Network


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

Hotspot


Dive into the research topics where Senthilkumar Muthusamy is active.

Publication


Featured researches published by Senthilkumar Muthusamy.


Aging Cell | 2011

Post-transcriptional regulation of IGF1R by key microRNAs in long-lived mutant mice

Ruqiang Liang; Amit Khanna; Senthilkumar Muthusamy; Na Li; Harshini Sarojini; John J. Kopchick; Michal M. Masternak; Andrzej Bartke; Eugenia Wang

Long‐lived mutant mice, both Ames dwarf and growth hormone receptor gene–disrupted or knockout strains, exhibit heightened cognitive robustness and altered IGF1 signaling in the brain. Here, we report, in both these long‐lived mice, that three up‐regulated lead microRNAs, miR‐470, miR‐669b, and miR‐681, are involved in posttranscriptional regulation of genes pertinent to growth hormone/IGF1 signaling. All three are most prominently localized in the hippocampus and correspond to reduced expression of key IGF1 signaling genes: IGF1, IGF1R, and PI3 kinase. The decline in these genes’ expression translates into decreased phosphorylation of downstream molecules AKT and FoxO3a. Cultures transfected with either miR‐470, miR‐669b, or miR‐681 show repressed endogenous expression of all three genes of the IGF1 signaling axis, most significantly IGF1R, while other similarly up‐regulated microRNAs, including let‐7g and miR‐509, do not induce the same levels of repression. Transduction study in IGF1‐responsive cell cultures shows significantly reduced IGF1R expression, and AKT to some extent, most notably by miR‐681. This is accompanied by decreased levels of downstream phosphorylated forms of AKT and FoxO3a upon IGF1 stimulation. Suppression of IGF1R by the three microRNAs is further validated by IGF1R 3′UTR reporter assays. Taken together, our results suggest that miR‐470, miR‐669b, and miR‐681 are all functionally able to suppress IGF1R and AKT, two upstream genes controlling FoxO3a phosphorylation status. Their up‐regulation in growth hormone signaling‐deficient mutant mouse brain suggests reduced IGF1 signaling at the posttranscriptional level, for numerous gains of neuronal function in these long‐lived mice.


Journal of Biological Chemistry | 2014

MicroRNA-539 is up-regulated in failing heart, and suppresses O-GlcNAcase expression.

Senthilkumar Muthusamy; Angelica M. DeMartino; Lewis J. Watson; Kenneth R. Brittian; Ayesha Zafir; Sujith Dassanayaka; Kyung U. Hong; Steven P. Jones

Background: Protein O-GlcNAcylation is nearly ubiquitous; however, regulation of the expression of key enzymes remains unknown. Results: miR-539 is up-regulated in the failing heart, binds to the 3′UTR, and negatively regulates O-GlcNAcase expression. Conclusion: Protein O-GlcNAcylation can be regulated by post-transcriptional mechanisms. Significance: miR-539 regulates one of the two enzymes responsible for O-GlcNAcylation in multicellular eukaryotes. Derangements in metabolism and related signaling pathways characterize the failing heart. One such signal, O-linked β-N-acetylglucosamine (O-GlcNAc), is an essential post-translational modification regulated by two enzymes, O-GlcNAc transferase and O-GlcNAcase (OGA), which modulate the function of many nuclear and cytoplasmic proteins. We recently reported reduced OGA expression in the failing heart, which is consistent with the pro-adaptive role of increased O-GlcNAcylation during heart failure; however, molecular mechanisms regulating these enzymes during heart failure remain unknown. Using miRNA microarray analysis, we observed acute and chronic changes in expression of several miRNAs. Here, we focused on miR-539 because it was predicted to target OGA mRNA. Indeed, co-transfection of the OGA-3′UTR containing reporter plasmid and miR-539 overexpression plasmid significantly reduced reporter activity. Overexpression of miR-539 in neonatal rat cardiomyocytes significantly suppressed OGA expression and consequently increased O-GlcNAcylation; conversely, the miR-539 inhibitor rescued OGA protein expression and restored O-GlcNAcylation. In conclusion, this work identifies the first target of miR-539 in the heart and the first miRNA that regulates OGA. Manipulation of miR-539 may represent a novel therapeutic target in the treatment of heart failure and other metabolic diseases.


Journal of Biological Chemistry | 2015

E2F1 Transcription Factor Regulates O-linked N-acetylglucosamine (O-GlcNAc) Transferase and O-GlcNAcase Expression.

Senthilkumar Muthusamy; Kyung U. Hong; Sujith Dassanayaka; Tariq Hamid; Steven P. Jones

Protein O-GlcNAcylation, which is controlled by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), has emerged as an important posttranslational modification that may factor in multiple diseases. Until recently, it was assumed that OGT/OGA protein expression was relatively constant. Several groups, including ours, have shown that OGT and/or OGA expression changes in several pathologic contexts, yet the cis and trans elements that regulate the expression of these enzymes remain essentially unexplored. Here, we used a reporter-based assay to analyze minimal promoters and leveraged in silico modeling to nominate several candidate transcription factor binding sites in both Ogt (i.e. the gene for OGT protein) and Mgea5 (i.e. the gene for OGA protein). We noted multiple E2F binding site consensus sequences in both promoters. We performed chromatin immunoprecipitation in both human and mouse cells and found that E2F1 bound to candidate E2F binding sites in both promoters. In HEK293 cells, we overexpressed E2F1, which significantly reduced OGT and MGEA5 expression. Conversely, E2F1-deficient mouse fibroblasts had increased Ogt and Mgea5 expression. Of the known binding partners for E2F1, we queried whether retinoblastoma 1 (Rb1) might be involved. Rb1-deficient mouse embryonic fibroblasts showed increased levels of Ogt and Mgea5 expression, yet overexpression of E2F1 in the Rb1-deficient cells did not alter Ogt and Mgea5 expression, suggesting that Rb1 is required for E2F1-mediated suppression. In conclusion, this work identifies and validates some of the promoter elements for mouse Ogt and Mgea5 genes. Specifically, E2F1 negatively regulates both Ogt and Mgea5 expression in an Rb1 protein-dependent manner.


Journal of Biological Chemistry | 2015

E2F1 transcription factor regulates O-GlcNAc transferase and O-GlcNAcase expression

Senthilkumar Muthusamy; Kyung U. Hong; Sujith Dassanayaka; Tariq Hamid; Steven P. Jones

Protein O-GlcNAcylation, which is controlled by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), has emerged as an important posttranslational modification that may factor in multiple diseases. Until recently, it was assumed that OGT/OGA protein expression was relatively constant. Several groups, including ours, have shown that OGT and/or OGA expression changes in several pathologic contexts, yet the cis and trans elements that regulate the expression of these enzymes remain essentially unexplored. Here, we used a reporter-based assay to analyze minimal promoters and leveraged in silico modeling to nominate several candidate transcription factor binding sites in both Ogt (i.e. the gene for OGT protein) and Mgea5 (i.e. the gene for OGA protein). We noted multiple E2F binding site consensus sequences in both promoters. We performed chromatin immunoprecipitation in both human and mouse cells and found that E2F1 bound to candidate E2F binding sites in both promoters. In HEK293 cells, we overexpressed E2F1, which significantly reduced OGT and MGEA5 expression. Conversely, E2F1-deficient mouse fibroblasts had increased Ogt and Mgea5 expression. Of the known binding partners for E2F1, we queried whether retinoblastoma 1 (Rb1) might be involved. Rb1-deficient mouse embryonic fibroblasts showed increased levels of Ogt and Mgea5 expression, yet overexpression of E2F1 in the Rb1-deficient cells did not alter Ogt and Mgea5 expression, suggesting that Rb1 is required for E2F1-mediated suppression. In conclusion, this work identifies and validates some of the promoter elements for mouse Ogt and Mgea5 genes. Specifically, E2F1 negatively regulates both Ogt and Mgea5 expression in an Rb1 protein-dependent manner.


Basic Research in Cardiology | 2017

Cardiomyocyte Ogt limits ventricular dysfunction in mice following pressure overload without affecting hypertrophy

Sujith Dassanayaka; Robert E. Brainard; Lewis J. Watson; Bethany W. Long; Kenneth R. Brittian; Angelica M. DeMartino; Allison L. Aird; Anna M. Gumpert; Timothy N. Audam; Peter J. Kilfoil; Senthilkumar Muthusamy; Tariq Hamid; Sumanth D. Prabhu; Steven P. Jones

The myocardial response to pressure overload involves coordination of multiple transcriptional, posttranscriptional, and metabolic cues. The previous studies show that one such metabolic cue, O-GlcNAc, is elevated in the pressure-overloaded heart, and the increase in O-GlcNAcylation is required for cardiomyocyte hypertrophy in vitro. Yet, it is not clear whether and how O-GlcNAcylation participates in the hypertrophic response in vivo. Here, we addressed this question using patient samples and a preclinical model of heart failure. Protein O-GlcNAcylation levels were increased in myocardial tissue from heart failure patients compared with normal patients. To test the role of OGT in the heart, we subjected cardiomyocyte-specific, inducibly deficient Ogt (i-cmOgt−/−) mice and Ogt competent littermate wild-type (WT) mice to transverse aortic constriction. Deletion of cardiomyocyte Ogt significantly decreased O-GlcNAcylation and exacerbated ventricular dysfunction, without producing widespread changes in metabolic transcripts. Although some changes in hypertrophic and fibrotic signaling were noted, there were no histological differences in hypertrophy or fibrosis. We next determined whether significant differences were present in i-cmOgt−/− cardiomyocytes from surgically naïve mice. Interestingly, markers of cardiomyocyte dedifferentiation were elevated in Ogt-deficient cardiomyocytes. Although no significant differences in cardiac dysfunction were apparent after recombination, it is possible that such changes in dedifferentiation markers could reflect a larger phenotypic shift within the Ogt-deficient cardiomyocytes. We conclude that cardiomyocyte Ogt is not required for cardiomyocyte hypertrophy in vivo; however, loss of Ogt may exert subtle phenotypic differences in cardiomyocytes that sensitize the heart to pressure overload-induced ventricular dysfunction.


PLOS ONE | 2015

O-GlcNAcylation Negatively Regulates Cardiomyogenic Fate in Adult Mouse Cardiac Mesenchymal Stromal Cells.

Ayesha Zafir; James A. Bradley; Bethany W. Long; Senthilkumar Muthusamy; Qianhong Li; Bradford G. Hill; Marcin Wysoczynski; Sumanth D. Prabhu; Aruni Bhatnagar; Roberto Bolli; Steven P. Jones

In both preclinical and clinical studies, cell transplantation of several cell types is used to promote repair of damaged organs and tissues. Nevertheless, despite the widespread use of such strategies, there remains little understanding of how the efficacy of cell therapy is regulated. We showed previously that augmentation of a unique, metabolically derived stress signal (i.e., O-GlcNAc) improves survival of cardiac mesenchymal stromal cells; however, it is not known whether enhancing O-GlcNAcylation affects lineage commitment or other aspects of cell competency. In this study, we assessed the role of O-GlcNAc in differentiation of cardiac mesenchymal stromal cells. Exposure of these cells to routine differentiation protocols in culture increased markers of the cardiomyogenic lineage such as Nkx2.5 and connexin 40, and augmented the abundance of transcripts associated with endothelial and fibroblast cell fates. Differentiation significantly decreased the abundance of O-GlcNAcylated proteins. To determine if O-GlcNAc is involved in stromal cell differentiation, O-GlcNAcylation was increased pharmacologically during the differentiation protocol. Although elevated O-GlcNAc levels did not significantly affect fibroblast and endothelial marker expression, acquisition of cardiomyocyte markers was limited. In addition, increasing O-GlcNAcylation further elevated smooth muscle actin expression. In addition to lineage commitment, we also evaluated proliferation and migration, and found that increasing O-GlcNAcylation did not significantly affect either; however, we found that O-GlcNAc transferase—the protein responsible for adding O-GlcNAc to proteins—is at least partially required for maintaining cellular proliferative and migratory capacities. We conclude that O-GlcNAcylation contributes significantly to cardiac mesenchymal stromal cell lineage and function. O-GlcNAcylation and pathological conditions that may affect O-GlcNAc levels (such as diabetes) should be considered carefully in the context of cardiac cell therapy.


Mechanisms of Ageing and Development | 2011

Increased expression of miR-34a and miR-93 in rat liver during aging, and their impact on the expression of Mgst1 and Sirt1

Na Li; Senthilkumar Muthusamy; Ruqiang Liang; Harshini Sarojini; Eugenia Wang


Aging (Albany NY) | 2011

Gain of survival signaling by down-regulation of three key miRNAs in brain of calorie-restricted mice

Amit Khanna; Senthilkumar Muthusamy; Ruqiang Liang; Harshini Sarojini; Eugenia Wang


Biochemical Journal | 2015

High glucose induces mitochondrial dysfunction independently of protein O-GlcNAcylation

Sujith Dassanayaka; Ryan Readnower; Joshua K. Salabei; Bethany W. Long; Allison L. Aird; Yuting Zheng; Senthilkumar Muthusamy; Heberty T. Facundo; Bradford G. Hill; Steven P. Jones


Journal of the American College of Cardiology | 2017

Myocardial Reparative Properties of Cardiac Mesenchymal Cells Isolated on the Basis of Adherence

Marcin Wysoczynski; Yiru Guo; Joseph B. Moore; Senthilkumar Muthusamy; Qianhong Li; Marjan Nasr; Hong Li; Yibing Nong; Wen-Jian Wu; Alex Tomlin; Xiaoping Zhu; Gregory N. Hunt; Anna M. Gumpert; Michael Book; Abdur Rahman Khan; Xian-Liang Tang; Roberto Bolli

Collaboration


Dive into the Senthilkumar Muthusamy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eugenia Wang

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyung U. Hong

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qianhong Li

University of Louisville

View shared research outputs
Researchain Logo
Decentralizing Knowledge