Network


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

Hotspot


Dive into the research topics where Andrea S. Bedrosian is active.

Publication


Featured researches published by Andrea S. Bedrosian.


Journal of Clinical Investigation | 2009

In liver fibrosis, dendritic cells govern hepatic inflammation in mice via TNF-α

Michael K. Connolly; Andrea S. Bedrosian; Jon Mallen-St. Clair; Aaron Mitchell; Junaid Ibrahim; Andrea Stroud; H. Leon Pachter; Dafna Bar-Sagi; Alan B. Frey; George Miller

Hepatic fibrosis occurs during most chronic liver diseases and is driven by inflammatory responses to injured tissue. Because DCs are central to modulating liver immunity, we postulated that altered DC function contributes to immunologic changes in hepatic fibrosis and affects the pathologic inflammatory milieu within the fibrotic liver. Using mouse models, we determined the contribution of DCs to altered hepatic immunity in fibrosis and investigated the role of DCs in modulating the inflammatory environment within the fibrotic liver. We found that DC depletion completely abrogated the elevated levels of many inflammatory mediators that are produced in the fibrotic liver. DCs represented approximately 25% of the fibrotic hepatic leukocytes and showed an elevated CD11b+CD8- fraction, a lower B220+ plasmacytoid fraction, and increased expression of MHC II and CD40. Moreover, after liver injury, DCs gained a marked capacity to induce hepatic stellate cells, NK cells, and T cells to mediate inflammation, proliferation, and production of potent immune responses. The proinflammatory and immunogenic effects of fibrotic DCs were contingent on their production of TNF-alpha. Therefore, modulating DC function may be an attractive approach to experimental therapeutics in fibro-inflammatory liver disease.


Journal of Experimental Medicine | 2012

MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells

Atsuo Ochi; Andrew H. Nguyen; Andrea S. Bedrosian; Harry Mushlin; Saman Zarbakhsh; Rocky Barilla; Constantinos P. Zambirinis; Nina Fallon; Adeel Rehman; Yuliya Pylayeva-Gupta; Sana Badar; Cristina H. Hajdu; Alan B. Frey; Dafna Bar-Sagi; George Miller

MyD88 blockade exaggerates the ability of dendritic cells to promote the transition from chronic pancreatitis to pancreatic cancer.


Journal of Leukocyte Biology | 2010

Distinct populations of metastases‐enabling myeloid cells expand in the liver of mice harboring invasive and preinvasive intra‐abdominal tumor

Michael K. Connolly; Jon Mallen-St. Clair; Andrea S. Bedrosian; Ashim Malhotra; Valery Vera; Junaid Ibrahim; Justin R. Henning; H. Leon Pachter; Dafna Bar-Sagi; Alan B. Frey; George Miller

The liver is the most common site of adenocarcinoma metastases, even in patients who initially present with early disease. We postulated that immune‐suppressive cells in the liver of tumor‐bearing hosts inhibit anti‐tumor T cells, thereby accelerating the growth of liver metastases. Using models of early preinvasive pancreatic neoplasia and advanced colorectal cancer, aims of this study were to determine immune phenotype, stimulus for recruitment, inhibitory effects, and tumor‐enabling function of immune‐suppressive cells in the liver of tumor‐bearing hosts. We found that in mice with intra‐abdominal malignancies, two distinct CD11b+Gr1+ populations with divergent phenotypic and functional properties accumulate in the liver, becoming the dominant hepatic leukocytes. Their expansion is contingent on tumor expression of KC. These cells are distinct from CD11b+Gr1+ populations in other tissues of tumor‐bearing hosts in terms of cellular phenotype and cytokine and chemokine profile. Liver CD11b+Gr1+ cells are highly suppressive of T cell activation, proliferation, and cytotoxicity and induce the development of Tregs. Moreover, liver myeloid‐derived suppressor cells accelerate the development of hepatic metastases by inactivation of cytotoxic T cells. These findings may explain the propensity of patients with intra‐abdominal cancers to develop liver metastases and suggest a promising target for experimental therapeutics.


Hepatology | 2011

Dendritic cell depletion exacerbates acetaminophen hepatotoxicity

Michael K. Connolly; Diego Ayo; Ashim Malhotra; Michael Hackman; Andrea S. Bedrosian; Junaid Ibrahim; Napoleon E. Cieza-Rubio; Andrew H. Nguyen; Justin R. Henning; Monica Dorvil-Castro; H. Leon Pachter; George Miller

Acetaminophen (APAP) overdose is one of the most frequent causes of acute liver failure in the United States and is primarily mediated by toxic metabolites that accumulate in the liver upon depletion of glutathione stores. However, cells of the innate immune system, including natural killer (NK) cells, neutrophils, and Kupffer cells, have also been implicated in the centrilobular liver necrosis associated with APAP. We have recently shown that dendritic cells (DCs) regulate intrahepatic inflammation in chronic liver disease and, therefore, postulated that DC may also modulate the hepatotoxic effects of APAP. We found that DC immune‐phenotype was markedly altered after APAP challenge. In particular, liver DC expressed higher MHC II, costimulatory molecules, and Toll‐like receptors, and produced higher interleukin (IL)‐6, macrophage chemoattractant protein‐1 (MCP‐1), and tumor necrosis factor alpha (TNF‐α). Conversely, spleen DC were unaltered. However, APAP‐induced centrilobular necrosis, and its associated mortality, was markedly exacerbated upon DC depletion. Conversely, endogenous DC expansion using FMS‐like tyrosine kinase 3 ligand (Flt3L) protected mice from APAP injury. Our mechanistic studies showed that APAP liver DC had the particular capacity to prevent NK cell activation and induced neutrophil apoptosis. Nevertheless, the exacerbated hepatic injury in DC‐depleted mice challenged with APAP was independent of NK cells and neutrophils or numerous immune modulatory cytokines and chemokines. Conclusion: Taken together, these data indicate that liver DC protect against APAP toxicity, whereas their depletion is associated with exacerbated hepatotoxicity. (HEPATOLOGY 2011;)


Journal of Immunology | 2010

In Hepatic Fibrosis, Liver Sinusoidal Endothelial Cells Acquire Enhanced Immunogenicity

Michael K. Connolly; Andrea S. Bedrosian; Ashim Malhotra; Justin R. Henning; Junaid Ibrahim; Valery Vera; Napoleon E. Cieza-Rubio; Burhan U. Hassan; H. Leon Pachter; Steven P. Cohen; Alan B. Frey; George Miller

The normal liver is characterized by immunologic tolerance. Primary mediators of hepatic immune tolerance are liver sinusoidal endothelial cells (LSECs). LSECs block adaptive immunogenic responses to Ag and induce the generation of T regulatory cells. Hepatic fibrosis is characterized by both intense intrahepatic inflammation and altered hepatic immunity. We postulated that, in liver fibrosis, a reversal of LSEC function from tolerogenic to proinflammatory and immunogenic may contribute to both the heightened inflammatory milieu and altered intrahepatic immunity. We found that, after fibrotic liver injury from hepatotoxins, LSECs become highly proinflammatory and secrete an array of cytokines and chemokines. In addition, LSECs gain enhanced capacity to capture Ag and induce T cell proliferation. Similarly, unlike LSECs in normal livers, in fibrosis, LSECs do not veto dendritic cell priming of T cells. Furthermore, whereas in normal livers, LSECs are active in the generation of T regulatory cells, in hepatic fibrosis LSECs induce an immunogenic T cell phenotype capable of enhancing endogenous CTLs and generating potent de novo CTL responses. Moreover, depletion of LSECs from fibrotic liver cultures mitigates the proinflammatory milieu characteristic of hepatic fibrosis. Our findings offer a critical understanding of the role of LSECs in modulating intrahepatic immunity and inflammation in fibro-inflammatory liver disease.


Gastroenterology | 2011

Dendritic Cells Promote Pancreatic Viability in Mice with Acute Pancreatitis

Andrea S. Bedrosian; Andrew H. Nguyen; Michael Hackman; Michael K. Connolly; Ashim Malhotra; Junaid Ibrahim; Napoleon E. Cieza–Rubio; Justin R. Henning; Rocky Barilla; Adeel Rehman; H. Leon Pachter; Marco V. Medina–Zea; Steven M. Cohen; Alan B. Frey; Devrim Acehan; George Miller

BACKGROUND & AIMS The cellular mediators of acute pancreatitis are incompletely understood. Dendritic cells (DCs) can promote or suppress inflammation, depending on their subtype and context. We investigated the roles of DC in development of acute pancreatitis. METHODS Acute pancreatitis was induced in CD11c.DTR mice using caerulein or L-arginine; DCs were depleted by administration of diphtheria toxin. Survival was analyzed using Kaplan-Meier method. RESULTS Numbers of major histocompatibility complex II(+)CD11c(+) DCs increased 100-fold in pancreata of mice with acute pancreatitis to account for nearly 15% of intrapancreatic leukocytes. Intrapancreatic DCs acquired a distinct immune phenotype in mice with acute pancreatitis; they expressed higher levels of major histocompatibility complex II and CD86 and increased production of interleukin-6, membrane cofactor protein-1, and tumor necrosis factor-α. However, rather than inducing an organ-destructive inflammatory process, DCs were required for pancreatic viability; the exocrine pancreas died in mice that were depleted of DCs and challenged with caerulein or L-arginine. All mice with pancreatitis that were depleted of DCs died from acinar cell death within 4 days. Depletion of DCs from mice with pancreatitis resulted in neutrophil infiltration and increased levels of systemic markers of inflammation. However, the organ necrosis associated with depletion of DCs did not require infiltrating neutrophils, activation of nuclear factor-κB, or signaling by mitogen-activated protein kinase or tumor necrosis factor-α. CONCLUSIONS DCs are required for pancreatic viability in mice with acute pancreatitis and might protect organs against cell stress.


Archive | 2018

Masters Program Bariatric Pathway: Adjustable Gastric Band

Andrea S. Bedrosian; Christine J. Ren Fielding

Every year, tens of thousands of patients worldwide undergo laparoscopic adjustable gastric banding (LAGB) for treatment of morbid obesity. With standard surgical technique, and under the auspices of an intensive postoperative follow-up program, LAGB can be a lifesaving companion for those suffering the physical, psychosocial, and economic effects of morbid obesity. Multiple studies have shown the safety and efficacy of this procedure. Long-term successful outcomes are dependent on frequent adjustments and close clinical and behavioral support from a dedicated bariatric surgical practice. This chapter reviews the accepted surgical technique for placement of the adjustable gastric band, as well as outcomes.


Journal of Surgical Research | 2012

The Role of Dendritic Cell and Pancreatic Stellate Cell Cross-Talk in Chronic Pancreatitis

Andrew H. Nguyen; Andrea S. Bedrosian; Michael K. Connolly; Justin R. Henning; V. Medina-Zea; Napoleon E. Cieza-Rubio; M. Dorvil-Castro; Christopher S. Graffeo; Michael Hackman; Adeel Rehman; Junaid Ibrahim; George Miller


Journal of The American College of Surgeons | 2011

Divergent lipid-laden dendritic cell subpopulations regulate hepatic tolerance and immunity

Junaid Ibrahim; Andrew H. Nguyen; Aaron Mitchell; Andrea S. Bedrosian; Justin R. Henning; Ahsan U. Khan; Adeel Rehman; Christopher S. Graffeo; Pamela Tan; George Miller


Journal of Surgical Research | 2011

Dendritic Cells Are Necessary For Pancreas Viability In Acute Pancreatitis

Andrew H. Nguyen; Andrea S. Bedrosian; Justin R. Henning; Michael K. Connolly; Ashim Malhotra; Napoleon E. Cieza-Rubio; Junaid Ibrahim; M. Marr; Rocky Barilla; M. Rapp; Diego Ayo; P. Tan; H. Mushlin; I.C. Berkowitz; Steven M. Cohen; George Miller

Collaboration


Dive into the Andrea S. Bedrosian'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