Network


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

Hotspot


Dive into the research topics where Jeffrey Schmeckpeper is active.

Publication


Featured researches published by Jeffrey Schmeckpeper.


Circulation Research | 2013

C3orf58, a Novel Paracrine Protein, Stimulates Cardiomyocyte Cell-Cycle Progression Through the PI3K–AKT–CDK7 Pathway

Farideh Beigi; Jeffrey Schmeckpeper; James Alan Payne; Lunan Zhang; Zhiping Zhang; Jing Huang; Maria Mirotsou; Victor J. Dzau

Rationale: The regenerative capacity of the heart is markedly diminished shortly after birth, coinciding with overall withdrawal of cardiomyocytes from cell cycle. Consequently, the adult mammalian heart has limited capacity to regenerate after injury. The discovery of factors that can induce cardiomyocyte proliferation is, therefore, of high interest and has been the focus of extensive investigation throughout the past years. Objective: We have recently identified C3orf58 as a novel hypoxia and Akt induced stem cell factor (HASF) secreted from mesenchymal stem cells, which can promote cardiac repair through cytoprotective mechanisms. Here, we tested the hypothesis that HASF can also contribute to cardiac regeneration by stimulating cardiomyocyte division and proliferation. Methods and Results: Neonatal ventricular cardiomyocytes were stimulated in culture for 7 days with purified recombinant HASF protein. Compared with control untreated cells, HASF-treated neonatal cardiomyocytes exhibited 60% increase in DNA synthesis as measured by bromodeoxyuridine incorporation. These results were confirmed by immunofluorescence confocal microscopy showing a 50% to 100% increase in the number of cardiomyocytes in the mitotic and cytokinesis phases. Importantly, in vivo cardiac overexpression of HASF in a transgenic mouse model resulted in enhanced level of DNA synthesis and cytokinesis in neonatal and adult cardiomyocytes. These proliferative effects were modulated by a phosphoinositide 3-kinase–protein kinase B–cycle-dependent kinase 7 pathway as revealed by the use of phosphoinositide 3-kinase -pathway–specific inhibitors and silencing of the Cdk7 gene. Conclusions: Our studies support the hypothesis that HASF induces cardiomyocyte proliferation via a phosphoinositide 3-kinase–protein kinase B–cycle-dependent kinase 7 pathway. The implications of this finding may be significant for cardiac regeneration biology and therapeutics.


Journal of The American Society of Nephrology | 2013

Adult Renal Mesenchymal Stem Cell–Like Cells Contribute to Juxtaglomerular Cell Recruitment

Hao Wang; Jose A. Gomez; Sabine Klein; Zhiping Zhang; Barbara Seidler; Yanqiang Yang; Jeffrey Schmeckpeper; Lunan Zhang; Garrett G. Muramoto; John P. Chute; Richard E. Pratt; Dieter Saur; Maria Mirotsou; Victor J. Dzau

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Stem Cells | 2013

Abi3bp Is a Multifunctional Autocrine/Paracrine Factor that Regulates Mesenchymal Stem Cell Biology†

Conrad P. Hodgkinson; Vinogran Naidoo; Karl Patti; Jose A. Gomez; Jeffrey Schmeckpeper; Zhiping Zhang; Bryce H. Davis; Richard E. Pratt; Maria Mirotsou; Victor J. Dzau

Mesenchymal stem cells (MSCs) transplanted into injured myocardium promote repair through paracrine mechanisms. We have previously shown that MSCs over‐expressing AKT1 (Akt‐MSCs) exhibit enhanced properties for cardiac repair. In this study, we investigated the relevance of Abi3bp toward MSC biology. Abi3bp formed extracellular deposits with expression controlled by Akt1 and ubiquitin‐mediated degradation. Abi3bp knockdown/knockout stabilized focal adhesions and promoted stress‐fiber formation. Furthermore, MSCs from Abi3bp knockout mice displayed severe deficiencies in osteogenic and adipogenic differentiation. Knockout or stable knockdown of Abi3bp increased MSC and Akt‐MSC proliferation, promoting S‐phase entry via cyclin‐d1, ERK1/2, and Src. Upon Abi3bp binding to integrin‐β1 Src associated with paxillin which inhibited proliferation. In vivo, Abi3bp knockout increased MSC number and proliferation in bone marrow, lung, and liver. In summary, we have identified a novel extracellular matrix protein necessary for the switch from proliferation to differentiation in MSCs. STEM Cells 2013;31:1669–1682


Journal of Molecular and Cellular Cardiology | 2015

Inhibition of Wnt6 by Sfrp2 regulates adult cardiac progenitor cell differentiation by differential modulation of Wnt pathways

Jeffrey Schmeckpeper; Amanda Verma; Lucy Yin; Farideh Beigi; Lunan Zhang; Alan Payne; Zhiping Zhang; Richard E. Pratt; Victor J. Dzau; Maria Mirotsou

Wnt signaling has recently emerged as an important regulator of cardiac progenitor cell proliferation and differentiation, but the exact mechanisms by which Wnt signaling modulates these effects are not known. Understanding these mechanisms is essential for advancing our knowledge of cardiac progenitor cell biology and applying this knowledge to enhance cardiac therapy. Here, we explored the effects of Sfrp2, a canonical Wnt inhibitor, in adult cardiac progenitor cell (CPC) differentiation and investigated the molecular mechanisms involved. Our data show that Sfrp2 treatment can promote differentiation of CPCs after ischemia-reperfusion injury. Treatment of CPCs with Sfrp2 inhibited CPC proliferation and primed them for cardiac differentiation. Sfrp2 binding to Wnt6 and inhibition of Wnt6 canonical pathway was essential for the inhibition of CPC proliferation. This inhibition of Wnt6 canonical signaling by Sfrp2 was important for activation of the non-canonical Wnt/Planar Cell Polarity (PCP) pathway through JNK, which in turn induced expression of cardiac transcription factors and CPC differentiation. Taken together, these results demonstrate a novel role of Sfrp2 and Wnt6 in regulating the dynamic process of CPC proliferation and differentiation, as well as providing new insights into the mechanisms of Wnt signaling in cardiac differentiation.


Journal of Molecular and Cellular Cardiology | 2018

Insights from molecular signature of in vivo cardiac c-Kit(+) cells following cardiac injury and β-catenin inhibition

Conrad P. Hodgkinson; Jose A. Gomez; Syeda Samara Baksh; Alan Payne; Jeffrey Schmeckpeper; Richard E. Pratt; Victor J. Dzau

There is much interest over resident c-Kit(+) cells in tissue regeneration. Their role in cardiac regeneration has been controversial. In this study we aim to understand the in vivo behavior of cardiac c-Kit(+) cells at baseline and after myocardial infarction and in response to Sfrp2. This approach can accurately study the in vivo transcript expressions of these cells in temporal response to injury and overcomes the limitations of the in vitro approach. RNA-seq was performed with c-Kit(+) cells and cardiomyocytes from healthy non-injured mice as well as c-Kit(+) cells from 1 day post-MI and 12 days post-MI mice. When compared to in vivo c-Kit(+) cells isolated from a healthy non-injured mouse heart, cardiomyocytes were enriched in transcripts that express anion channels, cation channels, developmental/differentiation pathway components, as well as proteins that inhibit canonical Wnt/β-catenin signaling. Myocardial infarction (MI) induced in vivo c-Kit(+) cells to transiently adopt the cardiomyocyte-specific signature: expression of a number of cardiomyocyte-specific transcripts was maximal 1 day post-MI and declined by 12 days post-MI. We next studied the effect of β-catenin inhibition on in vivo c-Kit(+) cells by administering the Wnt inhibitor Sfrp2 into the infarct border zone. Sfrp2 both enhanced and sustained cardiomyocyte-specific gene expression in the in vivo c-Kit(+) cells: expression of cardiomyocyte-specific transcripts was higher and there was no decline in expression by 12 days post-MI. Further analysis of the biology of c-Kit(+) cells identified that culture induced a significant and irreversible change in their molecular signature raising questions about reliability of in vitro studies. Our findings provide evidence that MI induces in vivo c-Kit(+) cells to adopt transiently a cardiomyocyte-specific pattern of gene expression, and Sfrp2 further enhances and induces sustained gene expression. Our approach is important for understanding c-Kit(+) cells in cardiac regeneration and also has broad implications in the investigation of in vivo resident stem cells in other areas of tissue regeneration.


Methods of Molecular Biology | 2016

Transcriptomic Analysis of Adult Renal Derived Mesenchymal Stem-Like Cells.

Jose A. Gomez; Jeffrey Schmeckpeper; Maria Mirotsou

Mesenchymal stem cells (MSC) from bone marrow or adult tissues are widely studied to evaluate their potential for tissue repair. Differences in tissue of origin, donor variation, or in vitro handling exist and it is still unclear how they affect cell function and regenerative potential. Large-scale gene expression analysis of these cells not only allows researchers to compare and contrast the differences between each MSC subset but also allows for the development of better analytical tools for their characterization and utilization. Here, we describe a protocol for transcriptomics analysis of MSC-like cells derived from adult kidneys.


Journal of Molecular and Cellular Cardiology | 2011

Paracrine mechanisms of stem cell reparative and regenerative actions in the heart

Maria Mirotsou; Tilanthi M. Jayawardena; Jeffrey Schmeckpeper; Massimiliano Gnecchi; Victor J. Dzau


Circulation | 2012

Abstract 18934: Inhibition of Wnt6 by Sfrp2 Regulates CPC Differentiation by Differential Modulation of Canonical and Non Canonical Wnt Pathways

Amanda Verma; Jeffrey Schmeckpeper; Lucy Yin; Lunan Zhang; Farideh Beigi; Alan Payne; Zhiping Zhang; R. H. Pratt; Victor J. Dzau; Maria Mirotsou


Circulation | 2011

Abstract 14016: Secreted Frizzled Related Protein 2 (Sfrp2) Regulates Resident Cardiac Progenitor Cell Proliferation and Differentiation by Modulating Wnt6 Canonical Signaling

Jeffrey Schmeckpeper; Farideh Beigi; Alan Payne; Zhiping Zhang; R. H. Pratt; Victor J. Dzau; Maria Mirotsou


Circulation | 2011

Abstract 13365: ABI Gene Family, Member 3 (NESH) Binding Protein (Abi3bp) Regulates MSC Proliferation in Autocrine/Paracrine Fashion

Conrad P. Hodgkinson; Vinogran Naidoo; Karl Patti; Jeffrey Schmeckpeper; Jose A. Gomez; Zhiping Zhang; Bryce H. Davis; R. H. Pratt; Maria Mirotsou; Victor J. Dzau

Collaboration


Dive into the Jeffrey Schmeckpeper's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Victor J. Dzau

New York Academy of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. H. Pratt

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge