice, and ectopic overexpression of ERR-? was sufficient to induce hepatic FGF23 production. In patients and in mice, AKI is accompanied by up-regulated systemic IL-6, which was previously identified as an upstream regulator of ERR-? expression in the liver. Administration of IL-6 neutralizing antibody to FA-treated mice or of recombinant IL-6 to healthy mice confirms IL-6 as an upstream regulator of hepatic ERR-?-mediated FGF23 production. A significant ( < 0.001) interconnection between high IL-6 and FGF23 levels as a predictor of AKI in patients that underwent cardiac surgery was also found, suggesting the clinical relevance of the finding. Finally, liver-specific depletion of ERR-? or treatment with an inverse ERR-? agonist decreased hepatic FGF23 expression and plasma FGF23 levels in mice with FA-induced AKI. Thus, inverse agonist of ERR-? may represent a therapeutic strategy to reduce adverse plasma FGF23 levels in AKI.
Guarda su PubMed -
An association between fibroblast growth factor 21 and cognitive impairment in iron-overload thalassemia.
Sci Rep2021 Apr;11(1):8057. doi: 10.1038/s41598-021-87597-x.
Theerajangkhaphichai Wasan, Sripetchwandee Jirapas, Sriwichaiin Sirawit, Svasti Saovaros, Chattipakorn Nipon, Tantiworawit Adisak, Chattipakorn Siriporn C,
Abstract
Although an increased fibroblast growth factor 21 (FGF21) level was related to mild cognitive impairment (MCI) in metabolic syndrome patients, any association regarding FGF21 and MCI in thalassemia patients as well as mechanistic insight are questionable. Therefore, the objectives of this study were: (1) to investigate the prevalence and associative risk factors of MCI in thalassemia patients, (2) to evaluate the association between levels of FGF21 and MCI in thalassemia patients, and (3) to investigate brain FGF21 signaling in iron-overload thalassemia. Thalassemia patients were enrolled onto the study (n?=?131). Montreal cognitive assessment (MoCA) was used to determine cognitive performance. Plasma FGF21 level was determined in all patients. Iron-overload ?-thalassemic (HT) mice were used to investigate brain FGF21 level and signaling, the expression of synaptic proteins, and Alzheimer's like pathology. We found that 70% of thalassemia patients developed MCI. FGF21 level was positively correlated with the MCI. Interestingly, brain FGF21 resistance, as indicated by increased brain FGF21 levels with impaired FGF21 signaling, was found in iron-overload HT mice. The reduced synaptic protein expression and increased Alzheimer's like pathology were also observed. These suggest that FGF21 may play a role in MCI in thalassemia patients.
Guarda su PubMed -
Retrospective analysis of factors associated with serum levels of fibroblast growth factor-21 in patients with diabetes.
Ann Palliat Med2021 Mar;10(3):3258-3266. doi: 10.21037/apm-21-525.
Ren Fang, Huang Jiafang, Dai Tianran, Gan Feng,
Abstract
BACKGROUND:
Fibroblast growth factor-21 (FGF-21) plays multiple roles in pathophysiological processes of the human body. Previous studies have evidenced FGF-21 to be an inhibitor of vascular calcification through a variety of mechanisms. Increased levels of serum FGF-21 are known to be associated with an elevated risk of coronary heart disease; however, the factors affecting the expression of FGF-21 are currently unclear. This study aimed to observe the effects of some medications and other factors on serum FGF-21 levels in patients with diabetes.
METHODS:
Baseline characteristics of patients with diabetes, including body mass index (BMI), medication, level of exercise, and other information, were collected and analyzed, and their baseline levels of serum FGF-21 were tested. The relationship of serum FGF-21 levels with these characteristics was analyzed.
RESULTS:
A total of 2118 patients were enrolled in the final analysis. Results revealed that the serum levels of FGF-21 in patients with a high BMI were elevated compared to those in patients with a normal or low BMI. Furthermore, the serum levels of FGF-21 in patients who engaged in regular exercise were higher than those in patients who exercised intermittently or not at all. No significant differences existed between patients who received different anti-diabetic drugs, or between patients treated with different antihyperlipidemic drugs. Also, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers had no obvious effects on serum levels of FGF-21 in patients with diabetes.
CONCLUSIONS:
Drugs used in the treatment of patients with diabetes have no significant effects on serum levels of FGF-21. Obese patients had higher serum levels of FGF-21 than did non-obese patients. Participating in sports might increase the levels of FGF-21 in patients with diabetes.
Guarda su PubMed -
Direct conversion of adult human fibroblasts into functional endothelial cells using defined factors.
Biomaterials2021 Mar;272():120781. doi: S0142-9612(21)00137-X.
Han Jung-Kyu, Shin Youngchul, Sohn Min-Hwan, Choi Saet-Byeol, Shin Dasom, You Youngrang, Shin Jong-Yeon, Seo Jeong-Sun, Kim Hyo-Soo,
Abstract
We aimed to directly convert adult human dermal fibroblasts (aHDFs) into functional endothelial cells (ECs). Lentiviral vectors encoding endothelial transcription factors (TFs) were constructed. We examined whether five TFs (FOXO1, ER71, KLF2, TAL1, and LMO2) used for the generation of mouse induced ECs (iECs) could convert the aHDFs into human iECs. Twenty-eight days after transduction with lentiviral constructs, 32.1 ± 5.1% cells expressed vascular endothelial (VE)-cadherin. Factor screening revealed that only three factors (3F: ER71, KLF2, and TAL1) were necessary to induce VE-cadherin (+) cells (49.4 ± 3.5%). However, whole transcriptome sequencing showed that VE-cadherin (+) cells were not completely reprogrammed. Mature iECs double-positive for VE-cadherin/Pecam1 (DP cells) with a cobblestone appearance were obtained at a frequency of only 5.1 ± 0.6%. Using whole transcriptome analysis, the potential factors that could block the conversion were screened. Among candidates TWIST1-knockdown enhanced efficiency of conversion. Rosiglitazone, an inhibitor of epithelial-mesenchymal transition (EMT), also improved the conversion efficiency. Moreover, a 2nd second-stage conversion process, in which VE-cadherin (+) cells were incubated for additional two weeks, further enhanced the efficiency. The final protocol for 6 weeks yielded a conversion rate of 19.6 ± 3.0% iECs, defined by DP cells depicting the nature of mature ECs in various analyses. Further analyses revealed that the genetic and epigenetic profiles of iECs resembled those of functional ECs. Collectively, aHDFs can be converted into functional ECs through the transduction of ER71, KLF2, and TAL1, combined with two EMT inhibitors (siTWIST1 and rosiglitazone), followed by 2nd stage conversion.
Copyright © 2021 Elsevier Ltd. All rights reserved.
Guarda su PubMed -
Donor cell memory confers a metastable state of directly converted cells.
Cell Stem Cell2021 Apr;():. doi: S1934-5909(21)00074-6.
Kim Kee-Pyo, Li Cui, Bunina Daria, Jeong Hyun-Woo, Ghelman Julia, Yoon Juyong, Shin Borami, Park Hongryeol, Han Dong Wook, Zaugg Judith B, Kim Johnny, Kuhlmann Tanja, Adams Ralf H, Noh Kyung-Min, Goldman Steven A, Schöler Hans R,
Abstract
Generation of induced oligodendrocyte progenitor cells (iOPCs) from somatic fibroblasts is a strategy for cell-based therapy of myelin diseases. However, iOPC generation is inefficient, and the resulting iOPCs exhibit limited expansion and differentiation competence. Here we overcome these limitations by transducing an optimized transcription factor combination into a permissive donor phenotype, the pericyte. Pericyte-derived iOPCs (PC-iOPCs) are stably expandable and functionally myelinogenic with high differentiation competence. Unexpectedly, however, we found that PC-iOPCs are metastable so that they can produce myelination-competent oligodendrocytes or revert to their original identity in a context-dependent fashion. Phenotypic reversion of PC-iOPCs is tightly linked to memory of their original transcriptome and epigenome. Phenotypic reversion can be disconnected from this donor cell memory effect, and in vivo myelination can eventually be achieved by transplantation of O4 pre-oligodendrocytes. Our data show that donor cell source and memory can contribute to the fate and stability of directly converted cells.
Copyright © 2021 Elsevier Inc. All rights reserved.
Guarda su PubMed
