Western diet dampens T regulatory cell function to fuel hepatic inflammation in nonalcoholic fatty liver disease
The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial. Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphth...
Saved in:
Published in | bioRxiv |
---|---|
Main Authors | , , , , , , , , |
Format | Journal Article Paper |
Language | English |
Published |
United States
Cold Spring Harbor Laboratory
25.03.2023
|
Edition | 1.1 |
Subjects | |
Online Access | Get full text |
Cover
Loading…
Abstract | The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial.
Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3
mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR.
WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD.
functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells.
Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD.
In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD. |
---|---|
AbstractList | The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial.Background and aimsThe immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial.Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3 DTR mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR.MethodsMice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3 DTR mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR.WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. Ex vivo functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells.ResultsWD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. Ex vivo functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells.Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD.ConclusionsOur findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD.In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD.Lay summaryIn this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD. The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial. Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3DTR mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR. WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. Ex vivo functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells. Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD. In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD. The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial. Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3 mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR. WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells. Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD. In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD. |
Author | Iyer, Smita S Pal, Pabitra B Grakoui, Arash Tedesco, Dana Raeman, Reben Singhi, Aatur D Chaudhary, Sudrishti Rai, Ravi Monga, Satdarshan P |
Author_xml | – sequence: 1 givenname: Sudrishti surname: Chaudhary fullname: Chaudhary, Sudrishti – sequence: 2 givenname: Ravi surname: Rai fullname: Rai, Ravi – sequence: 3 givenname: Pabitra B surname: Pal fullname: Pal, Pabitra B – sequence: 4 givenname: Dana surname: Tedesco fullname: Tedesco, Dana – sequence: 5 givenname: Aatur D surname: Singhi fullname: Singhi, Aatur D – sequence: 6 givenname: Satdarshan P surname: Monga fullname: Monga, Satdarshan P – sequence: 7 givenname: Arash surname: Grakoui fullname: Grakoui, Arash – sequence: 8 givenname: Smita S surname: Iyer fullname: Iyer, Smita S – sequence: 9 givenname: Reben surname: Raeman fullname: Raeman, Reben |
BackLink | https://www.ncbi.nlm.nih.gov/pubmed/36993495$$D View this record in MEDLINE/PubMed |
BookMark | eNpNUMtOwzAQtFARLaUfwAX5yKXFjySOj6jiJVXiUsQxcpwNNXLsECcV_XtcWh6X3ZFmNLsz52jkvAOELilZUEroDSOMLwhfxJlyLoU4QROWSTbPGUlH__AYzUJ4J4QwmVEukjM05pmUPJHpBLWvEHroHK4M9LhSTQsu4DXu4G2wqvfdDmuwFteD073xDvc-YrB4A63qjcbG1VY1jfomjcPxS2W133gbyVr1_Q5bs4UuHgigAlyg01rZALPjnqKX-7v18nG-en54Wt6u5iXNczFPhZJCs1yRkqcJJSBVRRMtOa_zmnFgMUnOapGBJIppmVKms1zTmpclUZXmU3R98C2N7z7Ntmg706huV-x7Kwgv4jz09idtO_8xxD6KxoR9auXAD6FgQjJJUpKwKL06SoeygerX9KdQ_gWdXXny |
ContentType | Journal Article Paper |
Copyright | 2023, Posted by Cold Spring Harbor Laboratory |
Copyright_xml | – notice: 2023, Posted by Cold Spring Harbor Laboratory |
DBID | NPM 7X8 FX. |
DOI | 10.1101/2023.03.23.533977 |
DatabaseName | PubMed MEDLINE - Academic bioRxiv |
DatabaseTitle | PubMed MEDLINE - Academic |
DatabaseTitleList | MEDLINE - Academic PubMed |
Database_xml | – sequence: 1 dbid: FX. name: bioRxiv url: https://www.biorxiv.org/ sourceTypes: Open Access Repository – sequence: 2 dbid: NPM name: PubMed url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed sourceTypes: Index Database |
DeliveryMethod | fulltext_linktorsrc |
Discipline | Biology |
EISSN | 2692-8205 |
Edition | 1.1 |
ExternalDocumentID | 2023.03.23.533977v1 36993495 |
Genre | Preprint |
GroupedDBID | NPM 7X8 8FE 8FH AFKRA ALMA_UNASSIGNED_HOLDINGS BBNVY BENPR BHPHI CCPQU HCIFZ LK8 M7P NQS PHGZM PHGZT PIMPY PQGLB PROAC RHI FX. |
ID | FETCH-LOGICAL-b1887-57a97c28a0b35410e9ad14c933f8f23e213782f76e90a2c9512c68c1f3bb0adc3 |
IEDL.DBID | FX. |
ISSN | 2692-8205 |
IngestDate | Tue Jan 07 18:58:47 EST 2025 Tue Aug 05 10:28:46 EDT 2025 Wed Feb 19 02:24:35 EST 2025 |
IsDoiOpenAccess | true |
IsOpenAccess | true |
IsPeerReviewed | false |
IsScholarly | false |
Keywords | NAFLD Inflammation NASH T regulatory cells Fibrosis |
Language | English |
License | The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. |
LinkModel | DirectLink |
MergedId | FETCHMERGED-LOGICAL-b1887-57a97c28a0b35410e9ad14c933f8f23e213782f76e90a2c9512c68c1f3bb0adc3 |
Notes | ObjectType-Article-2 SourceType-Scholarly Journals-1 ObjectType-Working Paper/Pre-Print-1 ObjectType-Feature-3 content type line 23 Competing Interest Statement: The authors have declared no competing interest. |
OpenAccessLink | https://www.biorxiv.org/content/10.1101/2023.03.23.533977 |
PMID | 36993495 |
PQID | 2792905042 |
PQPubID | 23479 |
PageCount | 35 |
ParticipantIDs | biorxiv_primary_2023_03_23_533977 proquest_miscellaneous_2792905042 pubmed_primary_36993495 |
PublicationCentury | 2000 |
PublicationDate | 2023-Mar-25 20230325 |
PublicationDateYYYYMMDD | 2023-03-25 |
PublicationDate_xml | – month: 03 year: 2023 text: 2023-Mar-25 day: 25 |
PublicationDecade | 2020 |
PublicationPlace | United States |
PublicationPlace_xml | – name: United States |
PublicationTitle | bioRxiv |
PublicationTitleAlternate | bioRxiv |
PublicationYear | 2023 |
Publisher | Cold Spring Harbor Laboratory |
Publisher_xml | – name: Cold Spring Harbor Laboratory |
References | Miyake, Kaise, Isono (2023.03.23.533977v1.30) 2007; 178 Inzaugarat, Ferreyra Solari, Billordo (2023.03.23.533977v1.5) 2011; 31 Rau, Schilling, Meertens (2023.03.23.533977v1.15) 2016; 196 Penaloza-MacMaster, Kamphorst, Wieland (2023.03.23.533977v1.25) 2014; 211 Stepanova, Kabbara, Mohess (2023.03.23.533977v1.3) 2022; 6 Younossi, Stepanova, Ong (2023.03.23.533977v1.2) 2021; 19 Gerriets, Kishton, Johnson (2023.03.23.533977v1.32) 2016; 17 Josefowicz, Lu, Rudensky (2023.03.23.533977v1.10) 2012; 30 Wang, Zhang, Wang (2023.03.23.533977v1.20) 2021; 75 Kim, Rasmussen, Rudensky (2023.03.23.533977v1.23) 2007; 8 Soderberg, Marmur, Eckes (2023.03.23.533977v1.14) 2011; 119 Van Herck, Vonghia, Kwanten (2023.03.23.533977v1.21) 1711; 11 Rau, Rehman, Dittrich (2023.03.23.533977v1.16) 2018; 6 Kamada, Togashi, Tay (2023.03.23.533977v1.31) 2019; 116 Dywicki, Buitrago-Molina, Noyan (2023.03.23.533977v1.19) 2022; 6 Boyman, Kovar, Rubinstein (2023.03.23.533977v1.26) 2006; 311 Ma, Kesarwala, Eggert (2023.03.23.533977v1.27) 2016; 531 Zhang, Zhang (2023.03.23.533977v1.8) 2020; 11 Thorburn, Frankel, Thorburn (2023.03.23.533977v1.29) 2003; 9 Bhattacharjee, Kumar, Arindkar (2023.03.23.533977v1.34) 2014; 25 Anstee, Targher, Day (2023.03.23.533977v1.1) 2013; 10 Her, Tan, Lim (2023.03.23.533977v1.18) 2020; 11 Bennett, Clausen (2023.03.23.533977v1.28) 2007; 28 Rahman, Desai, Iyer (2023.03.23.533977v1.24) 2016 Germain (2023.03.23.533977v1.9) 2002; 2 Ghazarian, Revelo, Nohr (2023.03.23.533977v1.12) 2017; 2 Wang, Li, Fu (2023.03.23.533977v1.11) 2022; 2022 Wei, Long, Yang (2023.03.23.533977v1.33) 2016; 17 Chapman, Boothby, Chi (2023.03.23.533977v1.6) 2020; 20 Seike, Mizukoshi, Yamada (2023.03.23.533977v1.13) 2020; 55 Sakaguchi, Mikami, Wing (2023.03.23.533977v1.7) 2020; 38 Wolf, Adili, Piotrowitz (2023.03.23.533977v1.35) 2014; 26 Barrow, Khan, Fredrickson (2023.03.23.533977v1.17) 2021; 74 Rai, Liu, Iyer (2023.03.23.533977v1.4) 2020 Snook, Kim, Williams (2023.03.23.533977v1.22) 2018; 3 |
References_xml | – volume: 26 start-page: 549 year: 2014 end-page: 64 ident: 2023.03.23.533977v1.35 article-title: Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes publication-title: Cancer Cell – volume: 31 start-page: 1120 year: 2011 end-page: 30 ident: 2023.03.23.533977v1.5 article-title: Altered phenotype and functionality of circulating immune cells characterize adult patients with nonalcoholic steatohepatitis publication-title: J Clin Immunol – volume: 311 start-page: 1924 year: 2006 end-page: 7 ident: 2023.03.23.533977v1.26 article-title: Selective stimulation of T cell subsets with antibody-cytokine immune complexes publication-title: Science – volume: 17 start-page: 1459 year: 2016 end-page: 1466 ident: 2023.03.23.533977v1.32 article-title: Foxp3 and Toll-like receptor signaling balance T(reg) cell anabolic metabolism for suppression publication-title: Nat Immunol – volume: 211 start-page: 1905 year: 2014 end-page: 18 ident: 2023.03.23.533977v1.25 article-title: Interplay between regulatory T cells and PD-1 in modulating T cell exhaustion and viral control during chronic LCMV infection publication-title: J Exp Med – year: 2016 ident: 2023.03.23.533977v1.24 article-title: Loss of Junctional Adhesion Molecule A Promotes Severe Steatohepatitis in Mice on a Diet High in Saturated Fat, Fructose, and Cholesterol publication-title: Gastroenterology – volume: 38 start-page: 541 year: 2020 end-page: 566 ident: 2023.03.23.533977v1.7 article-title: Regulatory T Cells and Human Disease publication-title: Annu Rev Immunol – volume: 11 issue: 580968 year: 2020 ident: 2023.03.23.533977v1.18 article-title: CD4(+) T Cells Mediate the Development of Liver Fibrosis in High Fat Diet-Induced NAFLD in Humanized Mice publication-title: Front Immunol – volume: 11 issue: 2020 year: 1711 ident: 2023.03.23.533977v1.21 article-title: Adoptive Cell Transfer of Regulatory T Cells Exacerbates Hepatic Steatosis in High-Fat High-Fructose Diet-Fed Mice publication-title: Front Immunol – volume: 9 start-page: 861 year: 2003 end-page: 5 ident: 2023.03.23.533977v1.29 article-title: Apoptosis by leukemia cell-targeted diphtheria toxin occurs via receptor-independent activation of Fas-associated death domain protein publication-title: Clin Cancer Res – year: 2020 ident: 2023.03.23.533977v1.4 article-title: Blocking integrin alpha4beta7-mediated CD4 T cell recruitment to the intestine and liver protects mice from western diet-induced non- alcoholic steatohepatitis publication-title: J Hepatol – volume: 55 start-page: 701 year: 2020 end-page: 711 ident: 2023.03.23.533977v1.13 article-title: Fatty acid-driven modifications in T-cell profiles in non-alcoholic fatty liver disease patients publication-title: J Gastroenterol – volume: 531 start-page: 253 year: 2016 end-page: 7 ident: 2023.03.23.533977v1.27 article-title: NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis publication-title: Nature – volume: 2022 start-page: 6250751 year: 2022 ident: 2023.03.23.533977v1.11 article-title: Correlation between T-Lymphocyte Subsets, Regulatory T Cells, and Hepatic Fibrosis in Patients with Nonalcoholic Fatty Liver publication-title: Evid Based Complement Alternat Med – volume: 116 start-page: 9999 year: 2019 end-page: 10008 ident: 2023.03.23.533977v1.31 article-title: PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer publication-title: Proc Natl Acad Sci U S A – volume: 6 start-page: 320 year: 2022 end-page: 333 ident: 2023.03.23.533977v1.19 article-title: The Detrimental Role of Regulatory T Cells in Nonalcoholic Steatohepatitis publication-title: Hepatol Commun – volume: 10 start-page: 330 year: 2013 end-page: 44 ident: 2023.03.23.533977v1.1 article-title: Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis publication-title: Nat Rev Gastroenterol Hepatol – volume: 2 year: 2017 ident: 2023.03.23.533977v1.12 article-title: Type I Interferon Responses Drive Intrahepatic T cells to Promote Metabolic Syndrome publication-title: Sci Immunol – volume: 6 start-page: 1496 year: 2018 end-page: 1507 ident: 2023.03.23.533977v1.16 article-title: Fecal SCFAs and SCFA-producing bacteria in gut microbiome of human NAFLD as a putative link to systemic T-cell activation and advanced disease publication-title: United European Gastroenterol J – volume: 196 start-page: 97 year: 2016 end-page: 105 ident: 2023.03.23.533977v1.15 article-title: Progression from Nonalcoholic Fatty Liver to Nonalcoholic Steatohepatitis Is Marked by a Higher Frequency of Th17 Cells in the Liver and an Increased Th17/Resting Regulatory T Cell Ratio in Peripheral Blood and in the Liver publication-title: J Immunol – volume: 19 start-page: 580 year: 2021 end-page: 589 ident: 2023.03.23.533977v1.2 article-title: Nonalcoholic Steatohepatitis Is the Most Rapidly Increasing Indication for Liver Transplantation in the United States publication-title: Clin Gastroenterol Hepatol – volume: 119 start-page: 412 year: 2011 end-page: 20 ident: 2023.03.23.533977v1.14 article-title: Microvesicular fat, inter cellular adhesion molecule-1 and regulatory T-lymphocytes are of importance for the inflammatory process in livers with non-alcoholic steatohepatitis publication-title: APMIS – volume: 8 start-page: 191 year: 2007 end-page: 7 ident: 2023.03.23.533977v1.23 article-title: Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice publication-title: Nat Immunol – volume: 178 start-page: 5001 year: 2007 end-page: 9 ident: 2023.03.23.533977v1.30 article-title: Protective role of macrophages in noninflammatory lung injury caused by selective ablation of alveolar epithelial type II Cells publication-title: J Immunol – volume: 20 start-page: 55 year: 2020 end-page: 70 ident: 2023.03.23.533977v1.6 article-title: Metabolic coordination of T cell quiescence and activation publication-title: Nat Rev Immunol – volume: 3 year: 2018 ident: 2023.03.23.533977v1.22 article-title: TCR signal strength controls the differentiation of CD4(+) effector and memory T cells publication-title: Sci Immunol – volume: 2 start-page: 309 year: 2002 end-page: 22 ident: 2023.03.23.533977v1.9 article-title: T-cell development and the CD4-CD8 lineage decision publication-title: Nat Rev Immunol – volume: 6 start-page: 1506 year: 2022 end-page: 1515 ident: 2023.03.23.533977v1.3 article-title: Nonalcoholic steatohepatitis is the most common indication for liver transplantation among the elderly: Data from the United States Scientific Registry of Transplant Recipients publication-title: Hepatol Commun – volume: 75 start-page: 1271 year: 2021 end-page: 1283 ident: 2023.03.23.533977v1.20 article-title: Regulatory T-cell and neutrophil extracellular trap interaction contributes to carcinogenesis in non-alcoholic steatohepatitis publication-title: J Hepatol – volume: 30 start-page: 531 year: 2012 end-page: 64 ident: 2023.03.23.533977v1.10 article-title: Regulatory T cells: mechanisms of differentiation and function publication-title: Annu Rev Immunol – volume: 28 start-page: 525 year: 2007 end-page: 31 ident: 2023.03.23.533977v1.28 article-title: DC ablation in mice: promises, pitfalls, and challenges publication-title: Trends Immunol – volume: 17 start-page: 277 year: 2016 end-page: 85 ident: 2023.03.23.533977v1.33 article-title: Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis publication-title: Nat Immunol – volume: 11 start-page: 1142 year: 2020 ident: 2023.03.23.533977v1.8 article-title: T Cells in Fibrosis and Fibrotic Diseases publication-title: Front Immunol – volume: 74 start-page: 704 year: 2021 end-page: 722 ident: 2023.03.23.533977v1.17 article-title: Microbiota-Driven Activation of Intrahepatic B Cells Aggravates NASH Through Innate and Adaptive Signaling publication-title: Hepatology – volume: 25 start-page: 219 year: 2014 end-page: 26 ident: 2023.03.23.533977v1.34 article-title: Role of immunodeficient animal models in the development of fructose induced NAFLD publication-title: J Nutr Biochem |
SSID | ssj0002961374 |
Score | 1.8292078 |
SecondaryResourceType | preprint |
Snippet | The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver... |
SourceID | biorxiv proquest pubmed |
SourceType | Open Access Repository Aggregation Database Index Database |
SubjectTerms | Pathology |
Title | Western diet dampens T regulatory cell function to fuel hepatic inflammation in nonalcoholic fatty liver disease |
URI | https://www.ncbi.nlm.nih.gov/pubmed/36993495 https://www.proquest.com/docview/2792905042 https://www.biorxiv.org/content/10.1101/2023.03.23.533977 |
hasFullText | 1 |
inHoldings | 1 |
isFullTextHit | |
isPrint | |
link | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1LS8NAEF60RfDm2_ooK3iNJLub11VpKYKlSIu9hX1ioSQlTcX-e2eSWDwoeFny3IFvJzvf7kxmCLkX2qE3SXnAXZ0nnHaeDJj2bCR9K3mUSFVHW4yj0Uw8z8P5j1JfGFapFkX5ufio_fgYsA2zb_Nx-wGu1TlmJYUWiAqQl33SBZUSWLVhOH_Yba-wFOxULFo_5q9vAuNtJf3NLmsrMzwi3Ylc2fKY7Nn8hBw0ZSK3p2T11qQzoGZhK2okMN18Tae0bOrIF-WW4v47RRuFONOqgGO7pO8W46U1BS2CgW9-UoQTmiP9rkvjwk0nq2pLlxifQVt3zRmZDQfTp5HXVkrwVICzRBjLNNYskb7ioQh8m0oTCJ1y7hLHuGWABYxFHNnUl0wDq2I6SnTguFK-NJqfkw6ItpeEhtzYyMnYAjcTXJnURGDBUujXamcM65G7FrVs1eTDyBDZzOcZtA2y8Mw3nhloK0Igc1ts1hmmK0z9EGaKHrlogN51wyPgSrBeu_qHhGtyiNcwGIyFN6RTlRt7C-ygUn3SfRyMJ6_9Wh-gHU9evgBvTbh7 |
linkProvider | Cold Spring Harbor Laboratory Press |
linkToHtml | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1LS8NAEF60RfTm2_pcwWtKsptskrNYqtbiocXewj6xIElJU7H_3pkk9qTgZUnYZAe-3ex8OzOZIeQu1A69ScoD7uq80GnnyYBpzwrpW8lFIlUdbTEWw2n4NItmrcFt2YZVqnlRfs0_az8-BmzD7tt83H6AZ3WOWUmhBaIC5KWPZupt0sVEZxjSNZj1NzYWloKyisPWmfnr60B7W3F_U8xa1Qz2SfdVLmx5QLZsfkh2mlqR6yOyeGtyGlAztxU1EuhuvqQTWjbF5ItyTdEIT1FRIdi0KuDaftB3i0HTmsJSgtlv_lSEG5ojB6_r40Knk1W1ph8YpEFbn80xmQ4eJvdDry2X4KkAt4oolmmsWSJ9xaMw8G0qTRDqlHOXOMYtAyxgQmJhU18yDdSKaZHowHGlfGk0PyEdEG3PCI24scLJ2AJBC7kyqRGgxlIY12pnDOuR2xa1bNEkxcgQ2cznGbQNsvDMD54ZLFmEQOa2WC0zzFmY-hFsFz1y2gC9GYYLIExwaDv_h4QbsjucvIyy0eP4-YLsYT9Gh7HoknSqcmWvgC5U6rpeE9_Kqbqs |
linkToPdf | http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwjV1LS8NAEF60RfHm2_pcwWtKspvnWQ31Qemhxd7CPrFQmpCmYv-9M0ksHhS8LAkbduHbyc63M7MzhNz5yqI3STrAXa3jW2Ud4THlmFC4RvAwFrKOthiGg4n_PA2mP-7CYFilnOXl5-yj9uNjwDbsvs3P7Xp4VueYlRRaICpAXvpopu4X2m6TLiY7Q8lOp_2NnYUloLAiv3Vo_joEUN92yr9pZq1u0n3SHYnClAdkyywOyU5TL3J9RIq3Jq8B1TNTUS2A8i6WdEzLpqB8Xq4pGuIpKisEnFY5PJs5fTcYOK0oiBNIQHNbEV7oAnl4XSMXOq2oqjWdY6AGbf02x2SSPo7vB05bMsGRHm4XQSSSSLFYuJIHvueaRGjPVwnnNraMGwZYwKJEoUlcwRTQK6bCWHmWS-kKrfgJ6cDU5ozQgGsTWhEZIGk-lzrRIaiyBMY1ymrNeuS2RS0rmsQYGSKbuTyDtkEWvvnGMwOxRQjEwuSrZYZ5CxM3gC2jR04boDfD8BBIExzczv8xww3ZHT2k2evT8OWC7GE3Boix4JJ0qnJlroAxVPK6FokvmJi7vQ |
openUrl | ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Western+diet+dampens+T+regulatory+cell+function+to+fuel+hepatic+inflammation+in+nonalcoholic+fatty+liver+disease&rft.jtitle=bioRxiv&rft.au=Chaudhary%2C+Sudrishti&rft.au=Rai%2C+Ravi&rft.au=Pal%2C+Pabitra+B&rft.au=Tedesco%2C+Dana&rft.date=2023-03-25&rft.issn=2692-8205&rft.eissn=2692-8205&rft_id=info:doi/10.1101%2F2023.03.23.533977&rft.externalDBID=NO_FULL_TEXT |
thumbnail_l | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=2692-8205&client=summon |
thumbnail_m | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=2692-8205&client=summon |
thumbnail_s | http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=2692-8205&client=summon |