Network


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

Hotspot


Dive into the research topics where Christopher T. Cottage is active.

Publication


Featured researches published by Christopher T. Cottage.


Nature Medicine | 2007

Pim-1 regulates cardiomyocyte survival downstream of Akt

John Muraski; Marcello Rota; Yu Misao; Jenna Fransioli; Christopher T. Cottage; Natalie Gude; Grazia Esposito; Francesca Delucchi; Michael L. Arcarese; Roberto Alvarez; Sailay Siddiqi; Gregory Emmanuel; Weitao Wu; Kimberlee Fischer; Joshua J. Martindale; Christopher C. Glembotski; Annarosa Leri; Jan Kajstura; Nancy S. Magnuson; Anton Berns; Remus M Beretta; Steven R. Houser; Erik Schaefer; Piero Anversa; Mark A. Sussman

The serine-threonine kinases Pim-1 and Akt regulate cellular proliferation and survival. Although Akt is known to be a crucial signaling protein in the myocardium, the role of Pim-1 has been overlooked. Pim-1 expression in the myocardium of mice decreased during postnatal development, re-emerged after acute pathological injury in mice and was increased in failing hearts of both mice and humans. Cardioprotective stimuli associated with Akt activation induced Pim-1 expression, but compensatory increases in Akt abundance and phosphorylation after pathological injury by infarction or pressure overload did not protect the myocardium in Pim-1–deficient mice. Transgenic expression of Pim-1 in the myocardium protected mice from infarction injury, and Pim-1 expression inhibited cardiomyocyte apoptosis with concomitant increases in Bcl-2 and Bcl-XL protein levels, as well as in Bad phosphorylation levels. Relative to nontransgenic controls, calcium dynamics were significantly enhanced in Pim-1–overexpressing transgenic hearts, associated with increased expression of SERCA2a, and were depressed in Pim-1–deficient hearts. Collectively, these data suggest that Pim-1 is a crucial facet of cardioprotection downstream of Akt.


Circulation | 2009

Enhancement of Myocardial Regeneration Through Genetic Engineering of Cardiac Progenitor Cells Expressing Pim-1 Kinase

Kimberlee Fischer; Christopher T. Cottage; Weitao Wu; Shabana Din; Natalie Gude; Daniele Avitabile; Pearl Quijada; Brett Collins; Jenna Fransioli; Mark A. Sussman

Background— Despite numerous studies demonstrating the efficacy of cellular adoptive transfer for therapeutic myocardial regeneration, problems remain for donated cells with regard to survival, persistence, engraftment, and long-term benefits. This study redresses these concerns by enhancing the regenerative potential of adoptively transferred cardiac progenitor cells (CPCs) via genetic engineering to overexpress Pim-1, a cardioprotective kinase that enhances cell survival and proliferation. Methods and Results— Intramyocardial injections of CPCs overexpressing Pim-1 were given to infarcted female mice. Animals were monitored over 4, 12, and 32 weeks to assess cardiac function and engraftment of Pim-1 CPCs with echocardiography, in vivo hemodynamics, and confocal imagery. CPCs overexpressing Pim-1 showed increased proliferation and expression of markers consistent with cardiogenic lineage commitment after dexamethasone exposure in vitro. Animals that received CPCs overexpressing Pim-1 also produced greater levels of cellular engraftment, persistence, and functional improvement relative to control CPCs up to 32 weeks after delivery. Salutary effects include reduction of infarct size, greater number of c-kit+ cells, and increased vasculature in the damaged region. Conclusions— Myocardial repair is significantly enhanced by genetic engineering of CPCs with Pim-1 kinase. Ex vivo gene delivery to enhance cellular survival, proliferation, and regeneration may overcome current limitations of stem cell–based therapeutic approaches.


Physiological Reviews | 2011

MYOCARDIAL AKT: THE OMNIPRESENT NEXUS

Mark A. Sussman; Mirko Völkers; Kimberlee Fischer; Brandi Bailey; Christopher T. Cottage; Shabana Din; Natalie Gude; Daniele Avitabile; Roberto Alvarez; Balaji Sundararaman; Pearl Quijada; Matt Mason; Mathias Konstandin; Amy Malhowski; Zhaokang Cheng; Mohsin Khan; Michael McGregor

One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.


Circulation Research | 2008

Activation of Notch-Mediated Protective Signaling in the Myocardium

Natalie Gude; Gregory Emmanuel; Weitao Wu; Christopher T. Cottage; Kimberlee Fischer; Pearl Quijada; John Muraski; Roberto Alvarez; Marta Rubio; Eric Schaefer; Mark A. Sussman

The Notch network regulates multiple cellular processes, including cell fate determination, development, differentiation, proliferation, apoptosis, and regeneration. These processes are regulated via Notch-mediated activity that involves hepatocyte growth factor (HGF)/c-Met receptor and phosphatidylinositol 3-kinase/Akt signaling cascades. The impact of HGF on Notch signaling was assessed following myocardial infarction as well as in cultured cardiomyocytes. Notch1 is activated in border zone cardiomyocytes coincident with nuclear c-Met following infarction. Intramyocardial injection of HGF enhances Notch1 and Akt activation in adult mouse myocardium. Corroborating evidence in cultured cardiomyocytes shows treatment with HGF or insulin increases levels of Notch effector Hes1 in immunoblots, whereas overexpression of activated Notch intracellular domain prompts a 3-fold increase in phosphorylated Akt. Infarcted hearts injected with adenoviral vector expressing Notch intracellular domain treatment exhibit improved hemodynamic function in comparison with control mice after 4 weeks, implicating Notch signaling in a cardioprotective role following cardiac injury. These results indicate Notch activation in cardiomyocytes is mediated through c-Met and Akt survival signaling pathways, and Notch1 signaling in turn enhances Akt activity. This mutually supportive crosstalk suggests a positive survival feedback mechanism between Notch and Akt signaling in adult myocardium following injury.


Journal of the American College of Cardiology | 2012

Human Cardiac Progenitor Cells Engineered With Pim-I Kinase Enhance Myocardial Repair

Sadia Mohsin; Mohsin Khan; Haruhiro Toko; Brandi Bailey; Christopher T. Cottage; Kathleen Wallach; Divya Nag; Andrew Lee; Sailay Siddiqi; Feng Lan; Kimberlee Fischer; Natalie Gude; Pearl Quijada; Daniele Avitabile; Silvia Truffa; Brett Collins; Walter P. Dembitsky; Joseph C. Wu; Mark A. Sussman

OBJECTIVES The goal of this study was to demonstrate the enhancement of human cardiac progenitor cell (hCPC) reparative and regenerative potential by genetic modification for the treatment of myocardial infarction. BACKGROUND Regenerative potential of stem cells to repair acute infarction is limited. Improved hCPC survival, proliferation, and differentiation into functional myocardium will increase efficacy and advance translational implementation of cardiac regeneration. METHODS hCPCs isolated from the myocardium of heart failure patients undergoing left ventricular assist device implantation were engineered to express green fluorescent protein (hCPCe) or Pim-1-GFP (hCPCeP). Functional tests of hCPC regenerative potential were performed with immunocompromised mice by using intramyocardial adoptive transfer injection after infarction. Myocardial structure and function were monitored by echocardiographic and hemodynamic assessment for 20 weeks after delivery. hCPCe and hCPCeP expressing luciferase were observed by using bioluminescence imaging to noninvasively track persistence. RESULTS hCPCeP exhibited augmentation of reparative potential relative to hCPCe control cells, as shown by significantly increased proliferation coupled with amelioration of infarction injury and increased hemodynamic performance at 20 weeks post-transplantation. Concurrent with enhanced cardiac structure and function, hCPCeP demonstrated increased cellular engraftment and differentiation with improved vasculature and reduced infarct size. Enhanced persistence of hCPCeP versus hCPCe was revealed by bioluminescence imaging at up to 8 weeks post-delivery. CONCLUSIONS Genetic engineering of hCPCs with Pim-1 enhanced repair of damaged myocardium. Ex vivo gene delivery to modify stem cells has emerged as a viable option addressing current limitations in the field. This study demonstrates that efficacy of hCPCs from the failing myocardium can be safely and significantly enhanced through expression of Pim-1 kinase, setting the stage for use of engineered cells in pre-clinical settings.


Stem Cells | 2008

Evolution of the c-kit-Positive Cell Response to Pathological Challenge in the Myocardium

Jenna Fransioli; Brandi Bailey; Natalie Gude; Christopher T. Cottage; John Muraski; Gregory Emmanuel; Weitao Wu; Roberto Alvarez; Marta Rubio; Sergio Ottolenghi; Erik Schaefer; Mark A. Sussman

Cumulative evidence indicates that myocardium responds to growth or injury by recruitment of stem and/or progenitor cells that participate in repair and regenerative processes. Unequivocal identification of this population has been hampered by lack of reagents or markers specific to the recruited population, leading to controversies regarding the nature of these cells. Use of a transgenic mouse expressing green fluorescent protein driven by the c‐kit promoter allows for unambiguous identification of this cell population. Green fluorescent protein (GFP) driven by the c‐kit promoter labels a fraction of the c‐kit+ cells recognized by antibody labeling for c‐kit protein. Expression of GFP by the c‐kit promoter and accumulation of GFP‐positive cells in the myocardium is relatively high at birth compared with adult and declines between postnatal weeks 1 and 2, which tracks in parallel with expression of c‐kit protein and c‐kit‐positive cells. Acute cardiomyopathic injury by infarction prompts increased expression of both GFP protein and GFP‐labeled cells in the region of infarction relative to remote myocardium. Similar increases were observed for c‐kit protein and cells with a slightly earlier onset and decline relative to the GFP signal. Cells coexpressing GFP, c‐kit, and cardiogenic markers were apparent at 1–2 weeks postinfarction. Cardiac‐resident c‐kit+ cell cultures derived from the transgenic line express GFP that is diminished in parallel with c‐kit by induction of differentiation. The use of genetically engineered mice validates and extends the concept of c‐kit+ cells participating in the response to myocardial injury.


Circulation Research | 2010

Cardiac Progenitor Cell Cycling Stimulated by Pim-1 Kinase

Christopher T. Cottage; Brandi Bailey; Kimberlee Fischer; Daniele Avitabile; Brett Collins; Savilla Tuck; Pearl Quijada; Natalie Gude; Roberto Alvarez; John Muraski; Mark A. Sussman

Rationale: Cardioprotective effects of Pim-1 kinase have been previously reported but the underlying mechanistic basis may involve a combination of cellular and molecular mechanisms that remain unresolved. The elucidation of the mechanistic basis for Pim-1 mediated cardioprotection provides important insights for designing therapeutic interventional strategies to treat heart disease. Objective: Effects of cardiac-specific Pim-1 kinase expression on the cardiac progenitor cell (CPC) population were examined to determine whether Pim-1 mediates beneficial effects through augmenting CPC activity. Methods and Results: Transgenic mice created with cardiac-specific Pim-1 overexpression (Pim-wt) exhibit enhanced Pim-1 expression in both cardiomyocytes and CPCs, both of which show increased proliferative activity assessed using 5-bromodeoxyuridine (BrdU), Ki-67, and c-Myc relative to nontransgenic controls. However, the total number of CPCs was not increased in the Pim-wt hearts during normal postnatal growth or after infarction challenge. These results suggest that Pim-1 overexpression leads to asymmetric division resulting in maintenance of the CPC population. Localization and quantitation of cell fate determinants Numb and &agr;-adaptin by confocal microscopy were used to assess frequency of asymmetric division in the CPC population. Polarization of Numb in mitotic phospho-histone positive cells demonstrates asymmetric division in 65% of the CPC population in hearts of Pim-wt mice versus 26% in nontransgenic hearts after infarction challenge. Similarly, Pim-wt hearts had fewer cells with uniform &agr;-adaptin staining indicative of symmetrically dividing CPCs, with 36% of the CPCs versus 73% in nontransgenic sections. Conclusions: These findings define a mechanistic basis for enhanced myocardial regeneration in transgenic mice overexpressing Pim-1 kinase.


Circulation Research | 2010

Pim-1 Kinase Protects Mitochondrial Integrity in Cardiomyocytes

Gwynngelle A. Borillo; Matt Mason; Pearl Quijada; Mirko Völkers; Christopher T. Cottage; Michael McGregor; Shabana Din; Kimberlee Fischer; Natalie Gude; Daniele Avitabile; Steven B. Barlow; Roberto Alvarez; Silvia Truffa; Ross Whittaker; Matthew S. Glassy; Åsa B. Gustafsson; Christopher C. Glembotski; Roberta A. Gottlieb; Joan Heller Brown; Mark A. Sussman

Rationale: Cardioprotective signaling mediates antiapoptotic actions through multiple mechanisms including maintenance of mitochondrial integrity. Pim-1 kinase is an essential downstream effector of AKT-mediated cardioprotection but the mechanistic basis for maintenance of mitochondrial integrity by Pim-1 remains unexplored. This study details antiapoptotic actions responsible for enhanced cell survival in cardiomyocytes with elevated Pim-1 activity. Objective: The purpose of this study is to demonstrate that the cardioprotective kinase Pim-1 acts to inhibit cell death by preserving mitochondrial integrity in cardiomyocytes. Methods and Results: A combination of biochemical, molecular, and microscopic analyses demonstrate beneficial effects of Pim-1 on mitochondrial integrity. Pim-1 protein level increases in the mitochondrial fraction with a corresponding decrease in the cytosolic fraction of myocardial lysates from hearts subjected to 30 minutes of ischemia followed by 30 minutes of reperfusion. Cardiac-specific overexpression of Pim-1 results in higher levels of antiapoptotic Bcl-XL and Bcl-2 compared to samples from normal hearts. In response to oxidative stress challenge, Pim-1 preserves the inner mitochondrial membrane potential. Ultrastructure of the mitochondria is maintained by Pim-1 activity, which prevents swelling induced by calcium overload. Finally, mitochondria isolated from hearts created with cardiac-specific overexpression of Pim-1 show inhibition of cytochrome c release triggered by a truncated form of proapoptotic Bid. Conclusion: Cardioprotective action of Pim-1 kinase includes preservation of mitochondrial integrity during cardiomyopathic challenge conditions, thereby raising the potential for Pim-1 kinase activation as a therapeutic interventional approach to inhibit cell death by antagonizing proapoptotic Bcl-2 family members that regulate the intrinsic apoptotic pathway.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Pim-1 preserves mitochondrial morphology by inhibiting dynamin-related protein 1 translocation.

Shabana Din; Matt Mason; Mirko Völkers; Bevan Johnson; Christopher T. Cottage; Zeping Wang; Anya Joyo; Pearl Quijada; Peter Erhardt; Nancy S. Magnuson; Mathias Konstandin; Mark A. Sussman

Mitochondrial morphological dynamics affect the outcome of ischemic heart damage and pathogenesis. Recently, mitochondrial fission protein dynamin-related protein 1 (Drp1) has been identified as a mediator of mitochondrial morphological changes and cell death during cardiac ischemic injury. In this study, we report a unique relationship between Pim-1 activity and Drp1 regulation of mitochondrial morphology in cardiomyocytes challenged by ischemic stress. Transgenic hearts overexpressing cardiac Pim-1 display reduction of total Drp1 protein levels, increased phosphorylation of Drp1-S637, and inhibition of Drp1 localization to the mitochondria. Consistent with these findings, adenoviral-induced Pim-1 neonatal rat cardiomyocytes (NRCMs) retain a reticular mitochondrial phenotype after simulated ischemia (sI) and decreased Drp1 mitochondrial sequestration. Interestingly, adenovirus Pim-dominant negative NRCMs show increased expression of Bcl-2 homology 3 (BH3)-only protein p53 up-regulated modulator of apoptosis (PUMA), which has been previously shown to induce Drp1 accumulation at mitochondria and increase sensitivity to apoptotic stimuli. Overexpression of the p53 up-regulated modulator of apoptosis–dominant negative adenovirus attenuates localization of Drp1 to mitochondria in adenovirus Pim-dominant negative NRCMs promotes reticular mitochondrial morphology and inhibits cell death during sI. Therefore, Pim-1 activity prevents Drp1 compartmentalization to the mitochondria and preserves reticular mitochondrial morphology in response to sI.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Pim-1 kinase antagonizes aspects of myocardial hypertrophy and compensation to pathological pressure overload

John Muraski; Kimberlee Fischer; Weitao Wu; Christopher T. Cottage; Pearl Quijada; Matt Mason; Shabana Din; Natalie Gude; Roberto Alvarez; Marcello Rota; Jan Kajstura; Zeping Wang; Erik Schaefer; Xiongen Chen; Scott MacDonnel; Nancy S. Magnuson; Stephen R. Houser; Piero Anversa; Mark A. Sussman

Pim-1 kinase exerts potent cardioprotective effects in the myocardium downstream of AKT, but the participation of Pim-1 in cardiac hypertrophy requires investigation. Cardiac-specific expression of Pim-1 (Pim-WT) or the dominant-negative mutant of Pim-1 (Pim-DN) in transgenic mice together with adenoviral-mediated overexpression of these Pim-1 constructs was used to delineate the role of Pim-1 in hypertrophy. Transgenic overexpression of Pim-1 protects mice from pressure-overload-induced hypertrophy relative to wild-type controls as evidenced by improved hemodynamic function, decreased apoptosis, increases in antihypertrophic proteins, smaller myocyte size, and inhibition of hypertrophic signaling after challenge. Similarly, Pim-1 overexpression in neonatal rat cardiomyocyte cultures inhibits hypertrophy induced by endothelin-1. On the cellular level, hearts of Pim-WT mice show enhanced incorporation of BrdU into myocytes and a hypercellular phenotype compared to wild-type controls after hypertrophic challenge. In comparison, transgenic overexpression of Pim-DN leads to dilated cardiomyopathy characterized by increased apoptosis, fibrosis, and severely depressed cardiac function. Furthermore, overexpression of Pim-DN leads to reduced contractility as evidenced by reduced Ca2+ transient amplitude and decreased percentage of cell shortening in isolated myocytes. These data support a pivotal role for Pim-1 in modulation of hypertrophy by impacting responses on molecular, cellular, and organ levels.

Collaboration


Dive into the Christopher T. Cottage's collaboration.

Top Co-Authors

Avatar

Mark A. Sussman

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Natalie Gude

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Kimberlee Fischer

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Roberto Alvarez

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Pearl Quijada

San Diego State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Muraski

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Brandi Bailey

San Diego State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shabana Din

San Diego State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge