Network


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

Hotspot


Dive into the research topics where Tanya Tarnovscki is active.

Publication


Featured researches published by Tanya Tarnovscki.


The Journal of Clinical Endocrinology and Metabolism | 2010

Altered autophagy in human adipose tissues in obesity.

Julia Kovsan; Matthias Blüher; Tanya Tarnovscki; Nora Klöting; Boris Kirshtein; Liron Madar; Iris Shai; Rachel Golan; Ilana Harman-Boehm; Michael R. Schön; Andrew S. Greenberg; Zvulun Elazar; Nava Bashan; Assaf Rudich

CONTEXT Autophagy is a housekeeping mechanism, involved in metabolic regulation and stress response, shown recently to regulate lipid droplets biogenesis/breakdown and adipose tissue phenotype. OBJECTIVE We hypothesized that in human obesity autophagy may be altered in adipose tissue in a fat depot and distribution-dependent manner. SETTING AND PATIENTS Paired omental (Om) and subcutaneous (Sc) adipose tissue samples were used from obese and nonobese (n = 65, cohort 1); lean, Sc-obese and intraabdominally obese (n = 196, cohort 2); severely obese persons without diabetes or obesity-associated morbidity, matched for being insulin sensitive or resistant (n = 60, cohort 3). RESULTS Protein and mRNA levels of the autophagy genes Atg5, LC3A, and LC3B were increased in Om compared with Sc, more pronounced among obese persons, particularly with intraabdominal fat accumulation. Both adipocytes and stromal-vascular cells contribute to the expression of autophagy genes. An increased number of autophagosomes and elevated autophagic flux assessed in fat explants incubated with lysosomal inhibitors were observed in obesity, particularly in Om. The degree of visceral adiposity and adipocyte hypertrophy accounted for approximately 50% of the variance in omental Atg5 mRNA levels by multivariate regression analysis, whereas age, sex, measures of insulin sensitivity, inflammation, and adipose tissue stress were excluded from the model. Moreover, in cohort 3, the autophagy marker genes were increased in those who were insulin resistant compared with insulin sensitive, particularly in Om. CONCLUSIONS Autophagy is up-regulated in adipose tissue of obese persons, especially in Om, correlating with the degree of obesity, visceral fat distribution, and adipocyte hypertrophy. This may co-occur with insulin resistance but precede the occurrence of obesity-associated morbidity.


PLOS ONE | 2013

Interleukin-1β regulates fat-liver crosstalk in obesity by auto-paracrine modulation of adipose tissue inflammation and expandability.

Ori Nov; Hagit Shapiro; Hilla Ovadia; Tanya Tarnovscki; Irit Dvir; Elad Shemesh; Julia Kovsan; Ilan Shelef; Yaron Carmi; Elena Voronov; Ron N. Apte; Eli C. Lewis; Yulia Haim; Daniel Konrad; Nava Bashan; Assaf Rudich

The inflammasome has been recently implicated in obesity-associated dys-metabolism. However, of its products, the specific role of IL-1β was clinically demonstrated to mediate only the pancreatic beta-cell demise, and in mice mainly the intra-hepatic manifestations of obesity. Yet, it remains largely unknown if IL-1β, a cytokine believed to mainly function locally, could regulate dysfunctional inter-organ crosstalk in obesity. Here we show that High-fat-fed (HFF) mice exhibited a preferential increase of IL-1β in portal compared to systemic blood. Moreover, portally-drained mesenteric fat transplantation from IL-1βKO donors resulted in lower pyruvate-glucose flux compared to mice receiving wild-type (WT) transplant. These results raised a putative endocrine function for visceral fat-derived IL-1β in regulating hepatic gluconeogenic flux. IL-1βKO mice on HFF exhibited only a minor or no increase in adipose expression of pro-inflammatory genes (including macrophage M1 markers), Mac2-positive crown-like structures and CD11b-F4/80-double-positive macrophages, all of which were markedly increased in WT-HFF mice. Further consistent with autocrine/paracrine functions of IL-1β within adipose tissue, adipose tissue macrophage lipid content was increased in WT-HFF mice, but significantly less in IL-1βKO mice. Ex-vivo, adipose explants co-cultured with primary hepatocytes from WT or IL-1-receptor (IL-1RI)-KO mice suggested only a minor direct effect of adipose-derived IL-1β on hepatocyte insulin resistance. Importantly, although IL-1βKOs gained weight similarly to WT-HFF, they had larger fat depots with similar degree of adipocyte hypertrophy. Furthermore, adipogenesis genes and markers (pparg, cepba, fabp4, glut4) that were decreased by HFF in WT, were paradoxically elevated in IL-1βKO-HFF mice. These local alterations in adipose tissue inflammation and expansion correlated with a lower liver size, less hepatic steatosis, and preserved insulin sensitivity. Collectively, we demonstrate that by promoting adipose inflammation and limiting fat tissue expandability, IL-1β supports ectopic fat accumulation in hepatocytes and adipose-tissue macrophages, contributing to impaired fat-liver crosstalk in nutritional obesity.


American Journal of Physiology-endocrinology and Metabolism | 2009

Depot-specific adipocyte cell lines reveal differential drug-induced responses of white adipocytes--relevance for partial lipodystrophy.

Julia Kovsan; Alexander Osnis; Adva Maissel; Livnat Mazor; Tanya Tarnovscki; Liat Hollander; Shira Ovadia; Britta Meier; Johannes Klein; Nava Bashan; Assaf Rudich

Intra-abdominal (IA) fat functionally differs from subcutaneous (SC) adipose tissue, likely contributing to its stronger association with obesity-induced morbidity and to differential response to medications. Drug-induced partial lipodystrophy, like in response to antiretroviral agents, is an extreme manifestation of the different response of different fat depots, with loss of SC but not IA. Investigating depot-specific adipocyte differences is limited by the low accessibility to IA fat and by the heterogenous cell population comprising adipose tissue. Here, we aimed at utilizing immortalized preadipocyte cell lines from IA (epididymal) or SC (inguinal) fat to investigate whether they differentially respond to the HIV protease inhibitor nelfinavir. Preadipocytes were readily amenable to adipogenesis, as evidenced by lipid accumulation, expression of adipose-specific genes, measurable lipolysis, and insulin responsiveness. Leptin secretion was higher by the SC line, consistent with known differences between IA and SC fat. As previously reported, nelfinavir inhibited adipogenesis downstream of C/EBPbeta, but similarly in both cell lines. In contrast, nelfinavirs capacity to diminish insulin signaling, decrease leptin secretion, enhance basal lipolysis, and decrease expression of the lipid droplet-associated protein perilipin occurred more robustly and/or at lower nelfinavir concentrations in the SC line. This was despite similar intracellular concentrations of nelfinavir (23.8 +/- 5.6 and 33.6 +/- 12.2 microg/mg protein for inguinal and epididymal adipocytes, respectively, P = 0.46). The cell lines recapitulated depot-differential effects of nelfinavir observed in differentiated primary preadipocytes and with whole tissue explants. Thus, we report the use of fat depot-specific adipocyte cell lines for unraveling depot-differential responses to a drug causing partial lipodystrophy.


PLOS ONE | 2013

Secreted Human Adipose Leptin Decreases Mitochondrial Respiration in HCT116 Colon Cancer Cells

Einav Yehuda-Shnaidman; Lili Nimri; Tanya Tarnovscki; Boris Kirshtein; Assaf Rudich; Betty Schwartz

Obesity is a key risk factor for the development of colon cancer; however, the endocrine/paracrine/metabolic networks mediating this connection are poorly understood. Here we hypothesize that obesity results in secreted products from adipose tissue that induce malignancy-related metabolic alterations in colon cancer cells. Human HCT116 colon cancer cells, were exposed to conditioned media from cultured human adipose tissue fragments of obese vs. non-obese subjects. Oxygen consumption rate (OCR, mostly mitochondrial respiration) and extracellular acidification rate (ECAR, mostly lactate production via glycolysis) were examined vis-à-vis cell viability and expression of related genes and proteins. Our results show that conditioned media from obese (vs. non-obese) subjects decreased basal (40%, p<0.05) and maximal (50%, p<0.05) OCR and gene expression of mitochondrial proteins and Bax without affecting cell viability or expression of glycolytic enzymes. Similar changes could be recapitulated by incubating cells with leptin, whereas, leptin-receptor specific antagonist inhibited the reduced OCR induced by conditioned media from obese subjects. We conclude that secreted products from the adipose tissue of obese subjects inhibit mitochondrial respiration and function in HCT116 colon cancer cells, an effect that is at least partly mediated by leptin. These results highlight a putative novel mechanism for obesity-associated risk of gastrointestinal malignancies, and suggest potential new therapeutic avenues.


Journal of Endocrinology | 2017

Adipose tissue supports normalization of macrophage and liver lipid handling in obesity reversal

Maayan Vatarescu; Sapir Bechor; Yulia Haim; Tal Pecht; Tanya Tarnovscki; Ori Nov; Hagit Shapiro; Avishai Shemesh; Angel Porgador; Nava Bashan; Assaf Rudich

Adipose tissue inflammation and dysfunction are considered central in the pathogenesis of obesity-related dysmetabolism, but their role in the rapid metabolic recovery upon obesity reversal is less well defined. We hypothesized that changes in adipose tissue endocrine and paracrine mechanisms may support the rapid improvement of obesity-induced impairment in cellular lipid handling. C57Bl-6J mice were fed ad libitum either normal chow (NC) or high-fat diet (HFF) for 10 weeks. A dietary obesity reversal group was fed HFF for 8 weeks and then switched to NC for 2 weeks (HFF→NC). Whole-body glucose homeostasis rapidly nearly normalized in the HFF→NC mice (fasting glucose and insulin fully normalized, glucose and insulin tolerance tests reversed 82% to the NC group levels). During 2 weeks of the dietary reversal, the liver was significantly cleared from ectopic fat, and functionally, glucose production from pyruvate, alanine or fructose was normalized. In contrast, adipose tissue inflammation (macrophage infiltration and polarization) largely remained as in HFF, though obesity-induced adipose tissue macrophage lipid accumulation decreased by ~50%, and adipose tissue MAP kinase hyperactivation was reversed. Ex vivo, mild changes in adipose tissue adipocytokine secretion profile were noted. These corresponded to partial or full reversal of the excess cellular lipid droplet accumulation induced by HFF adipose tissue conditioned media in hepatoma or macrophage cells, respectively. We propose that early after initiating reversal of nutritional obesity, rapid metabolic normalization largely precedes resolution of adipose tissue inflammation. Nevertheless, we demonstrate a hitherto unrecognized contribution of adipose tissue to the rapid improvement in lipid handling by the liver and by macrophages.


Endocrinology | 2007

Mitogen-Activated Protein Kinases, Inhibitory-κB Kinase, and Insulin Signaling in Human Omental Versus Subcutaneous Adipose Tissue in Obesity

Nava Bashan; Karina Dorfman; Tanya Tarnovscki; Ilana Harman-Boehm; Idit F. Liberty; Matthias Blüher; Shira Ovadia; Tali Maymon-Zilberstein; R. Potashnik; Michael Stumvoll; Eliezer Avinoach; Assaf Rudich


Autophagy | 2015

Elevated autophagy gene expression in adipose tissue of obese humans: A potential non-cell-cycle-dependent function of E2F1

Yulia Haim; Matthias Blüher; Nir Goldstein; Nora Klöting; Ilana Harman-Boehm; Boris Kirshtein; Doron Ginsberg; Martin Gericke; Esther Guiu Jurado; Julia Kovsan; Tanya Tarnovscki; Leonid Kachko; Nava Bashan; Yiftach Gepner; Iris Shai; Assaf Rudich


American Journal of Physiology-endocrinology and Metabolism | 2013

A chromatin immunoprecipitation (ChIP) protocol for use in whole human adipose tissue.

Yulia Haim; Tanya Tarnovscki; Dana Bashari; Assaf Rudich


Molecular metabolism | 2017

ASK1 (MAP3K5) is transcriptionally upregulated by E2F1 in adipose tissue in obesity, molecularly defining a human dys-metabolic obese phenotype

Yulia Haim; Matthias Blüher; Daniel Konrad; Nir Goldstein; Nora Klöting; Ilana Harman-Boehm; Boris Kirshtein; Doron Ginsberg; Tanya Tarnovscki; Yftach Gepner; Iris Shai; Assaf Rudich


Biochimica et Biophysica Acta | 2017

Adipose tissue conditioned media support macrophage lipid-droplet biogenesis by interfering with autophagic flux

Sapir Bechor; Dikla Nachmias; Natalie Elia; Yulia Haim; Maayan Vatarescu; Alicia Leikin-Frenkel; Martin Gericke; Tanya Tarnovscki; Guy Las; Assaf Rudich

Collaboration


Dive into the Tanya Tarnovscki's collaboration.

Top Co-Authors

Avatar

Assaf Rudich

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Nava Bashan

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Boris Kirshtein

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Ilana Harman-Boehm

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Yulia Haim

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Iris Shai

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Julia Kovsan

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Betty Schwartz

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge