Network


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

Hotspot


Dive into the research topics where Jiansheng Huang is active.

Publication


Featured researches published by Jiansheng Huang.


American Journal of Pathology | 2014

Elucidating the Metabolic Regulation of Liver Regeneration

Jiansheng Huang; David A. Rudnick

The regenerative capability of liver is well known, and the mechanisms that regulate liver regeneration are extensively studied. Such analyses have defined general principles that govern the hepatic regenerative response and implicated specific extracellular and intracellular signals as regulated during and essential for normal liver regeneration. Nevertheless, the most proximal events that stimulate liver regeneration and the distal signals that terminate this process remain incompletely understood. Recent data suggest that the metabolic response to hepatic insufficiency might be the proximal signal that initiates regenerative hepatocellular proliferation. This review provides an overview of the data in support of a metabolic model of liver regeneration and reflects on the clinical implications and areas for further study suggested by these findings.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2014

Fibroblast growth factor 15 deficiency impairs liver regeneration in mice

Bo Kong; Jiansheng Huang; Yan Zhu; Guodong Li; Jessica A. Williams; Steven Shen; Lauren M. Aleksunes; Jason R. Richardson; Udayan Apte; David A. Rudnick; Grace L. Guo

Fibroblast growth factor (FGF) 15 (human homolog, FGF19) is an endocrine FGF highly expressed in the small intestine of mice. Emerging evidence suggests that FGF15 is critical for regulating hepatic functions; however, the role of FGF15 in liver regeneration is unclear. This study assessed whether liver regeneration is altered in FGF15 knockout (KO) mice following 2/3 partial hepatectomy (PHx). The results showed that FGF15 KO mice had marked mortality, with the survival rate influenced by genetic background. Compared with wild-type mice, the KO mice displayed extensive liver necrosis and marked elevation of serum bile acids and bilirubin. Furthermore, hepatocyte proliferation was reduced in the KO mice because of impaired cell cycle progression. After PHx, the KO mice had weaker activation of signaling pathways that are important for liver regeneration, including signal transducer and activator of transcription 3, nuclear factor-κB, and mitogen-activated protein kinase. Examination of the KO mice at early time points after PHx revealed a reduced and/or delayed induction of immediate-early response genes, including growth-control transcription factors that are critical for liver regeneration. In conclusion, the results suggest that FGF15 deficiency severely impairs liver regeneration in mice after PHx. The underlying mechanism is likely the result of disrupted bile acid homeostasis and impaired priming of hepatocyte proliferation.


Hepatology | 2013

Characterization of the regulation and function of zinc‐dependent histone deacetylases during rodent liver regeneration

Jiansheng Huang; Emily Barr; David A. Rudnick

The studies reported here were undertaken to define the regulation and functional importance of zinc‐dependent histone deacetylase (Zn‐HDAC) activity during liver regeneration using the mouse partial hepatectomy (PH) model. The results showed that hepatic HDAC activity was significantly increased in nuclear and cytoplasmic fractions following PH. Further analyses showed isoform‐specific effects of PH on HDAC messenger RNA (mRNA) and protein expression, with increased expression of the class I HDACs, 1 and 8, and class II HDAC4 in regenerating liver. Hepatic expression of (class II) HDAC5 was unchanged after PH; however, HDAC5 exhibited transient nuclear accumulation in regenerating liver. These changes in hepatic HDAC expression, subcellular localization, and activity coincided with diminished histone acetylation in regenerating liver. The significance of these events was investigated by determining the effects of suberoylanilide hydroxyamic acid (SAHA, a specific inhibitor of Zn‐HDAC activity) on hepatic regeneration. The results showed that SAHA treatment suppressed the effects of PH on histone deacetylation and hepatocellular bromodeoxyuridine (BrdU) incorporation. Further examination showed that SAHA blunted hepatic expression and activation of cell cycle signals downstream of induction of cyclin D1 expression in mice subjected to PH. Conclusion: The data reported here demonstrate isoform‐specific regulation of Zn‐HDAC expression, subcellular localization, and activity in regenerating liver. These studies also indicate that HDAC activity promotes liver regeneration by regulating hepatocellular cell cycle progression at a step downstream of cyclin D1 induction. (HEPATOLOGY 2013)


Hepatology | 2012

Analysis of the role of hepatic PPARγ expression during mouse liver regeneration

Vered Gazit; Jiansheng Huang; Alexander Weymann; David A. Rudnick

Mice subjected to partial hepatectomy (PH) develop hypoglycemia, followed by increased systemic lipolysis and hepatic fat accumulation, prior to onset of hepatocellular proliferation. Strategies that disrupt these metabolic events inhibit regeneration. These observations suggest that alterations in metabolism in response to hepatic insufficiency promote liver regeneration. Hepatic expression of the peroxisome proliferator‐activated receptor gamma (PPARγ) influences fat accumulation in the liver. Therefore, the studies reported here were undertaken to assess the effects of disruption of hepatic PPARγ expression on hepatic fat accumulation and hepatocellular proliferation during liver regeneration. The results showed that liver regeneration was not suppressed, but rather modestly augmented in liver‐specific PPARγ null mice maintained on a normal diet. These animals also exhibited accelerated hepatic cyclin D1 expression. Because hepatic PPARγ expression is increased in experimental models of fatty liver disease in which liver regeneration is impaired, regeneration in liver‐specific PPARγ null mice with chronic hepatic steatosis was also examined. In contrast to the results described above, disruption of hepatic PPARγ expression in mice with diet‐induced hepatic steatosis resulted in significant suppression of hepatic regeneration. Conclusion: The metabolic and hepatocellular proliferative responses to PH are modestly augmented in liver‐specific PPARγ null mice, thus providing additional support for a metabolic model of liver regeneration. Furthermore, regeneration is significantly impaired in liver‐specific PPARγ null mice in the setting of diet‐induced chronic steatosis, suggesting that pharmacological strategies to augment hepatic PPARγ activity might improve regeneration of the fatty liver. (HEPATOLOGY 2012)


American Journal of Pathology | 2012

The Influence of Skeletal Muscle on the Regulation of Liver:Body Mass and Liver Regeneration

Jiansheng Huang; Martin Glauber; Zhaohua Qiu; Vered Gazit; Dennis J. Dietzen; David A. Rudnick

The relationship between liver and body mass is exemplified by the precision with which the liver:body mass ratio is restored after partial hepatic resection. Nevertheless, the compartments, against which liver mass is so exquisitely regulated, currently remain undefined. In the studies reported here, we investigated the role of skeletal muscle mass in the regulation of liver:body mass ratio and liver regeneration via the analysis of myostatin-null mice, in which skeletal muscle is hypertrophied. The results showed that liver mass is comparable and liver:body mass significantly diminished in the null animals compared to age-, sex-, and strain-matched controls. In association with these findings, basal hepatic Akt signaling is decreased, and the expression of the target genes of the constitutive androstane receptor and the integrin-linked kinase are dysregulated in the myostatin-null mice. In addition, the baseline expression levels of the regulators of the G1-S phase cell cycle progression in liver are suppressed in the null mice. The initiation of liver regeneration is not impaired in the null animals, although it progresses toward the lower liver:body mass set point. The data show that skeletal muscle is not the body component against which liver mass is positively regulated, and thus they demonstrate a previously unrecognized systemic compartmental specificity for the regulation of liver:body mass ratio.


Pediatric Research | 2015

Dietary Aflatoxin-Induced Stunting in a Novel Rat Model: Evidence for Toxin-Induced Liver Injury and Hepatic Growth Hormone Resistance

Brittany Knipstein; Jiansheng Huang; Emily Barr; Philip Sossenheimer; Dennis J. Dietzen; Patricia A. Egner; John D. Groopman; David A. Rudnick

Background:Despite a strong statistical correlation between dietary aflatoxin B1 (AFB1)-exposure and childhood stunting, the causal mechanism remains speculative. This issue is important because of emerging interest in reduction of human aflatoxin exposure to diminish the prevalence and complications of stunting. Pediatric liver diseases cause growth impairment, and AFB1 is hepatotoxic. Thus, liver injury might mediate AFB1-associated growth impairment. We have developed a rat model of dietary AFB1-induced stunting to investigate these questions.Methods:Newly-weaned rats were given AFB1-supplemented- or control-diets from age 3–9 wk, and then euthanized for serum- and tissue-collection. Food intake and weight were serially assessed, with tibial-length determined at the experimental endpoint. Serum AFB1-adducts, hepatic gene and protein expression, and liver injury markers were quantified using established methodologies.Results:AFB1-albumin adducts correlated with dietary toxin contamination, but such contamination did not affect food consumption. AFB1-exposed animals exhibited dose-dependent wasting and stunting, liver pathology, and suppression of hepatic targets of growth hormone (GH) signaling, but did not display increased mortality.Conclusion:These data establish toxin-dependent liver injury and hepatic GH-resistance as candidate mechanisms by which AFB1-exposure causes growth impairment in this mammalian model. Interrogation of modifiers of stunting using this model could guide interventions in at-risk and affected children.


Epigenetics | 2014

Identification of an epigenetic signature of early mouse liver regeneration that is disrupted by Zn-HDAC inhibition

Jiansheng Huang; Andrew E. Schriefer; Wei Yang; Paul F. Cliften; David A. Rudnick

Liver regeneration has been well studied with hope of discovering strategies to improve liver disease outcomes. Nevertheless, the signals that initiate such regeneration remain incompletely defined, and translation of mechanism-based pro-regenerative interventions into new treatments for hepatic diseases has not yet been achieved. We previously reported the isoform-specific regulation and essential function of zinc-dependent histone deacetylases (Zn-HDACs) during mouse liver regeneration. Those data suggest that epigenetically regulated anti-proliferative genes are deacetylated and transcriptionally suppressed by Zn-HDAC activity or that pro-regenerative factors are acetylated and induced by such activity in response to partial hepatectomy (PH). To investigate these possibilities, we conducted genome-wide interrogation of the liver histone acetylome during early PH-induced liver regeneration in mice using acetyL-histone chromatin immunoprecipitation and next generation DNA sequencing. We also compared the findings of that study to those seen during the impaired regenerative response that occurs with Zn-HDAC inhibition. The results reveal an epigenetic signature of early liver regeneration that includes both hyperacetylation of pro-regenerative factors and deacetylation of anti-proliferative and pro-apoptotic genes. Our data also show that administration of an anti-regenerative regimen of the Zn-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) not only disrupts gene-specific pro-regenerative changes in liver histone deacetylation but also reverses PH-induced effects on histone hyperacetylation. Taken together, these studies offer new insight into and suggest novel hypotheses about the epigenetic mechanisms that regulate liver regeneration.


American Journal of Pathology | 2016

Postponing the Hypoglycemic Response to Partial Hepatectomy Delays Mouse Liver Regeneration

Jiansheng Huang; Andrew E. Schriefer; Paul F. Cliften; Dennis J. Dietzen; Sakil Kulkarni; Sucha Sing; Satdarshan P.S. Monga; David A. Rudnick

All serious liver injuries alter metabolism and initiate hepatic regeneration. Recent studies using partial hepatectomy (PH) and other experimental models of liver regeneration implicate the metabolic response to hepatic insufficiency as an important source of signals that promote regeneration. Based on these considerations, the analyses reported here were undertaken to assess the impact of interrupting the hypoglycemic response to PH on liver regeneration in mice. A regimen of parenteral dextrose infusion that delays PH-induced hypoglycemia for 14 hours after surgery was identified, and the hepatic regenerative response to PH was compared between dextrose-treated and control mice. The results showed that regenerative recovery of the liver was postponed in dextrose-infused mice (versus vehicle control) by an interval of time comparable to the delay in onset of PH-induced hypoglycemia. The regulation of specific liver regeneration-promoting signals, including hepatic induction of cyclin D1 and S-phase kinase-associated protein 2 expression and suppression of peroxisome proliferator-activated receptor γ and p27 expression, was also disrupted by dextrose infusion. These data support the hypothesis that alterations in metabolism that occur in response to hepatic insufficiency promote liver regeneration, and they define specific pro- and antiregenerative molecular targets whose regenerative regulation is postponed when PH-induced hypoglycemia is delayed.


Current Pathobiology Reports | 2015

Elucidating Metabolic and Epigenetic Mechanisms that Regulate Liver Regeneration

Jiansheng Huang; David A. Rudnick

The regenerative capability of the liver is essential for recovery from all hepatic injuries. Although long studied, the signals that regulate such regeneration require further elucidation if knowledge about regenerative mechanisms is to be translated into improved clinical therapy. Alterations in metabolism have been the focus of recent experimental investigations as a possible source of essential signals that control liver regeneration. Although the specific mechanisms linking metabolism and regeneration remain unknown, specific growth factors, secondary messengers, and transcription factors have been suggested by published analyses. Epigenetic mechanisms are also emerging as potential intermediaries between hepatic insufficiency-induced changes in metabolism and regenerative hepatocellular proliferation. This article reviews the recent literature relevant to these considerations, with particular emphasis on contemporary data that link metabolic and epigenetic signals to the regulation of liver regulation. The relevance of metabolic–epigenetic regulation of experimental hepatic regeneration with respect to human liver diseases is also briefly considered.


Liver Regeneration#R##N#Basic Mechanisms, Relevant Models and Clinical Applications | 2015

Chapter 15 – Metabolic Regulation of Liver Regeneration

David A. Rudnick; Jiansheng Huang

Abstract All serious liver injuries alter metabolism. Those injuries also initiate hepatic regeneration. Experimental analyses using rodent partial hepatectomy and other models implicate the metabolic changes that occur in response to hepatic injury as vital regulators of liver regeneration. These studies also show that disrupting certain elements of the stereotypical metabolic response to hepatic insufficiency suppresses liver regeneration, while augmenting components of the response can accelerate regeneration. Such data raise the possibility that specific metabolic interventions might promote human liver regeneration for therapeutic benefit in liver disease. This chapter provides an update to recent reviews considering the functional connections between metabolism and liver regeneration and includes an overview of the current evidence in support of regenerative functions of liver injury-induced alterations in metabolism, discussions about important areas for further study, and reflections on provocative clinical implications raised by these considerations.

Collaboration


Dive into the Jiansheng Huang's collaboration.

Top Co-Authors

Avatar

David A. Rudnick

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dennis J. Dietzen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Paul F. Cliften

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Andrew E. Schriefer

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Emily Barr

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sakil Kulkarni

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vered Gazit

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alexander Weymann

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brittany Knipstein

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge