Network


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

Hotspot


Dive into the research topics where Samjhana Thapaliya is active.

Publication


Featured researches published by Samjhana Thapaliya.


Journal of Biological Chemistry | 2010

Adipocyte Apoptosis, a Link between Obesity, Insulin Resistance, and Hepatic Steatosis

Naim Alkhouri; Agnieszka Gornicka; Michael Berk; Samjhana Thapaliya; Laura J. Dixon; Sangeeta R. Kashyap; Philip R. Schauer; Ariel E. Feldstein

Adipocyte death has been reported in both obese humans and rodents. However, its role in metabolic disorders, including insulin resistance, hepatic steatosis, and inflammation associated with obesity has not been studied. We now show using real-time reverse transcription-PCR arrays that adipose tissue of obese mice display a pro-apoptotic phenotype. Moreover, caspase activation and adipocyte apoptosis were markedly increased in adipose tissue from both mice with diet-induced obesity and obese humans. These changes were associated with activation of both the extrinsic, death receptor-mediated, and intrinsic, mitochondrial-mediated pathways of apoptosis. Genetic inactivation of Bid, a key pro-apoptotic molecule that serves as a link between these two cell death pathways, significantly reduced caspase activation, adipocyte apoptosis, prevented adipose tissue macrophage infiltration, and protected against the development of systemic insulin resistance and hepatic steatosis independent of body weight. These data strongly suggest that adipocyte apoptosis is a key initial event that contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis associated with obesity in both mice and humans. Inhibition of adipocyte apoptosis may be a new therapeutic strategy for the treatment of obesity-associated metabolic complications.


Laboratory Investigation | 2012

Caspase 1-mediated regulation of fibrogenesis in diet-induced steatohepatitis

Laura J. Dixon; Michael Berk; Samjhana Thapaliya; Bettina G. Papouchado; Ariel E. Feldstein

Non-alcoholic steatohepatitis (NASH) is typically associated with pro-apoptotic caspase activation. A potential role for pro-inflammatory caspases remains incompletely understood. Our aims were to examine a potential role of caspase-1 in the development of liver damage and fibrosis in NASH. C57BL/6 wild type (WT) developed marked steatohepatitis, activation, fibrosis and increased hepatic caspase-1 and interleukin-1β expression when placed on the methionine- and choline-deficient (MCD) diet. Marked caspase-1 activation was detected in the liver of MCD-fed mice. Hepatocyte and non-parenchymal fractionation of the livers further demonstrated that caspase-1 activation after MCD feeding was mainly localized to non-parenchymal cells. Caspase-1-knockout (Casp1−/−) mice on the MCD diet showed marked reduction in mRNA expression of genes involved in inflammation and fibrogenesis (tumor necrosis factor-α was 7.6-fold greater in WT vs Casp1−/− MCD-fed mice; F4/80 was 1.5-fold greater in WT vs Casp1−/− MCD-fed mice; α-smooth muscle actin was 3.2-fold greater in WT vs Casp1−/− MCD-fed mice; collagen 1-α was 7.6-fold greater in WT vs Casp1−/− MCD-fed mice; transforming growth factor-β was 2.4-fold greater in WT vs Casp1−/− MCD-fed mice; cysteine- and glycine-rich protein 2 was 3.2-fold greater in WT vs Casp1−/− MCD-fed mice). Furthermore, Sirius red staining for hepatic collagen deposition was significantly reduced in Casp1−/− MCD-fed mice compared with WT MCD-fed animals. However, serum alanine aminotransferase levels, caspase-3 activity and terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells were similar in Casp1−/− and WT mice on the MCD diet. Selective Kupffer cell depletion by clodronate injection markedly suppressed MCD-induced caspase-1 activation and protected mice from fibrogenesis and fibrosis associated with this diet. The conclusion of this study is that it uncovers a novel role for caspase-1 in inflammation and fibrosis during NASH development.


Science Signaling | 2013

Lipid-Induced Toxicity Stimulates Hepatocytes to Release Angiogenic Microparticles That Require Vanin-1 for Uptake by Endothelial Cells

Davide Povero; Akiko Eguchi; Niesman Ir; Andronikou N; de Mollerat du Jeu X; Anny Mulya; Michael Berk; Milos Lazic; Samjhana Thapaliya; Maurizio Parola; Patel Hh; Ariel E. Feldstein

Fat-overloaded hepatocytes release microparticles that induce angiogenesis and worsening of fatty liver disease. Sending an Angiogenic Message Excess amounts of saturated fatty acids are a potential dietary trigger for the fatty liver disease steatohepatitis, in which the liver develops fat deposits and inflammation. Progression of the disease to more serious forms, which can include scarring and other serious complications, is associated with the formation of new blood vessels, a process called angiogenesis, which requires endothelial cells to migrate and form tubular structures. Povero et al. found that a hepatocyte cell line exposed to excess amounts of saturated fatty acids released membrane-bound microparticles that induced angiogenesis when administered to mice. Microparticles from the blood of mice with diet-induced steatohepatitis originated from the liver and triggered migration and tubular structure formation when applied to an endothelial cell line. The angiogenic effects of microparticles generated by a hepatocyte cell line exposed to saturated fatty acids or of those from mice with diet-induced steatohepatitis involved the uptake of the microparticles by endothelial cells, a process that required Vanin-1, an enzyme located on the surface of the microparticles. Thus, the pathological angiogenesis that can occur in steatohepatitis could be reduced by preventing endothelial cells from internalizing Vanin-1–positive microparticles from hepatocytes. Angiogenesis is a key pathological feature of experimental and human steatohepatitis, a common chronic liver disease that is associated with obesity. We demonstrated that hepatocytes generated a type of membrane-bound vesicle, microparticles, in response to conditions that mimicked the lipid accumulation that occurs in the liver in some forms of steatohepatitis and that these microparticles promoted angiogenesis. When applied to an endothelial cell line, medium conditioned by murine hepatocytes or a human hepatocyte cell line exposed to saturated free fatty acids induced migration and tube formation, two processes required for angiogenesis. Medium from hepatocytes in which caspase 3 was inhibited or medium in which the microparticles were removed by ultracentrifugation lacked proangiogenic activity. Isolated hepatocyte-derived microparticles induced migration and tube formation of an endothelial cell line in vitro and angiogenesis in mice, processes that depended on internalization of microparticles. Microparticle internalization required the interaction of the ectoenzyme Vanin-1 (VNN1), an abundant surface protein on the microparticles, with lipid raft domains of endothelial cells. Large quantities of hepatocyte-derived microparticles were detected in the blood of mice with diet-induced steatohepatitis, and microparticle quantity correlated with disease severity. Genetic ablation of caspase 3 or RNA interference directed against VNN1 protected mice from steatohepatitis-induced pathological angiogenesis in the liver and resulted in a loss of the proangiogenic effects of microparticles. Our data identify hepatocyte-derived microparticles as critical signals that contribute to angiogenesis and liver damage in steatohepatitis and suggest a therapeutic target for this condition.


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

Hyperammonemia in cirrhosis induces transcriptional regulation of myostatin by an NF-κB–mediated mechanism

Jia Qiu; Samjhana Thapaliya; Ashok Runkana; Yu Yang; Cynthia Tsien; Arvind Narayanan; Bijan Eghtesad; Paul E. Mozdziak; Christine McDonald; George R. Stark; Stephen Welle; Sathyamangla V. Naga Prasad; Srinivasan Dasarathy

Significance Loss of skeletal muscle mass, or sarcopenia, is nearly universal in cirrhosis and adversely affects the outcome of these patients. There are no established therapies to prevent or reverse sarcopenia because the mechanisms are not known. We show that the expression of myostatin, a negative regulator of skeletal muscle mass, is increased in the cirrhotic muscle and is mediated by increased ammonia concentration. Skeletal muscle ammonia concentrations are significantly increased in cirrhosis, resulting in activation of the transcription factor NF-κB, which in turn increases the expression of myostatin. Given the high prevalence of cirrhosis, these studies are of broad general interest because ammonia-lowering strategies, NF-κB antagonists, and myostatin blocker are potential therapies to reverse sarcopenia of cirrhosis. Loss of muscle mass, or sarcopenia, is nearly universal in cirrhosis and adversely affects patient outcome. The underlying cross-talk between the liver and skeletal muscle mediating sarcopenia is not well understood. Hyperammonemia is a consistent abnormality in cirrhosis due to impaired hepatic detoxification to urea. We observed elevated levels of ammonia in both plasma samples and skeletal muscle biopsies from cirrhotic patients compared with healthy controls. Furthermore, skeletal muscle from cirrhotics had increased expression of myostatin, a known inhibitor of skeletal muscle accretion and growth. In vivo studies in mice showed that hyperammonemia reduced muscle mass and strength and increased myostatin expression in wild-type compared with postdevelopmental myostatin knockout mice. We postulated that hyperammonemia is an underlying link between hepatic dysfunction in cirrhosis and skeletal muscle loss. Therefore, murine C2C12 myotubes were treated with ammonium acetate resulting in intracellular concentrations similar to those in cirrhotic muscle. In this system, we demonstrate that hyperammonemia stimulated myostatin expression in a NF-κB–dependent manner. This finding was also observed in primary murine muscle cell cultures. Hyperammonemia triggered activation of IκB kinase, NF-κB nuclear translocation, binding of the NF-κB p65 subunit to specific sites within the myostatin promoter, and stimulation of myostatin gene transcription. Pharmacologic inhibition or gene silencing of NF-κB abolished myostatin up-regulation under conditions of hyperammonemia. Our work provides unique insights into hyperammonemia-induced myostatin expression and suggests a mechanism by which sarcopenia develops in cirrhotic patients.


Hepatology | 2015

Metabolic and molecular responses to leucine‐enriched branched chain amino acid supplementation in the skeletal muscle of alcoholic cirrhosis

Cynthia Tsien; Gangarao Davuluri; Dharmvir Singh; Allawy Allawy; Gabriella A.M. Ten Have; Samjhana Thapaliya; John M. Schulze; David S. Barnes; Arthur J. McCullough; M.P. Engelen; Nicolaas E. P. Deutz; Srinivasan Dasarathy

Skeletal muscle loss (sarcopenia) is a major clinical complication in alcoholic cirrhosis with no effective therapy. Skeletal muscle autophagic proteolysis and myostatin expression (inhibitor of protein synthesis) are increased in cirrhosis and believed to contribute to anabolic resistance. A prospective study was performed to determine the mechanisms of sarcopenia in alcoholic cirrhosis and potential reversal by leucine. In six well‐compensated, stable, alcoholic patients with cirrhosis and eight controls, serial vastus lateralis muscle biopsies were obtained before and 7 hours after a single oral branched chain amino acid mixture enriched with leucine (BCAA/LEU). Primed‐constant infusion of l‐[ring‐2H5]‐phenylalanine was used to quantify whole‐body protein breakdown and muscle protein fractional synthesis rate using liquid chromatography/mass spectrometry. Muscle expression of myostatin, mammalian target of rapamycin (mTOR) targets, autophagy markers, protein ubiquitination, and the intracellular amino acid deficiency sensor general control of nutrition 2 were quantified by immunoblots and the leucine exchanger (SLC7A5) and glutamine transporter (SLC38A2), by real‐time polymerase chain reaction. Following oral administration, plasma BCAA concentrations showed a similar increase in patients with cirrhosis and controls. Skeletal muscle fractional synthesis rate was 9.63 ± 0.36%/hour in controls and 9.05 ± 0.68%/hour in patients with cirrhosis (P = 0.54). Elevated whole‐body protein breakdown in patients with cirrhosis was reduced with BCAA/LEU (P = 0.01). Fasting skeletal muscle molecular markers showed increased myostatin expression, impaired mTOR signaling, and increased autophagy in patients with cirrhosis compared to controls (P < 0.01). The BCAA/LEU supplement did not alter myostatin expression, but mTOR signaling, autophagy measures, and general control of nutrition 2 activation were consistently reversed in cirrhotic muscle (P < 0.01). Expression of SLC7A5 was higher in the basal state in patients with cirrhosis than controls (P < 0.05) but increased with BCAA/LEU only in controls (P < 0.001). Conclusions: Impaired mTOR1 signaling and increased autophagy in skeletal muscle of patients with alcoholic cirrhosis is acutely reversed by BCAA/LEU. (Hepatology 2015;61:2018‐2029)


Autophagy | 2014

Alcohol-induced autophagy contributes to loss in skeletal muscle mass

Samjhana Thapaliya; Ashok Runkana; Megan R. McMullen; Laura E. Nagy; Christine McDonald; Sathyamangla V. Naga Prasad; Srinivasan Dasarathy

Patients with alcoholic cirrhosis and hepatitis have severe muscle loss. Since ethanol impairs skeletal muscle protein synthesis but does not increase ubiquitin proteasome-mediated proteolysis, we investigated whether alcohol-induced autophagy contributes to muscle loss. Autophagy induction was studied in: A) Human skeletal muscle biopsies from alcoholic cirrhotics and controls, B) Gastrocnemius muscle from ethanol and pair-fed mice, and C) Ethanol-exposed murine C2C12 myotubes, by examining the expression of autophagy markers assessed by immunoblotting and real-time PCR. Expression of autophagy genes and markers were increased in skeletal muscle from humans and ethanol-fed mice, and in myotubes following ethanol exposure. Importantly, pulse-chase experiments showed suppression of myotube proteolysis upon ethanol-treatment with the autophagy inhibitor, 3-methyladenine (3MA) and not by MG132, a proteasome inhibitor. Correspondingly, ethanol-treated C2C12 myotubes stably expressing GFP-LC3B showed increased autophagy flux as measured by accumulation of GFP-LC3B vesicles with confocal microscopy. The ethanol-induced increase in LC3B lipidation was reversed upon knockdown of Atg7, a critical autophagy gene and was associated with reversal of the ethanol-induced decrease in myotube diameter. Consistently, CT image analysis of muscle area in alcoholic cirrhotics was significantly reduced compared with control subjects. In order to determine whether ethanol per se or its metabolic product, acetaldehyde, stimulates autophagy, C2C12 myotubes were treated with ethanol in the presence of the alcohol dehydrogenase inhibitor (4-methylpyrazole) or the acetaldehyde dehydrogenase inhibitor (cyanamide). LC3B lipidation increased with acetaldehyde treatment and increased further with the addition of cyanamide. We conclude that muscle autophagy is increased by ethanol exposure and contributes to sarcopenia.


Antioxidants & Redox Signaling | 2011

Transcriptional Profile of Genes Involved in Oxidative Stress and Antioxidant Defense in a Dietary Murine Model of Steatohepatitis

Agnieszka Gornicka; Gareth Morris-Stiff; Samjhana Thapaliya; Bettina G. Papouchado; Michael Berk; Ariel E. Feldstein

Oxidative stress is a core abnormality responsible for disease progression in nonalcoholic steatohepatitis (NASH). However, the relevant pathways that contribute to oxidative damage in vivo remain poorly understood. Here we explore the gene-expression patterns related to oxidative stress, antioxidant defense, and reactive oxygen metabolism in an established dietary murine model of NASH. C57BL/6 mice were placed on either a methionine- and choline-deficient (MCD) or a control (CTL) diet for 6 weeks. Hepatic oxidative damage and the development of NASH were monitored by biochemical and histologic indices. Analysis of 84 oxidative stress-related genes was performed by real-time reverse transcription polymerase chain reaction (PCR) in the livers of the two groups of mice. Mice on the MCD diet showed increased ALT, histologic features of NASH, and oxidative liver damage with increases in 4-hydroxynonenal and 3-nitrotyrosine. Of the genes analyzed, the GPx family were most significantly upregulated, whereas SCD1 was most significantly downregulated. Other genes that were significantly upregulated included Fmo2 and peroxiredoxins, whereas genes downregulated included Catalase and Serpinb1b. Our data demonstrate that oxidative stress-related genes are differentially expressed in the livers of mice with diet-induced NASH. These findings have important implications for NASH pathogenesis and the development of novel therapeutic strategies for patients with this condition.


Journal of Hepatology | 2016

Metabolic adaptation of skeletal muscle to hyperammonemia drives the beneficial effects of l-leucine in cirrhosis

Gangarao Davuluri; Dawid Krokowski; Bo Jhih Guan; Avinash Kumar; Samjhana Thapaliya; Dharmvir Singh; Maria Hatzoglou; Srinivasan Dasarathy

BACKGROUND & AIMS Increased skeletal muscle ammonia uptake with loss of muscle mass adversely affects clinical outcomes in cirrhosis. Hyperammonemia causes reduced protein synthesis and sarcopenia but the cellular responses to impaired proteostasis and molecular mechanism of l-leucine induced adaptation to ammonia induced stress were determined. METHODS Response to activation of amino acid deficiency sensor, GCN2, in the skeletal muscle from cirrhotic patients and the portacaval anastomosis (PCA) rat were quantified. During hyperammonemia and l-leucine supplementation, protein synthesis, phosphorylation of eIF2α, mTORC1 signaling, l-leucine transport and response to l-leucine supplementation were quantified. Adaptation to cellular stress via ATF4 and its target GADD34 were also determined. RESULTS Activation of the eIF2α kinase GCN2 and impaired mTORC1 signaling were observed in skeletal muscle from cirrhotic patients and PCA rats. Ammonia activated GCN2 mediated eIF2α phosphorylation (eIF2α-P) and impaired mTORC1 signaling that inhibit protein synthesis in myotubes and MEFs. Adaptation to ammonia induced stress did not involve translational reprogramming by activation transcription factor 4 (ATF4) dependent induction of the eIF2α-P phosphatase subunit GADD34. Instead, ammonia increased expression of the leucine/glutamine exchanger SLC7A5, l-leucine uptake and intracellular l-leucine levels, the latter not being sufficient to rescue the inhibition of protein synthesis, due to potentially enhanced mitochondrial sequestration of l-leucine. l-leucine supplementation rescued protein synthesis inhibition caused by hyperammonemia. CONCLUSIONS Response to hyperammonemia is reminiscent of the cellular response to amino acid starvation, but lacks the adaptive ATF4 dependent integrated stress response (ISR). Instead, hyperammonemia-induced l-leucine uptake was an adaptive response to the GCN2-mediated decreased protein synthesis. LAY SUMMARY Sarcopenia or skeletal muscle loss is the most frequent complication in cirrhosis but there are no treatments because the cause(s) of muscle loss in liver disease are not known. Results from laboratory experiments in animals and muscle cells were validated in human patients with cirrhosis to show that ammonia plays a key role in causing muscle loss in patients with cirrhosis. We identified a novel stress response to ammonia in the muscle that decreases muscle protein content that can be reversed by supplementation with the amino acid l-leucine.


The Journal of Physiology | 2016

Hyperammonaemia-induced skeletal muscle mitochondrial dysfunction results in cataplerosis and oxidative stress

Gangarao Davuluri; Allawy Allawy; Samjhana Thapaliya; Julie H. Rennison; Dharmvir Singh; Avinash Kumar; Yana Sandlers; David R. Van Wagoner; Chris A. Flask; Charles L. Hoppel; Takhar Kasumov; Srinivasan Dasarathy

Hyperammonaemia occurs in hepatic, cardiac and pulmonary diseases with increased muscle concentration of ammonia. We found that ammonia results in reduced skeletal muscle mitochondrial respiration, electron transport chain complex I dysfunction, as well as lower NAD+/NADH ratio and ATP content. During hyperammonaemia, leak of electrons from complex III results in oxidative modification of proteins and lipids. Tricarboxylic acid cycle intermediates are decreased during hyperammonaemia, and providing a cell‐permeable ester of αKG reversed the lower TCA cycle intermediate concentrations and increased ATP content. Our observations have high clinical relevance given the potential for novel approaches to reverse skeletal muscle ammonia toxicity by targeting the TCA cycle intermediates and mitochondrial ROS.


Gastroenterology | 2010

S1834 An Atherogenic Diet Induces Steatohepatitis in Mice

Agnieszka Gornicka; Samjhana Thapaliya; Michael Berk; Ariel E. Feldstein

Background and aims: Metabolic syndrome-related NASH is one of the emerging healththreatening problems in industrialized countries worldwide. Despite of numerous experimental/clinical challenges, therapeutic strategies for NASH have not been well established. In this study, we investigated the therapeutic effect of ursolic acid, a pentacyclic triterpene acid, on high-fat diet (HFD)-induced steatohepatitis in obese, diabetic KK-Ay mice.Methods: Male, 8-week old KK-Aymice were fed a diet containing 32% fat for 4 weeks, and subsequently given daily intra-gastric injections of UA (100 mg/kg BW, suspended in 0.5% metyl-cellulose solution) or vehicle alone in combination with continuous HFD-feeding for up to 4 weeks. Liver histology was assessed by H-E staining. Serum aminotransferases and tissue triglyceride levels were determined colorimetrically by standard enzymatic methods. Steady state mRNA levels for SREBP-1c, fatty acid synthase (FAS), enoyl-CoA hydratase (ECHD) and acyl-CoA oxidase 1 (ACOX1) in the liver were analyzed quantitatively by real-time RT-PCR. Results: All KK-Ay mice gained body weight constantly during 4-week pre-feeding period, and the control mice continued gaining body weight over 10% during the subsequent 4-week treatment period; however, mice given UA lost body weight nearly 7% (P<0.001 vs. control group) despite of the equivalent dietary consumption. Except for body weight loss, mice treated with UA appeared to be healthy. KK-Ay mice developed remarkable steatohepatitis following 4-week pre-feeding with HFD as expected; however, treatment with UA dramatically improved liver pathology within 1 week, and the effect was sustained for 4-week treatment period. Indeed, serum transaminases levels, as well as hepatic triglyceride contents, were decreased significantly in mice treated with UA. Interestingly, mice treated with UA showed significantly decreased expression of hepatic SREBP-1c and FAS mRNA levels, indicating that fatty acid synthesis is down-regulated by UA. Further, hepatic mRNA levels of β-oxidation enzymes such as ECHD and ACOX1 were also significantly lower in UAtreated mice.Conclusions: These findings clearly demonstrated that ursolic acid significantly improves HFD-induced severe steatohepatitis in KK-Ay mice without dietary restriction. The mechanisms underlying this therapeutic effect of UA most likely involve decreased fatty acid synthesis and radical-generating β-oxidation in the liver. Since this chemical is an active ingredient of herbal medicine, it is postulated that UA is useful for a new preventive/ therapeutic reagent for NASH.

Collaboration


Dive into the Samjhana Thapaliya'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

Akiko Eguchi

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge