Network


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

Hotspot


Dive into the research topics where Michael J. Bouchard is active.

Publication


Featured researches published by Michael J. Bouchard.


Journal of Virology | 2004

The Enigmatic X Gene of Hepatitis B Virus

Michael J. Bouchard; Robert J. Schneider

The exact function of HBx during HBV replication and its influence in HBV-associated hepatocellular carcinoma are still undefined, but common themes are emerging. Most studies agree that, if not absolutely essential for HBV infections, HBx at the very least plays a critical role in viral replication. HBx functions in the cytoplasm to activate various signaling pathways, many of which are controlled by modulation of cytosolic calcium. A number of groups have now demonstrated that HBx activities somehow involve modulation of cytosolic calcium, although the exact molecular mechanism is undefined. In the nucleus, HBx can regulate transcription through a direct interaction with different transcription factors and, in some cases, enhance their binding to specific transcription elements. Clearly, HBx can influence apoptotic and cell cycle regulatory pathways, but the exact consequences of these activities of HBx are likely affected by the cell types in which the studies have been conducted. In this regard, it is interesting to note that much of the seeming confusion or controversy regarding HBx activities is likely the result of the different assays performed, and the findings of different studies may ultimately be more compatible than expected. In most experiments, HBx activities have been analyzed based on the end product of what are long and complex pathways. For example, transcriptional reporter readout, apoptotic assays, cell cycle regulation, and mutagenic studies are judged based on an analysis of the final product of a given pathway without considering common effectors. HBx may initiate overlapping and very similar effectors in all the studies conducted, but the consequence of this will likely vary depending on the specific cellular phenotype and the assay involved. Moreover, the frequency or longevity of this stimulus may vary depending on the amount of HBx present, which in turn influences the results. Fluctuations in the frequency or longevity of a HBx-induced stimulus, such as calcium signaling, can have a dramatic influence on the final effect on a cell (reviewed in reference 19). Equally interesting is the possibility that HBx has different consequences for hepatocyte physiology as HBV-infected cells are targeted by the immune system or as hepatocytes in which HBx is expressed undergo transformation and progression to HCC. Both these processes are dynamic, and it is likely that cellular physiology is consequently altered. The demonstration that different signaling pathways can be activated based on the differentiation state of hepatocytes derived from the same parental cell line and suggestions that the cellular status of p53, p21, and p27 can affect HBx-dependent regulation of cell cycle control supports the notion that the consequence of HBx expression may change as hepatocytes become transformed (2, 62, 93). It remains to be determined whether similar results will be observed in the context of an authentic HBV infection. Such observations will add to the complexity of identifying and understanding the precise molecular mechanism of HBx activities during viral replication. However, there is the exciting possibility that the end product of HBx activities may change as the cell responds to the infection. Finally, studies of HBx activities during HBV replication should help clarify the many properties that have been ascribed to HBx expression, including those that are required for viral replication and those that may influence cellular transformation.


Journal of Virology | 2003

Activation and Inhibition of Cellular Calcium and Tyrosine Kinase Signaling Pathways Identify Targets of the HBx Protein Involved in Hepatitis B Virus Replication

Michael J. Bouchard; Robyn Puro; Lihua Wang; Robert J. Schneider

ABSTRACT Human hepatitis B virus (HBV) HBx protein is a multifunctional protein that activates cellular signaling pathways and is thought to be essential for viral infection. Woodchuck HBV mutants that lack HBx are unable to replicate in vivo or are severely impaired. HBV replication in HepG2 cells, a human hepatoblastoma cell line, is stimulated 5- to 10-fold by HBx protein. We have utilized the HepG2, HBx-dependent HBV replication system to study the effects of activators and inhibitors of cytosolic calcium and tyrosine kinase signaling pathways on viral replication. By transfecting either a wild-type HBV genome or an HBV genome that does not express HBx and then treating transfected cells with activators or inhibitors of signaling pathways, we identified compounds that either impair wild-type HBV replication or rescue HBx-deficient HBV replication. Geldanamycin or herbimycin A, tyrosine kinase inhibitors, blocked HBV replication. Derivatives of cyclosporine, i.e., cyclosporine A, cyclosporine H, and SDZ NIM811, which block cytosolic calcium signaling and specifically the mitochondrial permeability transition pore (SDZ NIM811), also impaired HBV replication. Treatment of cells with compounds that increase cytosolic calcium levels by a variety of mechanisms rescued replication of an HBx-deficient HBV mutant. Transcription of viral RNA and production of viral capsids were only minimally affected by these treatments. These results define a functional signaling circuit for HBV replication that includes calcium signaling and activation of cytosolic signaling pathways involving Src kinases, and they suggest that these pathways are stimulated by HBx acting on the mitochondrial transition pore.


Cancer Letters | 2011

Hepatitis B and C virus hepatocarcinogenesis: Lessons learned and future challenges

Michael J. Bouchard; Sonia Navas-Martin

Worldwide, hepatocellular carcinoma (HCC) is one of the most common cancers. It is thought that 80% of hepatocellular carcinomas are linked to chronic infections with the hepatitis B (HBV) or hepatitis C (HCV) viruses. Chronic HBV and HCV infections can alter hepatocyte physiology in similar ways and may utilize similar mechanisms to influence the development of HCC. There has been significant progress towards understanding the molecular biology of HBV and HCV and identifying the cellular signal transduction pathways that are altered by HBV and HCV infections. Although the precise molecular mechanisms that link HBV and HCV infections to the development of HCC are not entirely understood, there is considerable evidence that both inflammatory responses to infections with these viruses, and associated destruction and regeneration of hepatocytes, as well as activities of HBV- or HCV-encoded proteins, contribute to hepatocyte transformation. In this review, we summarize progress in defining mechanisms that may link HBV and HCV infections to the development of HCC, discuss the challenges of directly defining the processes that underlie HBV- and HCV-associated HCC, and describe areas that remain to be explored.


Journal of Virology | 2008

Hepatitis B Virus HBx Protein Localizes to Mitochondria in Primary Rat Hepatocytes and Modulates Mitochondrial Membrane Potential

Amy J. Clippinger; Michael J. Bouchard

ABSTRACT Over 350 million people are chronically infected with hepatitis B virus (HBV), and a significant number of chronically infected individuals develop primary liver cancer. HBV encodes seven viral proteins, including the nonstructural X (HBx) protein. The results of studies with immortalized or transformed cells and with HBx-transgenic mice demonstrated that HBx can interact with mitochondria. However, no studies with normal hepatocytes have characterized the precise mitochondrial localization of HBx or the effect of HBx on mitochondrial physiology. We have used cultured primary rat hepatocytes as a model system to characterize the mitochondrial localization of HBx and the effect of HBx expression on mitochondrial physiology. We now show that a fraction of HBx colocalizes with density-gradient-purified mitochondria and associates with the outer mitochondrial membrane. We also demonstrate that HBx regulates mitochondrial membrane potential in hepatocytes and that this function of HBx varies depending on the status of NF-κB activity. In primary rat hepatocytes, HBx activation of NF-κB prevented mitochondrial membrane depolarization; however, when NF-κB activity was inhibited, HBx induced membrane depolarization through modulation of the mitochondrial permeability transition pore. Collectively, these results define potential pathways through which HBx may act in order to modulate mitochondrial physiology, thereby altering many cellular activities and ultimately contributing to the development of HBV-associated liver cancer.


Journal of Virology | 2001

Hepatitis B virus HBx protein activation of cyclin A-cyclin-dependent kinase 2 complexes and G1 transit via a Src kinase pathway.

Michael J. Bouchard; Stavros Giannakopoulos; Edith H. Wang; Naoko Tanese; Robert J. Schneider

ABSTRACT Numerous studies have demonstrated that the hepatitis B virus HBx protein stimulates signal transduction pathways and may bind to certain transcription factors, particularly the cyclic AMP response element binding protein, CREB. HBx has also been shown to promote early cell cycle progression, possibly by functionally replacing the TATA-binding protein-associated factor 250 (TAFII250), a transcriptional coactivator, and/or by stimulating cytoplasmic signal transduction pathways. To understand the basis for early cell cycle progression mediated by HBx, we characterized the molecular mechanism by which HBx promotes deregulation of the G0 and G1 cell cycle checkpoints in growth-arrested cells. We demonstrate that TAFII250 is absolutely required for HBx activation of the cyclin A promoter and for promotion of early cell cycle transit from G0 through G1. Thus, HBx does not functionally replace TAFII250 for transcriptional activity or for cell cycle progression, in contrast to a previous report. Instead, HBx is shown to activate the cyclin A promoter, induce cyclin A–cyclin-dependent kinase 2 complexes, and promote cycling of growth-arrested cells into G1 through a pathway involving activation of Src tyrosine kinases. HBx stimulation of Src kinases and cyclin gene expression was found to force growth-arrested cells to transit through G1 but to stall at the junction with S phase, which may be important for viral replication.


ACS Nano | 2009

In Situ Intracellular Spectroscopy with Surface Enhanced Raman Spectroscopy (SERS)-Enabled Nanopipettes

Elina A. Vitol; Zulfiya Orynbayeva; Michael J. Bouchard; Jane Azizkhan-Clifford; Gary Friedman; Yury Gogotsi

We report on a new analytical approach to intracellular chemical sensing that utilizes a surface-enhanced Raman spectroscopy (SERS)-enabled nanopipette. The probe is comprised of a glass capillary with a 100-500 nm tip coated with gold nanoparticles. The fixed geometry of the gold nanoparticles allows us to overcome the limitations of the traditional approach for intracellular SERS using metal colloids. We demonstrate that the SERS-enabled nanopipettes can be used for in situ analysis of living cell function in real time. In addition, SERS functionality of these probes allows tracking of their localization in a cell. The developed probes can also be applied for highly sensitive chemical analysis of nanoliter volumes of chemicals in a variety of environmental and analytical applications.


Journal of Virology | 2006

Activation of Focal Adhesion Kinase by Hepatitis B Virus HBx Protein: Multiple Functions in Viral Replication

Michael J. Bouchard; Lihua Wang; Robert J. Schneider

ABSTRACT The hepatitis B virus (HBV) X protein (HBx) is a multifunctional regulator of cellular signal transduction and transcription pathways and has a critical role in HBV replication. Much of the cytoplasmic signal transduction activity associated with HBx expression and its stimulation of viral replication is attributable to HBx-induced activation of calcium signaling pathways involving Pyk2 and Src tyrosine kinases. To further characterize upstream signal transduction pathways that are required for HBx activity, including activation of Src and mitogen-activated protein kinase (MAPK) cascades, we determined whether focal adhesion kinase (FAK), a known regulator of Src family kinases and the other member of the Pyk2/FAK kinase family, is activated by HBx. We report that HBx activates FAK and that FAK activation is important for multiple HBx functions. Dominant inhibiting forms of FAK blocked HBx activation of Src kinases and downstream signal transduction, HBx stimulation of NF-κB and AP-1-dependent transcription, and HBV DNA replication. We also demonstrate that HBx-induced activation of FAK is dependent on cellular calcium signaling, which is modulated by HBx. Moreover, prolonged expression of HBx increases both FAK activity and its level of expression. FAK activation may play a role in cellular transformation and cancer progression. HBx stimulation of FAK activity and abundance may also be relevant as a potential cofactor in HBV-associated hepatocellular carcinoma.


Journal of Virology | 2010

The Hepatitis B Virus X Protein Modulates Hepatocyte Proliferation Pathways To Stimulate Viral Replication

Tricia L. Gearhart; Michael J. Bouchard

ABSTRACT Worldwide, there are over 350 million people who are chronically infected with the human hepatitis B virus (HBV); chronic HBV infections are associated with the development of hepatocellular carcinoma (HCC). The results of various studies suggest that the HBV X protein (HBx) has a role in the development of HBV-associated HCC. HBx can regulate numerous cellular signal transduction pathways, including those that modulate cell proliferation. Many previous studies that analyzed the impact of HBx on cell proliferation pathways were conducted using established or immortalized cell lines, and when HBx was expressed in the absence of HBV replication, and the precise effect of HBx on these pathways has often differed depending on experimental conditions. We have studied the effect of HBx on cell proliferation in cultured primary rat hepatocytes, a biologically relevant system. We demonstrate that HBx, both by itself and in the context of HBV replication, affected the levels and activities of various cell cycle-regulatory proteins to induce normally quiescent hepatocytes to enter the G1 phase of the cell cycle but not to proceed to S phase. We linked HBx regulation of cell proliferation to cytosolic calcium signaling and HBx stimulation of HBV replication. Cumulatively, our studies suggest that HBx induces normally quiescent hepatocytes to enter the G1 phase of the cell cycle and that this calcium-dependent HBx activity is required for HBV replication. These studies identify an essential function of HBx during HBV replication and a mechanism that may connect HBV infections to the development of HCC.


Journal of Virology | 2009

Hepatitis B Virus X Protein Modulates Apoptosis in Primary Rat Hepatocytes by Regulating both NF-κB and the Mitochondrial Permeability Transition Pore

Amy J. Clippinger; Tricia L. Gearhart; Michael J. Bouchard

ABSTRACT The hepatitis B virus (HBV) X protein (HBx) is a multifunctional protein that regulates numerous cellular signal transduction pathways, including those that modulate apoptosis. However, different HBx-dependent effects on apoptosis have been reported; these differences are likely the consequence of the exact conditions and cell types used in a study. Many of the previously reported studies that analyzed HBx regulation of apoptosis were conducted in immortalized or transformed cells, and the alterations that have transformed or immortalized these cells likely impact apoptotic pathways. We examined the effects of HBx on apoptotic pathways in cultured primary rat hepatocytes, a biologically relevant system that mimics normal hepatocytes in the liver. We analyzed the effects of HBx on apoptosis both when HBx was expressed in the absence of other HBV proteins and in the context of HBV replication. HBx stimulation of NF-κB inhibited the activation of apoptotic pathways in cultured primary rat hepatocytes. However, when HBx-induced activation of NF-κB was blocked, HBx stimulated apoptosis; blocking the activity of the mitochondrial permeability transition pore inhibited HBx activation of apoptosis. These results suggest that HBx can be either proapoptotic or antiapoptotic in hepatocytes, depending on the status of NF-κB, and confirm previous studies that link some HBx activities to modulation of the mitochondrial permeability transition pore. Overall, our studies define apoptotic pathways that are regulated by HBx in cultured primary hepatocytes and provide potential mechanisms for the development of HBV-associated liver cancer.


Journal of Virology | 2007

Hepatitis B Virus Replication Is Associated with an HBx-Dependent Mitochondrion-Regulated Increase in Cytosolic Calcium Levels

Stephanie L. McClain; Amy J. Clippinger; Rebecca Lizzano; Michael J. Bouchard

ABSTRACT The nonstructural hepatitis B virus (HBV) protein HBx has an important role in HBV replication and in HBV-associated liver disease. Many activities have been linked to HBx expression; however, the molecular mechanisms underlying many of these activities are unknown. One proposed HBx function is the regulation of cytosolic calcium. We analyzed calcium levels in HepG2 cells that expressed HBx or replicating HBV, and we demonstrated that HBx, expressed in the absence of other HBV proteins or in the context of HBV replication, elevates cytosolic calcium. We linked this elevation of cytosolic calcium to the association of HBx with the mitochondrial permeability transition pore.

Collaboration


Dive into the Michael J. Bouchard's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge