Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shashi Kumar Gupta is active.

Publication


Featured researches published by Shashi Kumar Gupta.


Circulation | 2011

MicroRNA-24 Regulates Vascularity After Myocardial Infarction

Jan Fiedler; Virginija Jazbutyte; Bettina C. Kirchmaier; Shashi Kumar Gupta; Johan M. Lorenzen; Dorothee Hartmann; Paolo Galuppo; Susanne Kneitz; John T.G. Pena; Cherin Sohn-Lee; Xavier Loyer; Juergen Soutschek; Thomas Brand; Thomas Tuschl; Joerg Heineke; Ulrich Martin; Stefan Schulte-Merker; Georg Ertl; Stefan Engelhardt; Johann Bauersachs; Thomas Thum

Background— Myocardial infarction leads to cardiac remodeling and development of heart failure. Insufficient myocardial capillary density after myocardial infarction has been identified as a critical event in this process, although the underlying mechanisms of cardiac angiogenesis are mechanistically not well understood. Methods and Results— Here, we show that the small noncoding RNA microRNA-24 (miR-24) is enriched in cardiac endothelial cells and considerably upregulated after cardiac ischemia. MiR-24 induces endothelial cell apoptosis, abolishes endothelial capillary network formation on Matrigel, and inhibits cell sprouting from endothelial spheroids. These effects are mediated through targeting of the endothelium-enriched transcription factor GATA2 and the p21-activated kinase PAK4, which were identified by bioinformatic predictions and validated by luciferase gene reporter assays. Respective downstream signaling cascades involving phosphorylated BAD (Bcl-XL/Bcl-2–associated death promoter) and Sirtuin1 were identified by transcriptome, protein arrays, and chromatin immunoprecipitation analyses. Overexpression of miR-24 or silencing of its targets significantly impaired angiogenesis in zebrafish embryos. Blocking of endothelial miR-24 limited myocardial infarct size of mice via prevention of endothelial apoptosis and enhancement of vascularity, which led to preserved cardiac function and survival. Conclusions— Our findings indicate that miR-24 acts as a critical regulator of endothelial cell apoptosis and angiogenesis and is suitable for therapeutic intervention in the setting of ischemic heart disease.


Nature Communications | 2012

The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy

Ahmet Ucar; Shashi Kumar Gupta; Jan Fiedler; Erdem Erikci; Kardasinski M; Sandor Batkai; Seema Dangwal; Regalla Kumarswamy; Claudia Bang; Angelika Holzmann; Janet Remke; Caprio M; Jentzsch C; Stefan Engelhardt; Geisendorf S; Glas C; Thomas G. Hofmann; Nessling M; Richter K; Schiffer M; Lars Christian Napp; Johann Bauersachs; Kamal Chowdhury; Thomas Thum

Pathological growth of cardiomyocytes (hypertrophy) is a major determinant for the development of heart failure, one of the leading medical causes of mortality worldwide. Here we show that the microRNA (miRNA)-212/132 family regulates cardiac hypertrophy and autophagy in cardiomyocytes. Hypertrophic stimuli upregulate cardiomyocyte expression of miR-212 and miR-132, which are both necessary and sufficient to drive the hypertrophic growth of cardiomyocytes. MiR-212/132 null mice are protected from pressure-overload-induced heart failure, whereas cardiomyocyte-specific overexpression of the miR-212/132 family leads to pathological cardiac hypertrophy, heart failure and death in mice. Both miR-212 and miR-132 directly target the anti-hypertrophic and pro-autophagic FoxO3 transcription factor and overexpression of these miRNAs leads to hyperactivation of pro-hypertrophic calcineurin/NFAT signalling and an impaired autophagic response upon starvation. Pharmacological inhibition of miR-132 by antagomir injection rescues cardiac hypertrophy and heart failure in mice, offering a possible therapeutic approach for cardiac failure.


Journal of Molecular and Cellular Cardiology | 2011

Diagnostic and prognostic impact of six circulating microRNAs in acute coronary syndrome

Christian Widera; Shashi Kumar Gupta; Johan M. Lorenzen; Claudia Bang; Johann Bauersachs; Kerstin Bethmann; Tibor Kempf; Kai C. Wollert; Thomas Thum

Circulating microRNAs may have diagnostic potential in acute coronary syndrome (ACS). Previous studies, however, were based on low patient numbers and could not assess the relation of microRNAs to clinical characteristics and their potential prognostic value. We thus assessed the diagnostic and prognostic value of cardiomyocyte-enriched microRNAs in the context of clinical variables and a sensitive myonecrosis biomarker in a larger ACS cohort. MiR-1, miR-133a, miR-133b, miR-208a, miR-208b, and miR-499 concentrations were measured by quantitative reverse transcription PCR in plasma samples obtained on admission from 444 patients with ACS. High-sensitivity troponin T (hsTnT) was measured by immunoassay. Patients were followed for 6 months regarding all-cause mortality. In a multiple linear regression analysis that included clinical variables and hsTnT, miR-1, miR-133a, miR-133b, and miR-208b were independently associated with hsTnT levels (all P<0.001). Patients with myocardial infarction presented with higher levels of miR-1, miR-133a, and miR-208b compared with patients with unstable angina. However, all six investigated microRNAs showed a large overlap between patients with unstable angina or myocardial infarction. MiR-133a and miR-208b levels were significantly associated with the risk of death in univariate and age- and gender-adjusted analyses. Both microRNAs lost their independent association with outcome upon further adjustment for hsTnT. The present study tempers speculations about the potential usefulness of cardiomyocyte-enriched microRNAs as diagnostic or prognostic markers in ACS.


Circulation-cardiovascular Genetics | 2010

Circulating MicroRNAs as Biomarkers and Potential Paracrine Mediators of Cardiovascular Disease

Shashi Kumar Gupta; Claudia Bang; Thomas Thum

MicroRNAs (miRNAs) comprise a class of small, noncoding RNAs that control expression of complementary target mRNAs. Dysregulation of intracellular miRNA expression has been described in various diseases, including a number of cardiovascular conditions. Functional studies have shown a role for miRNAs in cardiac fibrosis, hypertrophy, angiogenesis, and heart failure.1,–,3 These findings suggest a new therapeutic entry point for cardiac disease and illustrate the broad therapeutic potential of miRNA modulation. Initial reports have detected circulating extracellular miRNAs in the serum/plasma of patients with cancer.4 Despite the existence of RNases, miRNAs remain stable in serum and other body fluids. One explanation is the inclusion of miRNAs into lipid or lipoprotein complexes such as exosomes5 or microvesicles.6 Subsequently, altered concentrations of miRNAs have been found in patients with various cardiovascular diseases. Here, we review the current knowledge about circulating miRNAs during coronary artery disease (CAD), myocardial infarction, and heart failure (Table). A further new and exciting function of circulating miRNAs in the cardiovascular system may be their potential to serve as paracrine signaling molecules.7 The detection of circulating miRNAs in serum/plasma8,9 suggests that miRNAs may fulfill biological functions outside the cell and serve as potential biomarkers for diseases. Circulating miRNAs are protected from RNase-dependent degradation by several mechanisms, including their inclusion in microvesicles, exosomes, and apoptotic bodies as well as through the formation of protein-miRNA complexes resistant to degradation (Figure). In addition, circulating miRNAs are quite stable even after multiple freeze-thaw cycles.8 Recent studies have shown that miRNAs are actively secreted in microvesicles or exosomes from different cell types.6,10 Despite the current knowledge about the existence of circulating miRNAs and the intercellular transfer of miRNAs from donor …


Circulation Research | 2010

Short Communication: Asymmetric Dimethylarginine Impairs Angiogenic Progenitor Cell Function in Patients With Coronary Artery Disease Through a MicroRNA-21–Dependent Mechanism

Felix Fleissner; Virginija Jazbutyte; Jan Fiedler; Shashi Kumar Gupta; Xiaoke Yin; Qingbo Xu; Paolo Galuppo; Susanne Kneitz; Manuel Mayr; Georg Ertl; Johann Bauersachs; Thomas Thum

Rationale: The endogenous nitric oxide synthase inhibitor asymmetrical dimethylarginine (ADMA) is increased in patients with coronary artery disease and may regulate function of circulating angiogenic progenitor cells (APCs) by small regulatory RNAs. Objectives: To study the role of microRNAs in ADMA-mediated impairment of APCs. Methods and Results: By using microarray analyses, we established microRNA expression profiles of human APCs. We used ADMA to induce APC dysfunction and found 16 deregulated microRNAs. We focused on miR-21, which was 3-fold upregulated by ADMA treatment. Overexpression of miR-21 in human APCs impaired migratory capacity. To identify regulated miR-21 targets, we used proteome analysis, using difference in-gel electrophoresis followed by mass spectrometric analysis of regulated proteins. We found that transfection of miR-21 precursors significantly repressed superoxide dismutase 2 in APCs, which resulted in increased intracellular reactive oxygen species concentration and impaired nitric oxide bioavailability. MiR-21 further repressed sprouty-2, leading to Erk Map kinase–dependent reactive oxygen species formation and APC migratory defects. Small interference RNA–mediated superoxide dismutase 2 or sprouty-2 reduction also increased reactive oxygen species formation and impaired APC migratory capacity. ADMA-mediated reactive oxygen species formation and APC dysfunction was rescued by miR-21 blockade. APCs from patients with coronary artery disease and high ADMA plasma levels displayed >4-fold elevated miR-21 levels, low superoxide dismutase 2 expression, and impaired migratory capacity, which could be normalized by miR-21 antagonism. Conclusions: We identified a novel miR-21–dependent mechanism of ADMA-mediated APC dysfunction. MiR-21 antagonism therefore emerges as an interesting strategy to improve dysfunctional APCs in patients with coronary artery disease.


Clinical Journal of The American Society of Nephrology | 2011

Circulating miR-210 Predicts Survival in Critically Ill Patients with Acute Kidney Injury

Johan M. Lorenzen; Jan T. Kielstein; Carsten Hafer; Shashi Kumar Gupta; Philipp Kümpers; Robert Faulhaber-Walter; Hermann Haller; Danilo Fliser; Thomas Thum

BACKGROUND AND OBJECTIVES MicroRNAs (miRNAs) are small ribonucleotides regulating gene expression. MicroRNAs are present in the blood in a remarkably stable form. We tested whether circulating miRNAs in the plasma of critically ill patients with acute kidney injury (AKI) at the inception of renal replacement therapy are deregulated and may predict survival. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We profiled miRNAs using RNA isolated from the plasma of patients with AKI and healthy controls. The results were validated in 77 patients with acute kidney injury, 30 age-matched healthy controls, and 18 critically ill patients with acute myocardial infarction by quantitative real-time PCR. RESULTS Circulating levels of miR-16 and miR-320 were downregulated in the plasma of kidney injury AKI patients, whereas miR-210 was upregulated compared with healthy controls (all P < 0.0001) and disease controls (miR-210 and miR-16: P < 0.0001; miR-320: P = 0.03). Cox regression (P < 0.05) and Kaplan-Meier curve analysis (P = 0.03) revealed miR-210 as an independent and powerful predictor of 28-day survival. CONCLUSIONS Circulating miRNAs are altered in patients with kidney injury AKI. MiR-210 predicts mortality in this patient cohort and may serve as a novel biomarker AKI reflecting pathophysiological changes on a cellular level.


Basic Research in Cardiology | 2012

Role of miR-21 in the pathogenesis of atrial fibrosis.

Oliver Adam; Björn Löhfelm; Thomas Thum; Shashi Kumar Gupta; Sarah-Lena Puhl; Hans-Joachim Schäfers; Michael Böhm; Ulrich Laufs

Atrial fibrosis is important for the pathogenesis of atrial fibrillation (AF) but the underlying signal transduction is incompletely understood. We therefore studied the role of microRNA-21 (miR-21) and its downstream target Sprouty 1 (Spry1) during atrial fibrillation. Left atria (LA) from patients with AF showed a 2.5-fold increased expression of miR-21 compared to matched LA of patients in sinus rhythm. Increased miR-21 expression correlated positively with atrial collagen content and was associated with a reduced protein expression of Spry1 and increased expression of connective tissue growth factor (CTGF), lysyl oxidase and Rac1-GTPase. Neonatal cardiac fibroblasts treated with angiotensin II (AngII) or CTGF showed an increased miR-21 and decreased Spry1 expression. Pretreatment with an inhibitor of Rac1 GTPase, NSC23766, reduced the AngII-induced upregulation of miR-21. A small molecule inhibitor of lysyl oxidase, BAPN, prevented the AngII as well as the CTGF-induced miR-21 expression. Transgenic mice with cardiac overexpression of Rac1, which develop spontaneous AF and atrial fibrosis with increasing age, showed upregulation of miR-21 expression associated with reduced Spry1 expression. miR-21 expression and signalling in vivo were prevented by long-term treatment of the mice with statins. Direct inhibition of miR-21 by antagomir-21 prevented fibrosis of the atrial myocardium post-myocardial infarction. Left atria of patients with atrial fibrillation are characterized by upregulation of miR-21 und reduced expression of Spry1. Activation of Rac1 by angiotensin II leads to a CTGF- and lysyl oxidase-mediated increase of miR-21 expression contributing to structural remodelling of the atrial myocardium.


European Journal of Heart Failure | 2015

MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure.

Chris Watson; Shashi Kumar Gupta; Eoin O'Connell; Sabrina Thum; Nadezhda Glezeva; Jasmin Fendrich; Joe Gallagher; Mark Ledwidge; Lea Grote-Levi; Kenneth McDonald; Thomas Thum

Differentiation of heart failure with reduced (HFrEF) or preserved (HFpEF) ejection fraction independent of echocardiography is challenging in the community. Diagnostic strategies based on monitoring circulating microRNA (miRNA) levels may prove to be of clinical value in the near future. The aim of this study was to identify a novel miRNA signature that could be a useful HF diagnostic tool and provide valuable clinical information on whether a patient has HFrEF or HFpEF.


European Heart Journal | 2015

Osteopontin is indispensible for AP1-mediated angiotensin II-related miR-21 transcription during cardiac fibrosis

Johan M. Lorenzen; Celina Schauerte; Anika Hübner; Malte Kölling; Filippo Martino; Kristian Scherf; Sandor Batkai; Karina Zimmer; Ariana Foinquinos; Tamás Kaucsár; Jan Fiedler; Regalla Kumarswamy; Claudia Bang; Dorothee Hartmann; Shashi Kumar Gupta; Jan T. Kielstein; Andreas Jungmann; Hugo A. Katus; Frank Weidemann; Oliver J. Müller; Hermann Haller; Thomas Thum

Aims Osteopontin (OPN) is a multifunctional cytokine critically involved in cardiac fibrosis. However, the underlying mechanisms are unresolved. Non-coding RNAs are powerful regulators of gene expression and thus might mediate this process. Methods and results OPN and miR-21 were significantly increased in cardiac biopsies of patients with myocardial fibrosis. Ang II infusion via osmotic minipumps led to specific miRNA regulations with miR-21 being strongly induced in wild-type (WT) but not OPN knockout (KO) mice. This was associated with enhanced cardiac collagen content, myofibroblast activation, ERK-MAP kinase as well as AKT signalling pathway activation and a reduced expression of Phosphatase and Tensin Homologue (PTEN) as well as SMAD7 in WT but not OPN KO mice. In contrast, cardiotropic AAV9-mediated overexpression of OPN in vivo further enhanced cardiac fibrosis. In vitro, Ang II induced expression of miR-21 in WT cardiac fibroblasts, while miR-21 levels were unchanged in OPN KO fibroblasts. As pri-miR-21 was also increased by Ang II, we studied potential involved upstream regulators; Electrophoretic Mobility Shift and Chromatin Immunoprecipitation analyses confirmed activation of the miR-21 upstream-transcription factor AP-1 by Ang II. Recombinant OPN directly activated miR-21, enhanced fibrosis, and activated the phosphoinositide 3-kinase pathway. Locked nucleic acid-mediated miR-21 silencing ameliorated cardiac fibrosis development in vivo. Conclusion In cardiac fibrosis related to Ang II, miR-21 is transcriptionally activated and targets PTEN/SMAD7 resulting in increased fibroblast survival. OPN KO animals are protected from miR-21 increase and fibrosis development due to impaired AP-1 activation and fibroblast activation.


Clinical Chemistry | 2015

Circulating Long Noncoding RNA TapSAKI Is a Predictor of Mortality in Critically Ill Patients with Acute Kidney Injury

Johan M. Lorenzen; Celina Schauerte; Jan T. Kielstein; Anika Hübner; Filippo Martino; Jan Fiedler; Shashi Kumar Gupta; Robert Faulhaber-Walter; Regalla Kumarswamy; Carsten Hafer; Hermann Haller; Danilo Fliser; Thomas Thum

BACKGROUND Long noncoding RNAs (lncRNAs) are novel intracellular noncoding ribonucleotides regulating gene expression. Intriguingly, these RNA transcripts are detectable and stable in the blood of patients with cancer and cardiovascular disease. We tested whether circulating lncRNAs in plasma of critically ill patients with acute kidney injury (AKI) at inception of renal replacement therapy were deregulated and might predict survival. METHODS We performed a global lncRNA expression analysis using RNA isolated from plasma of patients with AKI, healthy controls, and ischemic disease controls. This global screen revealed several deregulated lncRNAs in plasma samples of patients with AKI. lncRNA-array-based alterations were confirmed in kidney biopsies of patients as well as in plasma of 109 patients with AKI, 30 age-matched healthy controls, and 30 disease controls by quantitative real-time PCR. RESULTS Circulating concentrations of the novel intronic antisense lncRNA TrAnscript Predicting Survival in AKI (TapSAKI) (P < 0.0001) were detectable in kidney biopsies and upregulated in plasma of patients with AKI. Cox regression and Kaplan-Meier curve analysis revealed TapSAKI as an independent predictor of 28-day survival (P < 0.01). TapSAKI was enriched in tubular epithelial cells subjected to ATP depletion (P = 0.03). CONCLUSIONS The alteration of circulating concentrations of lncRNAs in patients with AKI supports TapSAKI as a predictor of mortality in this patient cohort.

Collaboration


Dive into the Shashi Kumar Gupta's collaboration.

Top Co-Authors

Avatar

Thomas Thum

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Jan Fiedler

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudia Bang

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Johann Bauersachs

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge