Network


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

Hotspot


Dive into the research topics where Hagit Shapiro is active.

Publication


Featured researches published by Hagit Shapiro.


Nature | 2014

Artificial sweeteners induce glucose intolerance by altering the gut microbiota

Jotham Suez; Tal Korem; David Zeevi; Gili Zilberman-Schapira; Christoph A. Thaiss; Ori Maza; David Israeli; Niv Zmora; Shlomit Gilad; Adina Weinberger; Yael Kuperman; Alon Harmelin; Ilana Kolodkin-Gal; Hagit Shapiro; Zamir Halpern; Eran Segal; Eran Elinav

Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.


Cell | 2016

Microbiota Diurnal Rhythmicity Programs Host Transcriptome Oscillations

Christoph A. Thaiss; Maayan Levy; Tal Korem; Lenka Dohnalová; Hagit Shapiro; Diego Jaitin; Eyal David; Deborah R. Winter; Meital Gury-BenAri; Evgeny Tatirovsky; Timur Tuganbaev; Sara Federici; Niv Zmora; David Zeevi; Mally Dori-Bachash; Meirav Pevsner-Fischer; Elena Kartvelishvily; Alexander Brandis; Alon Harmelin; Oren Shibolet; Zamir Halpern; Kenya Honda; Ido Amit; Eran Segal; Eran Elinav

The intestinal microbiota undergoes diurnal compositional and functional oscillations that affect metabolic homeostasis, but the mechanisms by which the rhythmic microbiota influences host circadian activity remain elusive. Using integrated multi-omics and imaging approaches, we demonstrate that the gut microbiota features oscillating biogeographical localization and metabolome patterns that determine the rhythmic exposure of the intestinal epithelium to different bacterial species and their metabolites over the course of a day. This diurnal microbial behavior drives, in turn, the global programming of the host circadian transcriptional, epigenetic, and metabolite oscillations. Surprisingly, disruption of homeostatic microbiome rhythmicity not only abrogates normal chromatin and transcriptional oscillations of the host, but also incites genome-wide de novo oscillations in both intestine and liver, thereby impacting diurnal fluctuations of host physiology and disease susceptibility. As such, the rhythmic biogeography and metabolome of the intestinal microbiota regulates the temporal organization and functional outcome of host transcriptional and epigenetic programs.


Nature | 2016

Persistent microbiome alterations modulate the rate of post-dieting weight regain

Christoph A. Thaiss; Shlomik Itav; Daphna Rothschild; Mariska T. Meijer; Maayan Levy; Claudia Moresi; Lenka Dohnalová; Sofia Braverman; Shachar Rozin; Sergey Malitsky; Mally Dori-Bachash; Yael Kuperman; Inbal E. Biton; Arieh Gertler; Alon Harmelin; Hagit Shapiro; Zamir Halpern; Asaph Aharoni; Eran Segal; Eran Elinav

In tackling the obesity pandemic, considerable efforts are devoted to the development of effective weight reduction strategies, yet many dieting individuals fail to maintain a long-term weight reduction, and instead undergo excessive weight regain cycles. The mechanisms driving recurrent post-dieting obesity remain largely elusive. Here we identify an intestinal microbiome signature that persists after successful dieting of obese mice and contributes to faster weight regain and metabolic aberrations upon re-exposure to obesity-promoting conditions. Faecal transfer experiments show that the accelerated weight regain phenotype can be transmitted to germ-free mice. We develop a machine-learning algorithm that enables personalized microbiome-based prediction of the extent of post-dieting weight regain. Additionally, we find that the microbiome contributes to diminished post-dieting flavonoid levels and reduced energy expenditure, and demonstrate that flavonoid-based ‘post-biotic’ intervention ameliorates excessive secondary weight gain. Together, our data highlight a possible microbiome contribution to accelerated post-dieting weight regain, and suggest that microbiome-targeting approaches may help to diagnose and treat this common disorder.


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.


The Journal of Clinical Endocrinology and Metabolism | 2013

Adipose Tissue Foam Cells Are Present in Human Obesity

Hagit Shapiro; Tal Pecht; Ruthy Shaco-Levy; Ilana Harman-Boehm; Boris Kirshtein; Yael Kuperman; Alon Chen; Matthias Blüher; Iris Shai; Assaf Rudich

CONTEXT Adipose tissue macrophages (ATMs) are thought to engulf the remains of dead adipocytes in obesity, potentially resulting in increased intracellular neutral lipid content. Lipid-laden macrophages (foam cells [FCs]) have been described in atherosclerotic lesions and have been proposed to contribute to vascular pathophysiology, which is enhanced in obesity. OBJECTIVE The objective of this study was to determine whether a subclass of lipid-laden ATMs (adipose FCs) develop in obesity and to assess whether they may uniquely contribute to obesity-associated morbidity. SETTING AND PATIENTS Patients undergoing elective abdominal surgery from the Beer-Sheva (N = 94) and the Leipzig (N = 40) complementary cohorts were recruited. Paired abdominal subcutaneous (SC) and omental (Om) fat biopsy samples were collected and analyzed by histological and flow cytometry-based methods. Functional studies in mice included coculture of ATMs or FCs with adipose tissue. RESULTS ATM lipid content was increased 3-fold in Om compared with SC fat, particularly in obese persons. FCs could be identified in some patients and were most abundant in Om fat of obese persons, particularly those with intra-abdominal fat distribution. Stepwise multivariate models demonstrated depot differential associations: fasting glucose with SC FCs (β = 0.667, P < .001) and fasting insulin (β = 0.413, P = .006) and total ATM count (β = 0.310, P = .034) with Om FCs in models including age, body mass index, high-density lipoprotein cholesterol, and high-sensitivity C-reactive protein. When cocultured with adipose explants from lean mice, FCs induced attenuated insulin responsiveness compared with adipose explants cocultured with control ATMs with low lipid content. CONCLUSIONS FCs can be identified as an ATM subclass in human SC and Om adipose tissues in 2 independent cohorts, with distinct depot-related associations with clinical parameters. Once formed, they may engage in local cross-talk with adipocytes, contributing to adipose insulin resistance.


Current Opinion in Immunology | 2014

The cross talk between microbiota and the immune system: metabolites take center stage

Hagit Shapiro; Christoph A. Thaiss; Maayan Levy; Eran Elinav

The human meta-organism consists of more than 90% of microbial cells. The gastrointestinal tract harbors trillions of commensal microorganisms that influence the development and homeostasis of the host. Alterations in composition and function of the microbiota, termed dysbiosis, have been implicated in a multitude of metabolic and inflammatory diseases in humans. Thus, understanding the molecular underpinnings the cross talk between commensal bacteria and their host during homeostasis and dysbiosis may hold the key to understanding many idiopathic diseases. While most attention has focused on the innate recognition of immune-stimulatory bacterial molecules, such as cell wall components and nucleic acids, we emphasize here the impact of diet-dependent microbial metabolites on the development and function of the immune system.


Molecular metabolism | 2016

Non-alcoholic fatty liver and the gut microbiota.

Stavros Bashiardes; Hagit Shapiro; Shachar Rozin; Oren Shibolet; Eran Elinav

Background Non-alcoholic fatty liver (NAFLD) is a common, multi-factorial, and poorly understood liver disease whose incidence is globally rising. NAFLD is generally asymptomatic and associated with other manifestations of the metabolic syndrome. Yet, up to 25% of NAFLD patients develop a progressive inflammatory liver disease termed non-alcoholic steatohepatitis (NASH) that may progress towards cirrhosis, hepatocellular carcinoma, and the need for liver transplantation. In recent years, several lines of evidence suggest that the gut microbiome represents a significant environmental factor contributing to NAFLD development and its progression into NASH. Suggested microbiome-associated mechanisms contributing to NAFLD and NASH include dysbiosis-induced deregulation of the gut endothelial barrier function, which facilitates systemic bacterial translocation, and intestinal and hepatic inflammation. Furthermore, increased microbiome-modulated metabolites such as lipopolysaccharides, short chain fatty acids (SCFAs), bile acids, and ethanol, may affect liver pathology through multiple direct and indirect mechanisms. Scope of review Herein, we discuss the associations, mechanisms, and clinical implications of the microbiomes contribution to NAFLD and NASH. Understanding these contributions to the development of fatty liver pathogenesis and its clinical course may serve as a basis for development of therapeutic microbiome-targeting approaches for treatment and prevention of NAFLD and NASH. Major conclusions Intestinal host–microbiome interactions play diverse roles in the pathogenesis and progression of NAFLD and NASH. Elucidation of the mechanisms driving these microbial effects on the pathogenesis of NAFLD and NASH may enable to identify new diagnostic and therapeutic targets of these common metabolic liver diseases. This article is part of a special issue on microbiota.


Science | 2018

Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection

Christoph A. Thaiss; Maayan Levy; Inna Grosheva; Danping Zheng; Eliran Soffer; Eran Blacher; Sofia Braverman; Anouk C. Tengeler; Oren Barak; Maya Elazar; Rotem Ben-Zeev; Dana Lehavi-Regev; Meirav N. Katz; Meirav Pevsner-Fischer; Arieh Gertler; Zamir Halpern; Alon Harmelin; Suhail Aamar; Patricia Serradas; Alexandra Grosfeld; Hagit Shapiro; Benjamin Geiger; Eran Elinav

Metabolic syndrome, leaky guts, and infection Metabolic syndrome often accompanies obesity and hyperglycemia and is associated with a breakdown in the integrity of the intestinal barrier and increased risk of systemic infection. Thaiss et al. found that mice with systemic infection of a Salmonella analog, Citrobacter rodentium, also exhibited hyperglycemia. Deletion of the glucose transporter GLUT2 altered sensitivity to chemically induced epithelial permeability and protected mice from pathogen invasion. The authors also found a correlation in humans between glycated hemoglobin (an indicator of hyperglycemia) and serum levels of pathogen recognition receptor ligands. Science, this issue p. 1376 High blood sugar levels cause epithelial reprogramming, compromising gut barrier integrity and increasing susceptibility to pathogens. Obesity, diabetes, and related manifestations are associated with an enhanced, but poorly understood, risk for mucosal infection and systemic inflammation. Here, we show in mouse models of obesity and diabetes that hyperglycemia drives intestinal barrier permeability, through GLUT2-dependent transcriptional reprogramming of intestinal epithelial cells and alteration of tight and adherence junction integrity. Consequently, hyperglycemia-mediated barrier disruption leads to systemic influx of microbial products and enhanced dissemination of enteric infection. Treatment of hyperglycemia, intestinal epithelial–specific GLUT2 deletion, or inhibition of glucose metabolism restores barrier function and bacterial containment. In humans, systemic influx of intestinal microbiome products correlates with individualized glycemic control, indicated by glycated hemoglobin levels. Together, our results mechanistically link hyperglycemia and intestinal barrier function with systemic infectious and inflammatory consequences of obesity and diabetes.


Journal of Diabetes | 2017

Personalized microbiome‐based approaches to metabolic syndrome management and prevention

Hagit Shapiro; Jotham Suez; Eran Elinav

Personalized or precision medicine is a novel clinical approach targeted to the individual patient and based on integration of clinical, genetic, and environmental factors that define a patient uniquely from other individuals featuring similar clinical symptoms. Such a personalized medicine approach is increasingly applied for diagnosis, clinical stratification, and treatment of metabolic syndrome (MetS)‐associated risks and diseases, including obesity, type 2 diabetes, non‐alcoholic fatty liver disease, and their complications. One emerging factor that governs MetS manifestations is the microbiome, the composition, function, growth dynamics, associated metabolite profile and diverse effects of which on host immune and metabolic systems can all significantly affect metabolic homeostasis. Interindividual differences in microbiome composition and function, as well as personal variations in microbial‐derived products, pave the way towards microbiome‐based personalized medicine in treating MetS‐related diseases.


Trends in Immunology | 2017

NLRP6: A Multifaceted Innate Immune Sensor

Maayan Levy; Hagit Shapiro; Christoph A. Thaiss; Eran Elinav

NLRP6, a member of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) innate immune receptor family, regulates inflammation and host defense against microorganisms. Similar to other NLRs, NLRP6 not only participates in inflammasome formation, but is also involved in nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling regulation and facilitation of gastrointestinal antiviral effector functions. Additionally, NLRP6 contributes to the regulation of mucus secretion and antimicrobial peptide production, thereby impacting intestinal microbial colonization and associated microbiome-related infectious, autoinflammatory, metabolic, and neoplastic diseases. However, several of the mechanisms attributed to the functions of NLRP6 remain debatable, leaving open questions as to the relevant molecular mechanisms and interacting partners, and putative human relevance. We herein discuss recent findings related to NLRP6 activity, while highlighting outstanding questions and future perspectives in elucidating its roles in health and disease.

Collaboration


Dive into the Hagit Shapiro's collaboration.

Top Co-Authors

Avatar

Eran Elinav

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Christoph A. Thaiss

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Alon Harmelin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Zamir Halpern

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eran Segal

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Maayan Levy

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Jotham Suez

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

David Zeevi

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge