Ben Cosgrove: Difference between revisions

From OpenWetWare
Jump to navigationJump to search
No edit summary
No edit summary
Line 25: Line 25:
"Quantitative Analysis of Cytokine-Induced Hepatocyte Proliferation, Apoptosis, and Toxicity"
"Quantitative Analysis of Cytokine-Induced Hepatocyte Proliferation, Apoptosis, and Toxicity"


Many therapeutic agents, including viral gene therapy vectors and small molecule pharmaceutical compounds, are confounded by liver toxicity due to, in part, relationships with inflammatory stimuli in eliciting hepatocyte toxicity and/or death. Our work focuses on developing physiologically relevant in vitro approaches to quantitatively assess how hepatocytes regulate, through the activities of intracellular and extracellular signaling networks, cell fate decisions related to proliferation, survival, apoptosis, and differentiated function following cytokine stimulation in the presence of viral gene therapy agents or small molecule drugs.   
Many therapeutic agents, including viral gene therapy vectors and small molecule pharmaceutical compounds, are confounded by liver toxicity due to, in part, relationships with inflammatory stimuli in eliciting hepatocyte toxicity and/or death. Our work focuses on developing physiologically relevant in vitro approaches to quantitatively assess how hepatocytes regulate, through the activities of intracellular and extracellular signaling networks, cell fate decisions related to proliferation, survival, apoptosis, and differentiated function following cytokine stimulation in the presence of viral gene therapy agents or small molecule drugs.   


Initially, we examined the role of a specific inflammatory cytokine, tumor necrosis factor alpha (TNF), which regulates both hepatocyte proliferation and apoptosis in vivo.  We have shown that TNF stimulates hepatocyte proliferation in vitro through a contingent, inducible autocrine cascade containing the growth factor TGF-alpha and the cytokines IL-1alpha/beta. This TGF-alpha-IL-1 autocrine cascade regulates TNF-induced hepatocyte proliferation in a self-limiting manner as TGF-alpha positively regulates proliferation while also upregulating autocrine IL-1 release, which negatively regulates proliferation.  Similarly, we have shown that TNF potently stimulates apoptosis in hepatocytes infected with a replication-deficient adenovirus in vitro as mediated by the same TGF-alpha-IL-1 autocrine cascade. 
Initially, we examined the role of a specific inflammatory cytokine, tumor necrosis factor alpha (TNF), which regulates both hepatocyte proliferation and apoptosis in vivo.  We have shown that TNF stimulates hepatocyte proliferation and (adenoviral vector-sensitized) apoptosis in vitro through an inducible and time-varying autocrine cascade containing the growth factor TGF-α and the cytokines IL-/β and IL-1ra. This TGF-α–IL-1α/β–IL-1ra autocrine cascade regulates TNF-induced hepatocyte proliferation and apoptosis responses in a self-antagonizing manner by contributing to multiple signaling pathways, including Akt, ERK, JNK, p38, and IKK–NF-κB, downstream of TNFR.   
 
Currently, we are developing in vitro models of idiosyncratic drug hepatotoxicity by examining the interactions between multiple pharmaceutical compounds and inflammatory cytokines.  In this work, we aim to elucidate how certain idiosyncratic hepatotoxic drugs exhibit synergistic toxicity relationships with inflammatory cytokines by collecting systems-level intracellular signaling data (using multiplexed kinase activity assays and bead-based phosphoprotein detection schemes) and phenotypic cell response data (using imaging-based assays of differentiated function and high-throughput cell necrosis and apoptosis assays)This data set will be used to develop statistical signaling-outcome models through partial least squares regression (PLSR) approaches to identify and predict key signaling activities that regulate a diverse set of hepatocyte toxicity phenotypes and to inform future therapeutic strategies.


Currently, we are developing in vitro models of idiosyncratic drug hepatotoxicity by examining the interactions between multiple pharmaceutical compounds and inflammatory cytokines. In this work, we aim to elucidate how certain idiosyncratic hepatotoxic drugs exhibit synergistic toxicity relationships with inflammatory cytokines by collecting systems-level intracellular signaling data and phenotypic cellular toxicity data. This data set has been used to develop data-driven signaling-outcome models through partial least squares regression (PLSR) approaches to identify and predict key signaling activities that regulate a diverse set of hepatocyte toxicity phenotypes and to inform future therapeutic strategies.


''Member of:''
''Member of:''

Revision as of 13:13, 7 March 2008

Home        People        Research        Publications        Links        Internal       

Ben Cosgrove CV email Epernicus

Ph.D. Student

Biological Engineering Division webpage

Massachusetts Institute of Technology webpage


Research Advisors:


Research Summary:

"Quantitative Analysis of Cytokine-Induced Hepatocyte Proliferation, Apoptosis, and Toxicity"

Many therapeutic agents, including viral gene therapy vectors and small molecule pharmaceutical compounds, are confounded by liver toxicity due to, in part, relationships with inflammatory stimuli in eliciting hepatocyte toxicity and/or death. Our work focuses on developing physiologically relevant in vitro approaches to quantitatively assess how hepatocytes regulate, through the activities of intracellular and extracellular signaling networks, cell fate decisions related to proliferation, survival, apoptosis, and differentiated function following cytokine stimulation in the presence of viral gene therapy agents or small molecule drugs.

Initially, we examined the role of a specific inflammatory cytokine, tumor necrosis factor alpha (TNF), which regulates both hepatocyte proliferation and apoptosis in vivo. We have shown that TNF stimulates hepatocyte proliferation and (adenoviral vector-sensitized) apoptosis in vitro through an inducible and time-varying autocrine cascade containing the growth factor TGF-α and the cytokines IL-1α/β and IL-1ra. This TGF-α–IL-1α/β–IL-1ra autocrine cascade regulates TNF-induced hepatocyte proliferation and apoptosis responses in a self-antagonizing manner by contributing to multiple signaling pathways, including Akt, ERK, JNK, p38, and IKK–NF-κB, downstream of TNFR.

Currently, we are developing in vitro models of idiosyncratic drug hepatotoxicity by examining the interactions between multiple pharmaceutical compounds and inflammatory cytokines. In this work, we aim to elucidate how certain idiosyncratic hepatotoxic drugs exhibit synergistic toxicity relationships with inflammatory cytokines by collecting systems-level intracellular signaling data and phenotypic cellular toxicity data. This data set has been used to develop data-driven signaling-outcome models through partial least squares regression (PLSR) approaches to identify and predict key signaling activities that regulate a diverse set of hepatocyte toxicity phenotypes and to inform future therapeutic strategies.

Member of:


Funding: