1
|
Myung J, Vitet H, Truong VH, Ananthasubramaniam B. The role of the multiplicity of circadian clocks in mammalian systems. Sleep Med 2025; 131:106518. [PMID: 40222295 DOI: 10.1016/j.sleep.2025.106518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/22/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Circadian clocks regulate rhythmic biological processes in nearly every tissue, aligning physiology and behavior with the 24-h light-dark cycle. While the central circadian clock in the suprachiasmatic nucleus (SCN) has been extensively studied, emerging evidence indicates that virtually every cell in the body possesses its own locally autonomous circadian clock. This raises a fundamental question: why do multicellular organisms utilize multiple circadian clocks instead of a single master clock broadcasting time cues? Here, we examine how distributed local clocks differ from phase-resettable cycles and ensure robust temporal scheduling of physiological processes. We discuss how internal entrainment among local clocks governs self-sustained, yet flexible, circadian organization of tissue-specific responses to environmental changes. We also examine how the organization of clocks contributes to seasonal homeostasis, and the implications for disease when coordination among these clocks is disrupted.
Collapse
Affiliation(s)
- Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Hélène Vitet
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | - Vuong Hung Truong
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | | |
Collapse
|
2
|
Li X, Rao Z, Hu W, Lu W, Luo Y. Treating metabolic dysfunction-associated steatohepatitis: The fat-trimming FGF21 approach. Obes Rev 2025; 26:e13861. [PMID: 39546893 DOI: 10.1111/obr.13861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/10/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a condition characterized by hepatosteatosis, inflammation, and tissue damage, with steatosis as the initial stage, which involves chronic, excess deposition of lipids in hepatic lipid droplets. Despite the growing prevalence and serious risks it poses, including liver decompensation, the need for transplantation, and increased patient mortality, MASH currently faces no approved pharmacotherapy. Several promising treatment candidates have emerged from recent clinical trials, including analogs of FGF21 and agonists of the associated FGFR1-KLB complex. These agents were well-tolerated in trials and have demonstrated significant improvements in both histological and biochemical markers of liver fat content, inflammation, injury, and fibrosis in patients with MASH. Endocrine FGF21 plays a vital role in maintaining homeostasis of lipid, glucose, and energy metabolism. It achieves this through pathways that target lipids or lipid droplets in adipocytes and hepatocytes. Mechanistically, pharmacological FGF21 acts as a potent catabolic factor to promote lipid or lipid droplet lipolysis, fatty acid oxidation, mitochondrial catabolic flux, and heat-dissipating energy expenditure, leading to effective clearance of hepatic and systemic gluco-lipotoxicity and inflammatory stress, thereby preventing obesity, diabetes, and MASH pathologies. In this review, we aim to provide an update on the outcomes of clinical trials for several FGF21 mimetics. We compare these outcomes with preclinical studies and offer a lipid-centric perspective on the mechanisms underlying the clinical benefits of these agents for MASH.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
| | - Zhiheng Rao
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
| | - Wenhao Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqin Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas, USA
| | - Yongde Luo
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Feng JN, Shao W, Yang L, Pang J, Ling W, Liu D, Wheeler MB, He HH, Jin T. Hepatic fibroblast growth factor 21 is required for curcumin or resveratrol in exerting their metabolic beneficial effect in male mice. Nutr Diabetes 2025; 15:4. [PMID: 39929809 PMCID: PMC11811165 DOI: 10.1038/s41387-025-00363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Our mechanistic understanding on metabolic beneficial effects of dietary polyphenols has been hampered for decades due to the lack of functional receptors for those compounds and their extremely low plasma concentrations. Recent studies by our team and others suggest that those dietary polyphenols target gut microbiome, and gut-liver axis and that hepatic fibroblast factor 21 (FGF21) serves as a common target for various dietary interventions. METHODS Utilizing liver-specific FGF21 null mice (lFgf21-/-), we are asking a straightforward question: Is hepatic FGF21 required for curcumin or resveratrol, two typical dietary polyphenols, in exerting their metabolic beneficial effect in obesogenic diet-induced obesity mouse models. RESULTS On low-fat diet feeding, no appreciable defect on glucose disposal was observed in male or female lFgf21-/- mice, while fat tolerance was moderately impaired in male but not in female lFgf21-/- mice, associated with elevated random and fasting serum triglyceride (TG) levels, and reduced hepatic expression of Ehhadh and Ppargc1a, which encodes the two downstream effectors of FGF21. On high-fat-high-fructose (HFHF) diet challenge, Fgf21fl/fl but not lFgf21-/- mice exhibited response to curcumin intervention on reducing fasting serum TG, and on improving fat tolerance. Resveratrol intervention also affected FGF21 expression or its downstream effectors. Metabolic beneficial effects of resveratrol intervention observed in HFHF diet-challenged Fgf21fl/fl mice were either absent or attenuated in lFgf21-/- mice. CONCLUSION AND SIGNIFICANCE We conclude that hepatic FGF21 is required for curcumin or resveratrol in exerting their major metabolic beneficial effect. The recognition that FGF21 as the common target of dietary intervention, demonstrated in current as well as previous investigations, brings us a novel angle in understanding metabolic disease treatment and prevention. It remains to be further explored how various dietary interventions regulate FGF21 expression and function, via certain common or unique gut-liver or gut-brain-liver axis.
Collapse
Affiliation(s)
- Jia Nuo Feng
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Weijuan Shao
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lin Yang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Juan Pang
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, PR China
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, PR China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, PR China
| | - Dinghui Liu
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Michael B Wheeler
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Ghosh S, Ganguly A, Habib M, Shin BC, Thamotharan S, Andersson S, Devaskar SU. Hepatic and Pancreatic Cellular Response to Early Life Nutritional Mismatch. Endocrinology 2025; 166:bqaf007. [PMID: 39823439 PMCID: PMC11815087 DOI: 10.1210/endocr/bqaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/22/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
To determine the basis for perinatal nutritional mismatch causing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus, we examined adult phenotype, hepatic transcriptome, and pancreatic β-islet function. In prenatal caloric-restricted rats with intrauterine growth restriction (IUGR) and postnatal exposure to high fat with fructose (HFhf) or high carbohydrate, we investigated male and female IUGR-HFhf and IUGR-high carbohydrate, vs HFhf and control offspring. Males more than females displayed adiposity, glucose intolerance, insulin resistance, hyperlipidemia, and hepatomegaly with hepatic steatosis. Male hepatic triglyceride synthesis, de novo lipogenesis genes increased, while female lipolysis, β-oxidation, fatty acid efflux, and FGF21 genes increased. IUGR-HFhf males demonstrated reduced β-islet insulin and humanin, and type 1 diabetes mellitus human amniotic fluid increased humanin. Humanin suppression disabled glucose stimulated insulin, ATP production, with apoptotic diminished β-islet viability. Humanin and FGF21 may reverse perinatal nutritional mismatched phenotype by restoring functional β islets and preventing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus.
Collapse
Affiliation(s)
- Shubhamoy Ghosh
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Amit Ganguly
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Manal Habib
- Division of Endocrinology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Bo-Chul Shin
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Shanthie Thamotharan
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Sture Andersson
- Department of Pediatrics, Helsinki University Central Hospital, 00290 Helsinki, Finland
| | - Sherin U Devaskar
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| |
Collapse
|
5
|
Nascimento Júnior JXD, Gomes JDC, Imbroisi Filho R, Valença HDM, Branco JR, Araújo AB, Moreira ADOE, Crepaldi LD, Paixão LP, Ochioni AC, Demaria TM, Leandro JGB, Casanova LM, Sola-Penna M, Zancan P. Dietary caloric input and tumor growth accelerate senescence and modulate liver and adipose tissue crosstalk. Commun Biol 2025; 8:18. [PMID: 39775048 PMCID: PMC11707351 DOI: 10.1038/s42003-025-07451-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Metabolic alterations are related to tumorigenesis and other age-related diseases that are accelerated by "Westernized" diets. In fact, hypercaloric nutrition is associated with an increased incidence of cancers and faster aging. Conversely, lifespan-extending strategies, such as caloric restriction, impose beneficial effects on both processes. Here, we investigated the metabolic consequences of hypercaloric-induced aging on tumor growth in female mice. Our findings indicate that a high-fat high-sucrose diet increases tumor growth mainly due to the boosted oxidation of glucose and fatty acids. Consequently, through an increased expression of lactate, IGFBP3, and PTHLH, tumors modulate liver and white adipose tissue metabolism. In the liver, the induced tumor increases fibrosis and accelerates the senescence process, despite the lower systemic pro-inflammatory state. Importantly, the induced tumor induces the wasting and browning of white adipose tissue, thereby reversing diet-induced insulin resistance. Finally, we suggest that tumor growth alters liver-adipose tissue crosstalk that upregulates Fgf21, induces senescence, and negatively modulates lipids and carbohydrates metabolism even in caloric-restricted-fed mice.
Collapse
Affiliation(s)
- José Xavier do Nascimento Júnior
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Júlia da Conceição Gomes
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Imbroisi Filho
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helber de Maia Valença
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jéssica Ristow Branco
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Bandeira Araújo
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda de Oliveira Esteves Moreira
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Letícia Diniz Crepaldi
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Pereira Paixão
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alan C Ochioni
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thainá M Demaria
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Gabriel Bernardo Leandro
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Livia Marques Casanova
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro Sola-Penna
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Zancan
- The MetaboliZSm GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Chhabra Y, Bielefeldt-Ohmann H, Brooks TL, Brooks AJ, Waters MJ. Roles of Growth Hormone-Dependent JAK-STAT5 and Lyn Kinase Signaling in Determining Lifespan and Cancer Incidence. Endocrinology 2024; 165:bqae136. [PMID: 39378329 PMCID: PMC11500606 DOI: 10.1210/endocr/bqae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
In rodents, loss of growth hormone (GH) or its receptor is associated with extended lifespan. We aimed to determine the signaling process resulting in this longevity using GH receptor (GHR)-mutant mice with key signaling pathways deleted and correlate this with cancer incidence and expression of genes associated with longevity. GHR uses both canonical janus kinase (JAK)2-signal transducer and activator of transcription (STAT) signaling as well as signaling via the LYN-ERK1/2 pathway. We used C57BL/6 mice with loss of key receptor tyrosines and truncation resulting in 1) loss of most STAT5 response to GH; 2) total inability to generate STAT5 to GH; 3) loss of Box1 to prevent activation of JAK2 but not LYN kinase; or 4) total knockout of the receptor. For each mutant we analyzed lifespan, histopathology to determine likely cause of death, and hepatic gene and protein expression. The extended lifespan is evident in the Box1-mutant males (retains Lyn activation), which have a median lifespan of 1016 days compared to 890 days for the Ghr-/- males. In the females, GhrBox1-/- mice have a median lifespan of 970 days compared to 911 days for the knockout females. Sexually dimorphic GHR-STAT5 is repressive for longevity, since its removal results in a median lifespan of 1003 days in females compared to 734 days for wild-type females. Numerous transcripts related to insulin sensitivity, oxidative stress response, and mitochondrial function are regulated by GHR-STAT5; however, LYN-responsive genes involve DNA repair, cell cycle control, and anti-inflammatory response. There appears to be a yin-yang relationship between JAK2 and LYN that determines lifespan.
Collapse
Affiliation(s)
- Yash Chhabra
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Helle Bielefeldt-Ohmann
- School of Chemistry & Molecular Biosciences, University of Queensland, St Lucia 4069, Australia
| | - Tania Louise Brooks
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
| | - Andrew James Brooks
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Michael J Waters
- Institute for Molecular Bioscience, University of Queensland, St Lucia 4069, Australia
| |
Collapse
|
7
|
Pacheco-Bernal I, Becerril-Pérez F, Bustamante-Zepeda M, González-Suárez M, Olmedo-Suárez MA, Hernández-Barrientos LR, Alarcón-Del-Carmen A, Escalante-Covarrubias Q, Mendoza-Viveros L, Hernández-Lemus E, León-Del-Río A, de la Rosa-Velázquez IA, Orozco-Solis R, Aguilar-Arnal L. Transitions in chromatin conformation shaped by fatty acids and the circadian clock underlie hepatic transcriptional reorganization in obese mice. Cell Mol Life Sci 2024; 81:309. [PMID: 39060446 PMCID: PMC11335233 DOI: 10.1007/s00018-024-05364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
The circadian clock system coordinates metabolic, physiological, and behavioral functions across a 24-h cycle, crucial for adapting to environmental changes. Disruptions in circadian rhythms contribute to major metabolic pathologies like obesity and Type 2 diabetes. Understanding the regulatory mechanisms governing circadian control is vital for identifying therapeutic targets. It is well characterized that chromatin remodeling and 3D structure at genome regulatory elements contributes to circadian transcriptional cycles; yet the impact of rhythmic chromatin topology in metabolic disease is largely unexplored. In this study, we explore how the spatial configuration of the genome adapts to diet, rewiring circadian transcription and contributing to dysfunctional metabolism. We describe daily fluctuations in chromatin contacts between distal regulatory elements of metabolic control genes in livers from lean and obese mice and identify specific lipid-responsive regions recruiting the clock molecular machinery. Interestingly, under high-fat feeding, a distinct interactome for the clock-controlled gene Dbp strategically promotes the expression of distal metabolic genes including Fgf21. Alongside, new chromatin loops between regulatory elements from genes involved in lipid metabolism control contribute to their transcriptional activation. These enhancers are responsive to lipids through CEBPβ, counteracting the circadian repressor REVERBa. Our findings highlight the intricate coupling of circadian gene expression to a dynamic nuclear environment under high-fat feeding, supporting a temporally regulated program of gene expression and transcriptional adaptation to diet.
Collapse
Affiliation(s)
- Ignacio Pacheco-Bernal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Fernando Becerril-Pérez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Marcia Bustamante-Zepeda
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Mirna González-Suárez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Miguel A Olmedo-Suárez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis Ricardo Hernández-Barrientos
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alejandro Alarcón-Del-Carmen
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Quetzalcoatl Escalante-Covarrubias
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucía Mendoza-Viveros
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Cronobiología, Metabolismo y Envejecimiento, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), Mexico City, México
- Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, Mexico
| | - Enrique Hernández-Lemus
- Department of Computational Genomics, Centro de Ciencias de La Complejidad (C3), Instituto Nacional de Medicina Genómica (INMEGEN), Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfonso León-Del-Río
- Departamento de Medicina Genómica y Toxicología Ambiental, Programa Institucional de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Inti A de la Rosa-Velázquez
- Genomics Laboratory, Red de Apoyo a la Investigación-CIC, Universidad Nacional Autónoma de México, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080, Mexico City, Mexico
- Next Generation Sequencing Core Facility, Helmholtz Zentrum Muenchen, Ingolstaedter Landstr 1, 85754, Neuherberg, Germany
| | - Ricardo Orozco-Solis
- Laboratorio de Cronobiología, Metabolismo y Envejecimiento, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), Mexico City, México
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
8
|
Zaman K, Mun HC, Solon-Biet SM, Senior AM, Raubenheimer D, Simpson SJ, Conigrave AD. Mice Regulate Dietary Amino Acid Balance and Energy Intake by Selecting between Complementary Protein Sources. J Nutr 2024; 154:1766-1780. [PMID: 38583524 DOI: 10.1016/j.tjnut.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND A balanced intake of protein and constituent amino acids (AAs) requires adjustments to total food intake (protein leverage [PL]) and food selection to balance deficits and excesses (complementary feeding). We provided mice with choices of casein and whey, 2 protein sources that are complementary in AA balance, across a range of protein concentrations (P%) of digestible energy (DE). OBJECTIVES We aimed to determine if: 1) PL operates similarly for casein and whey; 2) one protein source is preferred at control P%; 3) the preference changes as P% falls; and 4) AA intakes under control and low P% levels identify AAs that drive changes in protein selection. METHODS Food intake and plasma fibroblast growth factor-21 (FGF21) concentrations were measured in mice at various P% (P7.5%-P33%). For direct comparisons, defined diets were used in which the protein source was either casein or whey. In food choice studies, mice had access to foods in which both casein and whey were provided at the same P% level at the same time. RESULTS PL operated at different P% thresholds in casein (13%)- and whey (10%)-based diets, and the magnitude of PL was greater for casein. Although mice preferred casein under control conditions (P23%), a pronounced preference shift to whey occurred as P% fell to P13% and P10%. At low P%, increases in food intake were accompanied by increases in plasma FGF21, a protein hunger signal. Among AAs deficient in casein and enriched in whey, the intake of Cys was the most invariant as P% changed between P23% and P10%, appearing to drive the switch in protein preference. CONCLUSIONS Mice selected between complementary protein sources, casein and whey, achieving stable total energy intake and regulated intake of AAs as P% varied. Supplementation of low P% casein diets with one whey-enriched AA, Cys, suppressed plasma FGF21 and total food intake.
Collapse
Affiliation(s)
- Kamrul Zaman
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Hee-Chang Mun
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Arthur D Conigrave
- Charles Perkins Centre and School of Life & Environmental Sciences, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Hauck AK, Mehmood R, Carpenter BJ, Frankfurter MT, Tackenberg MC, Inoue SI, Krieg MK, Cassim Bawa FN, Midha MK, Zundell DM, Batmanov K, Lazar MA. Nuclear receptor corepressors non-canonically drive glucocorticoid receptor-dependent activation of hepatic gluconeogenesis. Nat Metab 2024; 6:825-836. [PMID: 38622413 PMCID: PMC11459266 DOI: 10.1038/s42255-024-01029-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 03/07/2024] [Indexed: 04/17/2024]
Abstract
Nuclear receptor corepressors (NCoRs) function in multiprotein complexes containing histone deacetylase 3 (HDAC3) to alter transcriptional output primarily through repressive chromatin remodelling at target loci1-5. In the liver, loss of HDAC3 causes a marked hepatosteatosis largely because of de-repression of genes involved in lipid metabolism6,7; however, the individual roles and contribution of other complex members to hepatic and systemic metabolic regulation are unclear. Here we show that adult loss of both NCoR1 and NCoR2 (double knockout (KO)) in hepatocytes phenocopied the hepatomegalic fatty liver phenotype of HDAC3 KO. In addition, double KO livers exhibited a dramatic reduction in glycogen storage and gluconeogenic gene expression that was not observed with hepatic KO of individual NCoRs or HDAC3, resulting in profound fasting hypoglycaemia. This surprising HDAC3-independent activation function of NCoR1 and NCoR2 is due to an unexpected loss of chromatin accessibility on deletion of NCoRs that prevented glucocorticoid receptor binding and stimulatory effect on gluconeogenic genes. These studies reveal an unanticipated, non-canonical activation function of NCoRs that is required for metabolic health.
Collapse
Affiliation(s)
- Amy K Hauck
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rashid Mehmood
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bryce J Carpenter
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maxwell T Frankfurter
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Tackenberg
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shin-Ichi Inoue
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria K Krieg
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fathima N Cassim Bawa
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohit K Midha
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Delaine M Zundell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kirill Batmanov
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Yang Z, Zarbl H, Guo GL. Circadian Regulation of Endocrine Fibroblast Growth Factors on Systemic Energy Metabolism. Mol Pharmacol 2024; 105:179-193. [PMID: 38238100 PMCID: PMC10877735 DOI: 10.1124/molpharm.123.000831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/05/2024] [Indexed: 02/17/2024] Open
Abstract
The circadian clock is an endogenous biochemical timing system that coordinates the physiology and behavior of organisms to earth's ∼24-hour circadian day/night cycle. The central circadian clock synchronized by environmental cues hierarchically entrains peripheral clocks throughout the body. The circadian system modulates a wide variety of metabolic signaling pathways to maintain whole-body metabolic homeostasis in mammals under changing environmental conditions. Endocrine fibroblast growth factors (FGFs), namely FGF15/19, FGF21, and FGF23, play an important role in regulating systemic metabolism of bile acids, lipids, glucose, proteins, and minerals. Recent evidence indicates that endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between peripheral clocks and energy homeostasis by regulating the expression of metabolic enzymes and hormones. Circadian disruption induced by environmental stressors or genetic ablation is associated with metabolic dysfunction and diurnal disturbances in FGF signaling pathways that contribute to the pathogenesis of metabolic diseases. Time-restricted feeding strengthens the circadian pattern of metabolic signals to improve metabolic health and prevent against metabolic diseases. Chronotherapy, the strategic timing of medication administration to maximize beneficial effects and minimize toxic effects, can provide novel insights into linking biologic rhythms to drug metabolism and toxicity within the therapeutical regimens of diseases. Here we review the circadian regulation of endocrine FGF signaling in whole-body metabolism and the potential effect of circadian dysfunction on the pathogenesis and development of metabolic diseases. We also discuss the potential of chrononutrition and chronotherapy for informing the development of timing interventions with endocrine FGFs to optimize whole-body metabolism in humans. SIGNIFICANCE STATEMENT: The circadian timing system governs physiological, metabolic, and behavioral functions in living organisms. The endocrine fibroblast growth factor (FGF) family (FGF15/19, FGF21, and FGF23) plays an important role in regulating energy and mineral metabolism. Endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between circadian clocks and metabolic homeostasis. Chronic disruption of circadian rhythms increases the risk of metabolic diseases. Chronological interventions such as chrononutrition and chronotherapy provide insights into linking biological rhythms to disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenning Yang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| | - Helmut Zarbl
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| |
Collapse
|
11
|
Zangerolamo L, Carvalho M, Velloso LA, Barbosa HCL. Endocrine FGFs and their signaling in the brain: Relevance for energy homeostasis. Eur J Pharmacol 2024; 963:176248. [PMID: 38056616 DOI: 10.1016/j.ejphar.2023.176248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Since their discovery in 2000, there has been a continuous expansion of studies investigating the physiology, biochemistry, and pharmacology of endocrine fibroblast growth factors (FGFs). FGF19, FGF21, and FGF23 comprise a subfamily with attributes that distinguish them from typical FGFs, as they can act as hormones and are, therefore, referred to as endocrine FGFs. As they participate in a broad cross-organ endocrine signaling axis, endocrine FGFs are crucial lipidic, glycemic, and energetic metabolism regulators during energy availability fluctuations. They function as powerful metabolic signals in physiological responses induced by metabolic diseases, like type 2 diabetes and obesity. Pharmacologically, FGF19 and FGF21 cause body weight loss and ameliorate glucose homeostasis and energy expenditure in rodents and humans. In contrast, FGF23 expression in mice and humans has been linked with insulin resistance and obesity. Here, we discuss emerging concepts in endocrine FGF signaling in the brain and critically assess their putative role as therapeutic targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Lucas Zangerolamo
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Marina Carvalho
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Helena C L Barbosa
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil.
| |
Collapse
|
12
|
Na K, Park YJ. Protein Restriction in Metabolic Health: Lessons from Rodent Models. Nutrients 2024; 16:229. [PMID: 38257122 PMCID: PMC10819042 DOI: 10.3390/nu16020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Consumption of protein-rich diets and supplements has been increasingly advocated by individuals seeking to optimize metabolic health and mitigate the effects of aging. Protein intake is postulated to support muscle mass retention and enhance longevity, underscoring its perceived benefits in age-related metabolic regulation. However, emerging evidence presents a paradox; while moderate protein consumption contributes to health maintenance, an excessive intake is associated with an elevated risk of chronic diseases, notably obesity and diabetes. Furthermore, recent studies suggest that reducing the ratio of protein intake to macronutrients improves metabolic parameters and extends lifespan. The aim of this study is to review the current evidence concerning the metabolic effects of protein-restricted diets and their potential mechanisms. Utilizing rodent models, investigations have revealed that protein-restricted diets exert a notable influence over food intake and energy consumption, ultimately leading to body weight loss, depending on the degree of dietary protein restriction. These phenotypic alterations are primarily mediated by the FGF21 signaling pathway, whose activation is likely regulated by ATF4 and the circadian clock. The evidence suggests that protein-restricted diets as an alternative approach to calorie-restricted regimes, particularly in overweight or obese adults. However, more research is needed to determine the optimal level of restriction, duration, and long-term effects of such interventions.
Collapse
Affiliation(s)
- Khuhee Na
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea;
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea;
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
13
|
Zhang Q, Chen Y, Li J, Xia H, Tong Y, Liu Y. Recent Advances in Hepatic Metabolic Regulation by the Nuclear Factor Rev-erbɑ. Curr Drug Metab 2024; 25:2-12. [PMID: 38409696 DOI: 10.2174/0113892002290055240212074758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Rev-erbɑ (NR1D1) is a nuclear receptor superfamily member that plays a vital role in mammalian molecular clocks and metabolism. Rev-erbɑ can regulate the metabolism of drugs and the body's glucose metabolism, lipid metabolism, and adipogenesis. It is even one of the important regulatory factors regulating the occurrence of metabolic diseases (e.g., diabetes, fatty liver). Metabolic enzymes mediate most drug metabolic reactions in the body. Rev-erbɑ has been recognized to regulate drug metabolic enzymes (such as Cyp2b10 and Ugt1a9). Therefore, this paper mainly reviewed that Rev-erbɑ regulates I and II metabolic enzymes in the liver to affect drug pharmacokinetics. The expression of these drug metabolic enzymes (up-regulated or down-regulated) is related to drug exposure and effects/ toxicity. In addition, our discussion extends to Rev-erbɑ regulating some transporters (such as P-gp, Mrp2, and Bcrp), as they also play an essential role in drug metabolism. Finally, we briefly describe the role and mechanism of nuclear receptor Rev-erbɑ in lipid and glucose homeostasis, obesity, and metabolic disorders syndrome. In conclusion, this paper aims to understand better the role and mechanism of Rev-erbɑ in regulating drug metabolism, lipid, glucose homeostasis, obesity, and metabolic disorders syndrome, which explores how to target Rev-erbɑ to guide the design and development of new drugs and provide scientific reference for the molecular mechanism of new drug development, rational drug use, and drug interaction.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yutong Chen
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jingqi Li
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Haishan Xia
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yongbin Tong
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yuyu Liu
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
14
|
Zhang-Sun ZY, Xu XZ, Escames G, Lei WR, Zhao L, Zhou YZ, Tian Y, Ren YN, Acuña-Castroviejo D, Yang Y. Targeting NR1D1 in organ injury: challenges and prospects. Mil Med Res 2023; 10:62. [PMID: 38072952 PMCID: PMC10712084 DOI: 10.1186/s40779-023-00495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Nuclear receptor subfamily 1, group D, member 1 (NR1D1, also known as REV-ERBα) belongs to the nuclear receptor (NR) family, and is a heme-binding component of the circadian clock that consolidates circadian oscillators. In addition to repressing the transcription of multiple clock genes associated with circadian rhythms, NR1D1 has a wide range of downstream target genes that are intimately involved in many physiopathological processes, including autophagy, immunity, inflammation, metabolism and aging in multiple organs. This review focuses on the pivotal role of NR1D1 as a key transcription factor in the gene regulatory network, with particular emphasis on the milestones of the latest discoveries of NR1D1 ligands. NR1D1 is considered as a promising drug target for treating diverse diseases and may contribute to research on innovative biomarkers and therapeutic targets for organ injury-related diseases. Further research on NR1D1 ligands in prospective human trials may pave the way for their clinical application in many organ injury-related disorders.
Collapse
Affiliation(s)
- Zi-Yin Zhang-Sun
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Xue-Zeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Germaine Escames
- Biomedical Research Center, Department of Physiology, Faculty of Medicine, Institute of Biotechnology, Technological Park of Health Sciences, University of Granada, 18016, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Ibs.Granada, San Cecilio University Hospital, 18016, Granada, Spain
| | - Wang-Rui Lei
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ya-Zhe Zhou
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Ye Tian
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Ya-Nan Ren
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Department of Physiology, Faculty of Medicine, Institute of Biotechnology, Technological Park of Health Sciences, University of Granada, 18016, Granada, Spain.
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Ibs.Granada, San Cecilio University Hospital, 18016, Granada, Spain.
- UGC of Clinical Laboratories, San Cecilio Clinical University Hospital, 18016, Granada, Spain.
| | - Yang Yang
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China.
| |
Collapse
|
15
|
Liu B, Zhong Y, Huang D, Yang L, Wang P, Yang L, Zhang F, Li X, Liang M, Huang K, Du M. LncRNA Nron deficiency protects mice from diet-induced adiposity and hepatic steatosis. Metabolism 2023; 148:155609. [PMID: 37277059 DOI: 10.1016/j.metabol.2023.155609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 06/07/2023]
Abstract
Obesity, as a worldwide healthcare problem, has attracted more and more attention. Here we identify a long non-coding RNA NRON, which is highly conserved across species, as an important regulator of glucose/lipid metabolism and whole-body energy expenditure. Depletion of Nron leads to metabolic benefits in DIO (diet-induced obesity) mice, including reduced body weight and fat mass, improved insulin sensitivity and serum lipid parameters, attenuated hepatic steatosis and enhanced adipose function. Mechanistically, Nron deletion improves hepatic lipid homeostasis via PER2/Rev-Erbα/FGF21 axis coupled with AMPK activation, and enhances adipose function via activating the process of triacylglycerol hydrolysis and fatty acid re-esterification (TAG/FA cycling) and coupled metabolic network. These interactive and integrative effects cooperatively account for a healthier metabolic phenotype in NKO (Nron knockout) mice. Genetic or pharmacological inhibition of Nron may have potential for future therapy of obesity.
Collapse
Affiliation(s)
- Bing Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zhong
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuye Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengchao Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China.
| | - Meng Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China; Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Wang J, Luo LZ, Liang DM, Guo C, Huang ZH, Jian XH, Wen J. Recent progress in understanding mitokines as diagnostic and therapeutic targets in hepatocellular carcinoma. World J Clin Cases 2023; 11:5416-5429. [PMID: 37637689 PMCID: PMC10450380 DOI: 10.12998/wjcc.v11.i23.5416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent tumors worldwide and the leading contributor to cancer-related deaths. The progression and metastasis of HCC are closely associated with altered mitochondrial metabolism, including mitochondrial stress response. Mitokines, soluble proteins produced and secreted in response to mitochondrial stress, play an essential immunomodulatory role. Immunotherapy has emerged as a crucial treatment option for HCC. However, a positive response to therapy is typically dependent on the interaction of tumor cells with immune regulation within the tumor microenvironment. Therefore, exploring the specific immunomodulatory mechanisms of mitokines in HCC is essential for improving the efficacy of immunotherapy. This study provides a comprehensive overview of the association between HCC and the immune microenvironment and highlights recent progress in understanding the involvement of mitochondrial function in preserving liver function. In addition, a systematic review of mitokines-mediated immunomodulation in HCC is presented. Finally, the potential diagnostic and therapeutic roles of mitokines in HCC are prospected and summarized. Recent progress in mitokine research represents a new prospect for mitochondrial therapy. Considering the potential of mitokines to regulate immune function, investigating them as a relevant molecular target holds great promise for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Jiang Wang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Lan-Zhu Luo
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Dao-Miao Liang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Chao Guo
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhi-Hong Huang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Xiao-Hong Jian
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
17
|
Zhong D, Cai J, Hu C, Chen J, Zhang R, Fan C, Li S, Zhang H, Xu Z, Jia Z, Guo D, Sun Y. Inhibition of mPGES-2 ameliorates NASH by activating NR1D1 via heme. Hepatology 2023; 78:547-561. [PMID: 35839302 DOI: 10.1002/hep.32671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/27/2022] [Accepted: 07/12/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD), a complex metabolic syndrome, has limited therapeutic options. Microsomal prostaglandin E synthase-2 (mPGES-2) was originally discovered as a prostaglandin E 2 (PGE 2 ) synthase; however, it does not produce PGE 2 in the liver. Moreover, the role of mPGES-2 in NAFLD remains undefined. Herein, we aimed to determine the function and mechanism of mPGES-2 in liver steatosis and steatohepatitis. APPROACH AND RESULTS To evaluate the role of mPGES-2 in NAFLD, whole-body or hepatocyte-specific mPGES-2-deficient mice fed a high-fat or methionine-choline-deficient diet were used. Compared with control mice, mPGES-2-deficient mice showed reduced hepatic lipid accumulation, along with ameliorated liver injury, inflammation, and fibrosis. Furthermore, the protective effect of mPGES-2 deficiency against NAFLD was dependent on decreased cytochrome P450 4A14 and increased acyl-CoA thioesterase 4 levels regulated by the heme receptor nuclear receptor subfamily 1 group D member 1 (NR1D1), but not PGE 2 . Heme regulated the increased NR1D1 activity mediated by mPGES-2 deficiency. Further, we confirmed the protective role of the mPGES-2 inhibitor SZ0232 in NAFLD therapy. CONCLUSION Our study indicates the pathogenic role of mPGES-2 and outlines the mechanism in mediating NAFLD, thereby highlighting the therapeutic potential of mPGES-2 inhibition in liver steatosis and steatohepatitis.
Collapse
Affiliation(s)
- Dandan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jie Cai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Cheng Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jingshuo Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Rumeng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Department of Pharmacology , Xuzhou Central Hospital , Xuzhou , Jiangsu , China
| | - Chenyu Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Shanshan Li
- Jiangsu Medical Engineering Research Center of Gene Detection , Xuzhou , Jiangsu , China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhou Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| |
Collapse
|
18
|
Navarro-Perez J, Vidal-Puig A, Carobbio S. Recent developments in adipose tissue-secreted factors and their target organs. Curr Opin Genet Dev 2023; 80:102046. [PMID: 37099831 DOI: 10.1016/j.gde.2023.102046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023]
Abstract
The white adipose tissue's primary roles are to store and mobilise energy, which is very different from the brown adipose tissue's function of using fuel to generate heat and maintain the body temperature. The adipose tissues (ATs), co-ordinately with the other organs, sense energetic demands and inform of their reserves before embarking on energetically demanding physiological functions. It is not surprising that ATs exhibit highly integrated regulatory mechanisms mediated by a diversified secretome, including adipokines, lipokines, metabolites and a repertoire of extracellular miRNAs that contribute to integrating the function of the AT niche and connect the AT through paracrine and endocrine effects with the whole organism. Characterising the adipose secretome, its changes in health and disease, regulation by ageing and gender and their contribution to energy homoeostasis is necessary to optimise its use for personalised strategies to prevent or reverse metabolic diseases.
Collapse
Affiliation(s)
- Jaime Navarro-Perez
- Centro de Investigacion Principe Felipe, Valencia, Spain. https://twitter.com/@JaimeNavarroPr1
| | - Antonio Vidal-Puig
- Centro de Investigacion Principe Felipe, Valencia, Spain; Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
19
|
Zhang Y, Liu L, Zhao X, Yan S, Zeng F, Zhou D. New insight into ischemic stroke: Circadian rhythm in post-stroke angiogenesis. Front Pharmacol 2022; 13:927506. [PMID: 36016550 PMCID: PMC9395980 DOI: 10.3389/fphar.2022.927506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
The circadian rhythm is an endogenous clock system that coordinates and optimizes various physiological and pathophysiological processes, which accord with the master and the peripheral clock. Increasing evidence indicates that endogenous circadian rhythm disruption is involved in the lesion volume and recovery of ischemic stroke. As a critical recovery mechanism in post-stroke, angiogenesis reestablishes the regional blood supply and enhances cognitive and behavioral abilities, which is mainly composed of the following processes: endothelial cell proliferation, migration, and pericyte recruitment. The available evidence revealed that the circadian governs many aspects of angiogenesis. This study reviews the mechanism by which circadian rhythms regulate the process of angiogenesis and its contribution to functional recovery in post-stroke at the aspects of the molecular level. A comprehensive understanding of the circadian clock regulating angiogenesis in post-stroke is expected to develop new strategies for the treatment of cerebral infarction.
Collapse
Affiliation(s)
- Yuxing Zhang
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Desheng Zhou,
| |
Collapse
|
20
|
Meng W, Xiao T, Liang X, Wen J, Peng X, Wang J, Zou Y, Liu J, Bialowas C, Luo H, Zhang Y, Liu B, Zhang J, Hu F, Liu M, Dong LQ, Zhou Z, Liu F, Bai J. The miR-182-5p/FGF21/acetylcholine axis mediates the crosstalk between adipocytes and macrophages to promote beige fat thermogenesis. JCI Insight 2021; 6:150249. [PMID: 34264867 PMCID: PMC8492300 DOI: 10.1172/jci.insight.150249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022] Open
Abstract
A dynamically regulated microenvironment, which is mediated by crosstalk between adipocytes and neighboring cells, is critical for adipose tissue homeostasis and function. However, information on key molecules and/or signaling pathways regulating the crosstalk remains limited. In this study, we identify adipocyte miRNA-182-5p (miR-182-5p) as a crucial antiobesity molecule that stimulated beige fat thermogenesis by promoting the crosstalk between adipocytes and macrophages. miR-182-5p was highly enriched in thermogenic adipocytes, and its expression was markedly stimulated by cold exposure in mice. In contrast, miR-182-5p expression was significantly reduced in adipose tissues of obese humans and mice. Knockout of miR-185-5p decreased cold-induced beige fat thermogenesis whereas overexpression of miR-185-5p increased beiging and thermogenesis in mice. Mechanistically, miR-182-5p promoted FGF21 expression and secretion in adipocytes by suppressing nuclear receptor subfamily 1 group D member 1 (Nr1d1) at 5'-UTR, which in turn stimulates acetylcholine synthesis and release in macrophages. Increased acetylcholine expression activated the nicotine acetylcholine receptor in adipocytes, which stimulated PKA signaling and consequent thermogenic gene expression. Our study reveals a key role of the miR-182-5p/FGF21/acetylcholine/acetylcholine receptor axis that mediates the crosstalk between adipocytes and macrophages to promote beige fat thermogenesis. Activation of the miR-182-5p-induced signaling pathway in adipose tissue may be an effective approach to ameliorate obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Wen Meng
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ting Xiao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiuci Liang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Wen
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pharmacology and
| | - Xinyi Peng
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pharmacology and
| | - Jing Wang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yi Zou
- Greehey Children’s Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Jiahao Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Christie Bialowas
- Division of Plastic Surgery, Department of Surgery, Albany Medical Center, Albany, New York, USA
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yacheng Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Ürümqi, China
| | - Bilian Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Meilian Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Lily Q. Dong
- Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pharmacology and
| | - Juli Bai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pharmacology and
| |
Collapse
|
21
|
Mitochondrial Metabolic Signatures in Hepatocellular Carcinoma. Cells 2021; 10:cells10081901. [PMID: 34440674 PMCID: PMC8391498 DOI: 10.3390/cells10081901] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. HCC progression and metastasis are closely related to altered mitochondrial metabolism, including mitochondrial stress responses, metabolic reprogramming, and mitoribosomal defects. Mitochondrial oxidative phosphorylation (OXPHOS) defects and reactive oxygen species (ROS) production are attributed to mitochondrial dysfunction. In response to oxidative stress caused by increased ROS production, misfolded or unfolded proteins can accumulate in the mitochondrial matrix, leading to initiation of the mitochondrial unfolded protein response (UPRmt). The mitokines FGF21 and GDF15 are upregulated during UPRmt and their levels are positively correlated with liver cancer development, progression, and metastasis. In addition, mitoribosome biogenesis is important for the regulation of mitochondrial respiration, cell viability, and differentiation. Mitoribosomal defects cause OXPHOS impairment, mitochondrial dysfunction, and increased production of ROS, which are associated with HCC progression in mouse models and human HCC patients. In this paper, we focus on the role of mitochondrial metabolic signatures in the development and progression of HCC. Furthermore, we provide a comprehensive review of cell autonomous and cell non-autonomous mitochondrial stress responses during HCC progression and metastasis.
Collapse
|
22
|
Abstract
Mammals undergo regular cycles of fasting and feeding that engage dynamic transcriptional responses in metabolic tissues. Here we review advances in our understanding of the gene regulatory networks that contribute to hepatic responses to fasting and feeding. The advent of sequencing and -omics techniques have begun to facilitate a holistic understanding of the transcriptional landscape and its plasticity. We highlight transcription factors, their cofactors, and the pathways that they impact. We also discuss physiological factors that impinge on these responses, including circadian rhythms and sex differences. Finally, we review how dietary modifications modulate hepatic gene expression programs.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Rohith Nagari
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
23
|
Badakhshi Y, Jin T. Current understanding and controversies on the clinical implications of fibroblast growth factor 21. Crit Rev Clin Lab Sci 2020; 58:311-328. [PMID: 33382006 DOI: 10.1080/10408363.2020.1864278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metabolic functions of the hepatic hormone fibroblast growth factor 21 (FGF21) have been recognized for more than a decade in studying the responses of human subjects and rodent models to nutritional stresses such as fasting, high-fat diet or ketogenic diet consumption, and ethanol intake. Our interest in the beneficial metabolic effects of FGF21 has risen due to its potential ability to serve as a therapeutic agent for various metabolic disorders, including type 2 diabetes, obesity, and fatty liver diseases, as well as its potential to act as a diagnostic or prognostic biomarker for metabolic and other disorders. Here, we briefly review the FGF21 gene and protein structures, its expression pattern, and cellular signaling cascades that mediate FGF21 production and function. We mainly focus on discussing experimental and clinical literature pertaining to FGF21 as a therapeutic agent. Furthermore, we present several lines of investigation, including a few studies conducted by our team, suggesting that FGF21 expression and function can be regulated by dietary polyphenol interventions. Finally, we discuss the literature debating FGF21 as a potential biomarker in various disorders.
Collapse
Affiliation(s)
- Yasaman Badakhshi
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Banting and Best Diabetes Center, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Banting and Best Diabetes Center, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Song X, Hu H, Zhao M, Ma T, Gao L. Prospects of circadian clock in joint cartilage development. FASEB J 2020; 34:14120-14135. [PMID: 32946614 DOI: 10.1096/fj.202001597r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022]
Abstract
Altering the food intake, exercise, and sleep patterns have a great influence on the homeostasis of the biological clock. This leads to accelerated aging of the articular cartilage, susceptibility to arthropathy and other aspects. Deficiency or overexpression of certain circadian clock-related genes accelerates the cartilage deterioration and leads to phenotypic variation in different joints. The process of joint cartilage development includes the formation of joint site, interzone, joint cavitation, epiphyseal ossification center, and cartilage maturation. The mechanism by which, biological clock regulates the cell-cycle, growth, metabolism, and other biological processes of chondrocytes is poorly understood. Here, we summarized the interaction between biological clock proteins and developmental pathways in chondrogenesis and provided the evidence from other tissues that further predicts the molecular patterns of these protein-protein networks in activation, proliferation, and differentiation. The purpose of this review is to gain deeper understanding of the evolution of cartilage and its irreversibility seen in damage and aging.
Collapse
Affiliation(s)
- Xiaopeng Song
- Heilongjiang Key Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hailong Hu
- Heilongjiang Key Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingchao Zhao
- Heilongjiang Key Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tianwen Ma
- Heilongjiang Key Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Li Gao
- Heilongjiang Key Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
25
|
Flicker D, Sancak Y, Mick E, Goldberger O, Mootha VK. Exploring the In Vivo Role of the Mitochondrial Calcium Uniporter in Brown Fat Bioenergetics. Cell Rep 2020; 27:1364-1375.e5. [PMID: 31042465 PMCID: PMC7231522 DOI: 10.1016/j.celrep.2019.04.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/28/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
The mitochondrial calcium uniporter has been proposed to coordinate the organelle’s energetics with calcium signaling. Uniporter current has previously been reported to be extremely high in brown adipose tissue (BAT), yet it remains unknown how the uniporter contributes to BAT physiology. Here, we report the generation and characterization of a mouse model lacking Mcu, the pore forming subunit of the uniporter, specifically in BAT (BAT-Mcu-KO). BAT-Mcu-KO mice lack uniporter-based calcium uptake in BAT mitochondria but exhibit unaffected cold tolerance, diet-induced obesity, and transcriptional response to cold in BAT. Unexpectedly, we found in wild-type animals that cold powerfully activates the ATF4-dependent integrated stress response (ISR) in BAT and up-regulates circulating FGF21 and GDF15, raising the hypothesis that the ISR partly underlies the pleiotropic effects of BAT on systemic metabolism. Our study demonstrates that the uniporter is largely dispensable for BAT thermogenesis and demonstrates activation of the ISR in BAT in response to cold. Flicker et al. generate a mouse lacking mitochondrial calcium uniporter activity in brown fat. They show that the uniporter is dispensable for brown fat bioenergetics. Unexpectedly, they find that in wild type animals, cold stress induces ATF4 signaling in normal brown fat, suggesting a mechanism for cold-induced GDF15 and FGF21 elevation.
Collapse
Affiliation(s)
- Daniel Flicker
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA
| | - Yasemin Sancak
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA.
| | - Eran Mick
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA
| | - Olga Goldberger
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02141, USA.
| |
Collapse
|
26
|
Li T, Zhang G, Wang L, Li S, Xu X, Gao Y. Defects in mTORC1 Network and mTORC1-STAT3 Pathway Crosstalk Contributes to Non-inflammatory Hepatocellular Carcinoma. Front Cell Dev Biol 2020; 8:225. [PMID: 32363190 PMCID: PMC7182440 DOI: 10.3389/fcell.2020.00225] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023] Open
Abstract
Background and Aims Mammalian target of rapamycin complex 1 (mTORC1) is frequently hyperactivated in hepatocellular carcinoma (HCC). Cases of HCC without inflammation and cirrhosis are not rarely seen in clinics. However, the molecular basis of non-inflammatory HCC remains unclear. Methods Spontaneous non-inflammatory HCC in mice was triggered by constitutive elevation of mTORC1 by liver-specific TSC1 knockout (LTsc1KO). A multi-omics approach was utilized on tumor tissues to better understand the molecular basis for the development of HCC in the LTsc1KO model. Results We showed that LTsc1KO in mice triggered spontaneous non-inflammatory HCC, with molecular characteristics similar to those of diethylnitrosamine-mediated non-cirrhotic HCC. Mitochondrial and autophagy defects, as well as hepatic metabolic disorder were manifested in HCC development by LTsc1KO. mTORC1 activation on its own regulated an oncogenic network (DNA-damage-inducible transcript 4, nuclear protein 1, and fibroblast growth factor 21), and mTORC1-signal transducer and activator of transcription pathway crosstalk that altered specific metabolic pathways contributed to the development of non-inflammatory HCC. Conclusion Our findings reveal the mechanisms of mTORC1-driven non-inflammatory HCC and provide insight into further development of a protective strategy against non-inflammatory HCC.
Collapse
Affiliation(s)
- Ting Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guohong Zhang
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, Shantou University Medical College, Shantou, China.,Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linlin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Susu Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoping Xu
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Artificial Organs and Tissue Engineering Centre of Guangdong Province, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Artificial Organs and Tissue Engineering Centre of Guangdong Province, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Léveillé M, Besse-Patin A, Jouvet N, Gunes A, Sczelecki S, Jeromson S, Khan NP, Baldwin C, Dumouchel A, Correia JC, Jannig PR, Boulais J, Ruas JL, Estall JL. PGC-1α isoforms coordinate to balance hepatic metabolism and apoptosis in inflammatory environments. Mol Metab 2020; 34:72-84. [PMID: 32180561 PMCID: PMC7011010 DOI: 10.1016/j.molmet.2020.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/31/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Objective The liver is regularly exposed to changing metabolic and inflammatory environments. It must sense and adapt to metabolic need while balancing resources required to protect itself from insult. Peroxisome proliferator activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional coactivator expressed as multiple, alternatively spliced variants transcribed from different promoters that coordinate metabolic adaptation and protect against inflammation. It is not known how PGC-1α integrates extracellular signals to balance metabolic and anti-inflammatory outcomes. Methods Primary mouse hepatocytes were used to evaluate the role(s) of different PGC-1α proteins in regulating hepatic metabolism and inflammatory signaling downstream of tumor necrosis factor alpha (TNFα). Gene expression and signaling analysis were combined with biochemical measurement of apoptosis using gain- and loss-of-function in vitro and in vivo. Results Hepatocytes expressed multiple isoforms of PGC-1α, including PGC-1α4, which microarray analysis showed had common and isoform-specific functions linked to metabolism and inflammation compared with canonical PGC-1α1. Whereas PGC-1α1 primarily impacted gene programs of nutrient metabolism and mitochondrial biology, TNFα signaling showed several pathways related to innate immunity and cell death downstream of PGC-1α4. Gain- and loss-of-function models illustrated that PGC-1α4 uniquely enhanced expression of anti-apoptotic gene programs and attenuated hepatocyte apoptosis in response to TNFα or lipopolysaccharide (LPS). This was in contrast to PGC-1α1, which decreased the expression of a wide inflammatory gene network but did not prevent hepatocyte death in response to cytokines. Conclusions PGC-1α variants have distinct, yet complementary roles in hepatic responses to metabolism and inflammation, and we identify PGC-1α4 as an important mitigator of apoptosis. Multiple isoforms of PGC-1α are expressed in hepatocytes, including PGC-1α4. PGC-1α1 and PGC-1α4 share many metabolic targets, but PGC-1α4 has unique functions linked to hepatic inflammatory signalling. PGC-1α4 attenuates hepatocyte apoptosis in response to TNFα and LPS in vitro and in vivo. Inflammatory signaling influences PGC-1α4 localization in hepatocytes.
Collapse
Affiliation(s)
- Mélissa Léveillé
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Aurèle Besse-Patin
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Nathalie Jouvet
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Aysim Gunes
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Sarah Sczelecki
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Stewart Jeromson
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada
| | - Naveen P Khan
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Cindy Baldwin
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada
| | - Annie Dumouchel
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada
| | - Jorge C Correia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Paulo R Jannig
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Boulais
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jennifer L Estall
- Institut de recherches cliniques de Montreal (IRCM), Montreal, Quebec, Canada; Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
28
|
Fibroblast growth Factor-21 promotes ketone body utilization in neurons through activation of AMP-dependent kinase. Mol Cell Neurosci 2019; 101:103415. [DOI: 10.1016/j.mcn.2019.103415] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
|
29
|
Desterke C, Chiappini F. Lipid Related Genes Altered in NASH Connect Inflammation in Liver Pathogenesis Progression to HCC: A Canonical Pathway. Int J Mol Sci 2019; 20:ijms20225594. [PMID: 31717414 PMCID: PMC6888337 DOI: 10.3390/ijms20225594] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is becoming a public health problem worldwide. While the number of research studies on NASH progression rises every year, sometime their findings are controversial. To identify the most important and commonly described findings related to NASH progression, we used an original bioinformatics, integrative, text-mining approach that combines PubMed database querying and available gene expression omnibus dataset. We have identified a signature of 25 genes that are commonly found to be dysregulated during steatosis progression to NASH and cancer. These genes are implicated in lipid metabolism, insulin resistance, inflammation, and cancer. They are functionally connected, forming the basis necessary for steatosis progression to NASH and further progression to hepatocellular carcinoma (HCC). We also show that five of the identified genes have genome alterations present in HCC patients. The patients with these genes associated to genome alteration are associated with a poor prognosis. In conclusion, using an integrative literature- and data-mining approach, we have identified and described a canonical pathway underlying progression of NASH. Other parameters (e.g., polymorphisms) can be added to this pathway that also contribute to the progression of the disease to cancer. This work improved our understanding of the molecular basis of NASH progression and will help to develop new therapeutic approaches.
Collapse
Affiliation(s)
| | - Franck Chiappini
- Laboratoire Croissance, Régénération, Réparation et Régénération Tissulaires (CRRET)/ EAC CNRS 7149, Univ Paris-Est Créteil (UPEC), F-94010 Créteil, France
- Correspondence: ; Tel.: +33-(0)1-45177080; Fax: +33-(0)1-45171816
| |
Collapse
|
30
|
Stölzel U, Doss MO, Schuppan D. Clinical Guide and Update on Porphyrias. Gastroenterology 2019; 157:365-381.e4. [PMID: 31085196 DOI: 10.1053/j.gastro.2019.04.050] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 12/24/2022]
Abstract
Physicians should be aware of porphyrias, which could be responsible for unexplained gastrointestinal, neurologic, or skin disorders. Despite their relative rarity and complexity, most porphyrias can be easily defined and diagnosed. They are caused by well-characterized enzyme defects in the complex heme biosynthetic pathway and are divided into categories of acute vs non-acute or hepatic vs erythropoietic porphyrias. Acute hepatic porphyrias (acute intermittent porphyria, variegate porphyria, hereditary coproporphyria, and aminolevulinic acid dehydratase deficient porphyria) manifest in attacks and are characterized by overproduction of porphyrin precursors, producing often serious abdominal, psychiatric, neurologic, or cardiovascular symptoms. Patients with variegate porphyria and hereditary coproporphyria can present with skin photosensitivity. Diagnosis relies on measurement of increased urinary 5-aminolevulinic acid (in patients with aminolevulinic acid dehydratase deficient porphyria) or increased 5-aminolevulinic acid and porphobilinogen (in patients with other acute porphyrias). Management of attacks requires intensive care, strict avoidance of porphyrinogenic drugs and other precipitating factors, caloric support, and often heme therapy. The non-acute porphyrias are porphyria cutanea tarda, erythropoietic protoporphyria, X-linked protoporphyria, and the rare congenital erythropoietic porphyria. They lead to the accumulation of porphyrins that cause skin photosensitivity and occasionally severe liver damage. Secondary elevated urinary or blood porphyrins can occur in patients without porphyria, for example, in liver diseases, or iron deficiency. Increases in porphyrin precursors and porphyrins are also found in patients with lead intoxication. Patients with porphyria cutanea tarda benefit from iron depletion, hydroxychloroquine therapy, and, if applicable, elimination of the hepatitis C virus. An α-melanocyte-stimulating hormone analogue can reduce sunlight sensitivity in patients with erythropoietic protoporphyria or X-linked protoporphyria. Strategies to address dysregulated or dysfunctional steps within the heme biosynthetic pathway are in development.
Collapse
Affiliation(s)
- Ulrich Stölzel
- Saxony Porphyria Center, Department of Internal Medicine II, Klinikum Chemnitz, Chemnitz, Germany
| | - Manfred O Doss
- German Competence Center for Porphyria Diagnosis and Consultation, Marburg, Germany; Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
31
|
Yuan D, Xiao D, Gao Q, Zeng L. PGC-1α activation: a therapeutic target for type 2 diabetes? Eat Weight Disord 2019; 24:385-395. [PMID: 30498989 DOI: 10.1007/s40519-018-0622-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/24/2018] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) has gained popularity as a very attractive target for diabetic therapies due to its role in lipid and glucose metabolism. Pharmacological activation of PGC-1α is thought to elicit health benefits. However, this notion has been questioned by increasing evidence, which suggests that insulin resistant is exacerbated when PGC-1α expression is far beyond normal physiological limits and is prevented under the condition of PGC-1α deficiency. This narrative review suggests that PGC-1α, as a master metabolic regulator, exerts roles in insulin sensitivity in a tissue-specific manner and in a physical activity/age-dependent fashion. When using PGC-1α as a target for therapeutic strategies against insulin resistance and T2DM, we should take these factors into consideration.Level of evidence: Level V, narrative review.
Collapse
Affiliation(s)
- Daixiu Yuan
- Department of Medicine, Jishou University, Jishou, 41600, Hunan, China
| | - Dingfu Xiao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Qian Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Liming Zeng
- Science College of Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| |
Collapse
|
32
|
Delezie J, Handschin C. Endocrine Crosstalk Between Skeletal Muscle and the Brain. Front Neurol 2018; 9:698. [PMID: 30197620 PMCID: PMC6117390 DOI: 10.3389/fneur.2018.00698] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/02/2018] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle is an essential regulator of energy homeostasis and a potent coordinator of exercise-induced adaptations in other organs including the liver, fat or the brain. Skeletal muscle-initiated crosstalk with other tissues is accomplished though the secretion of myokines, protein hormones which can exert autocrine, paracrine and long-distance endocrine effects. In addition, the enhanced release or uptake of metabolites from and into contracting muscle cells, respectively, likewise can act as a powerful mediator of tissue interactions, in particular in regard to the central nervous system. The present review will discuss the current stage of knowledge regarding how exercise and the muscle secretome improve a broad range of brain functions related to vascularization, neuroplasticity, memory, sleep and mood. Even though the molecular and cellular mechanisms underlying the communication between muscle and brain is still poorly understood, physical activity represents one of the most effective strategies to reduce the prevalence and incidence of depression, cognitive, metabolic or degenerative neuronal disorders, and thus warrants further study.
Collapse
|
33
|
Zhang Y, Higgins CB, Mayer AL, Mysorekar IU, Razani B, Graham MJ, Hruz PW, DeBosch BJ. TFEB-dependent induction of thermogenesis by the hepatocyte SLC2A inhibitor trehalose. Autophagy 2018; 14:1959-1975. [PMID: 29996716 PMCID: PMC6152536 DOI: 10.1080/15548627.2018.1493044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023] Open
Abstract
The macroautophagy/autophagy-inducing disaccharide, trehalose, has been proposed to be a promising therapeutic agent against neurodegenerative and cardiometabolic diseases. We recently showed that trehalose attenuates hepatic steatosis in part by blocking hepatocyte glucose transport to induce hepatocyte autophagic flux. However, although every major demonstration of trehalose action invokes activating autophagic flux as its primary function, the mechanism of action of trehalose in whole-body energy metabolism remains poorly defined. Here, we demonstrate that trehalose induces hepatocyte TFEB (transcription factor EB)-dependent thermogenesis in vivo, concomitant with upregulation of hepatic and white adipose expression of UCP1 (uncoupling protein 1 [mitochondrial, protein carrier]). Mechanistically, we provide evidence that hepatocyte fasting transcriptional and metabolic responses depend upon PPARGC1A (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha), TFEB, and FGF21 (fibroblast growth factor 21) signaling. Strikingly, hepatocyte-selective TFEB knockdown abrogated trehalose induction of thermogenesis and white adipose tissue UCP1 upregulation in vivo. In contrast, we found that trehalose action on thermogenesis was independent of LEP (leptin) and the autophagy pathway, as there was robust thermogenic induction in trehalose-treated ob/ob, Becn1, Atg16l1, and Epg5 mutant mice. We conclude that trehalose induces metabolically favorable effects on whole-body thermogenesis in part via hepatocyte-centered fasting-like mechanisms that appear to be independent of autophagic flux. Our findings elucidate a novel mechanism by which trehalose acts as a metabolic therapeutic agent by activating hepatic fasting responses. More broadly, the hepatic glucose fasting response may be of clinical utility against overnutrition-driven disease, such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yiming Zhang
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Allyson L. Mayer
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Indira U. Mysorekar
- Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Babak Razani
- Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark J. Graham
- IONIS Pharmaceuticals, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul W. Hruz
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J. DeBosch
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
Komatsu M, Kanda T, Urai H, Kurokochi A, Kitahama R, Shigaki S, Ono T, Yukioka H, Hasegawa K, Tokuyama H, Kawabe H, Wakino S, Itoh H. NNMT activation can contribute to the development of fatty liver disease by modulating the NAD + metabolism. Sci Rep 2018; 8:8637. [PMID: 29872122 PMCID: PMC5988709 DOI: 10.1038/s41598-018-26882-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Nicotinamide N-methyltransferase (NNMT) catalyses the reaction between nicotinamide (NAM) and S-adenosylmethionine to produce 1-methylnicotinamide and S-adenosylhomocysteine. Recently, this enzyme has also been reported to modulate hepatic nutrient metabolism, but its role in the liver has not been fully elucidated. We developed transgenic mice overexpressing NNMT to elucidate its role in hepatic nutrient metabolism. When fed a high fat diet containing NAM, a precursor for nicotinamide adenine dinucleotide (NAD)+, these NNMT-overexpressing mice exhibit fatty liver deterioration following increased expression of the genes mediating fatty acid uptake and decreased very low-density lipoprotein secretion. NNMT overactivation decreased the NAD+ content in the liver and also decreased gene activity related to fatty acid oxidation by inhibiting NAD+-dependent deacetylase Sirt3 function. Moreover, the transgenic mice showed liver fibrosis, with the induction of inflammatory and fibrosis genes. Induced NNMT expression decreased the tissue methylation capacity, thereby reducing methylation of the connective tissue growth factor (CTGF) gene promoter, resulting in increased CTGF expression. These data indicate that NNMT links the NAD+ and methionine metabolic pathways and promotes liver steatosis and fibrosis. Therefore, targeting NNMT may serve as a therapeutic strategy for treating fatty liver and fibrosis.
Collapse
Affiliation(s)
- Motoaki Komatsu
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Takeshi Kanda
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Hidenori Urai
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Arata Kurokochi
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Rina Kitahama
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | | | | | | | - Kazuhiro Hasegawa
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Hirobumi Tokuyama
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | | | - Shu Wakino
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
| | - Hiroshi Itoh
- Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
35
|
Staiger H, Keuper M, Berti L, Hrabe de Angelis M, Häring HU. Fibroblast Growth Factor 21-Metabolic Role in Mice and Men. Endocr Rev 2017; 38:468-488. [PMID: 28938407 DOI: 10.1210/er.2017-00016] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022]
Abstract
Since its identification in 2000, the interest of scientists in the hepatokine fibroblast growth factor (FGF) 21 has tremendously grown, and still remains high, due to a wealth of very robust data documenting this factor's favorable effects on glucose and lipid metabolism in mice. For more than ten years now, intense in vivo and ex vivo experimentation addressed the physiological functions of FGF21 in humans as well as its pathophysiological role and pharmacological effects in human metabolic disease. This work produced a comprehensive collection of data revealing overlaps in FGF21 expression and function but also significant differences between mice and humans that have to be considered before translation from bench to bedside can be successful. This review summarizes what is known about FGF21 in mice and humans with a special focus on this factor's role in glucose and lipid metabolism and in metabolic diseases, such as obesity and type 2 diabetes mellitus. We highlight the discrepancies between mice and humans and try to decipher their underlying reasons.
Collapse
Affiliation(s)
- Harald Staiger
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Interfaculty Center for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Michaela Keuper
- Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany.,Chair for Experimental Genetics, Technical University Munich, 85764 Neuherberg, Germany
| | - Hans-Ulrich Häring
- Interfaculty Center for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
36
|
Abstract
The physiological identity of every cell is maintained by highly specific transcriptional networks that establish a coherent molecular program that is in tune with nutritional conditions. The regulation of cell-specific transcriptional networks is accomplished by an epigenetic program via chromatin-modifying enzymes, whose activity is directly dependent on metabolites such as acetyl-coenzyme A, S-adenosylmethionine, and NAD+, among others. Therefore, these nuclear activities are directly influenced by the nutritional status of the cell. In addition to nutritional availability, this highly collaborative program between epigenetic dynamics and metabolism is further interconnected with other environmental cues provided by the day-night cycles imposed by circadian rhythms. Herein, we review molecular pathways and their metabolites associated with epigenetic adaptations modulated by histone- and DNA-modifying enzymes and their responsiveness to the environment in the context of health and disease.
Collapse
|
37
|
Harris LALS, Smith GI, Patterson BW, Ramaswamy RS, Okunade AL, Kelly SC, Porter LC, Klein S, Yoshino J, Mittendorfer B. Alterations in 3-Hydroxyisobutyrate and FGF21 Metabolism Are Associated With Protein Ingestion-Induced Insulin Resistance. Diabetes 2017; 66:1871-1878. [PMID: 28473464 PMCID: PMC5482083 DOI: 10.2337/db16-1475] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/24/2017] [Indexed: 12/17/2022]
Abstract
Systemic hyperaminoacidemia, induced by either intravenous amino acid infusion or protein ingestion, reduces insulin-stimulated glucose disposal. Studies of mice suggest that the valine metabolite 3-hydroxyisobutyrate (3-HIB), fibroblast growth factor 21 (FGF21), adiponectin, and nonesterified fatty acids (NEFAs) may be involved in amino acid-mediated insulin resistance. We therefore measured in 30 women the rate of glucose disposal, and plasma 3-HIB, FGF21, adiponectin, and NEFA concentrations, under basal conditions and during a hyperinsulinemic-euglycemic clamp procedure (HECP), with and without concomitant ingestion of protein (n = 15) or an amount of leucine that matched the amount of protein (n = 15). We found that during the HECP without protein or leucine ingestion, the grand mean ± SEM plasma 3-HIB concentration decreased (from 35 ± 2 to 14 ± 1 µmol/L) and the grand median [quartiles] FGF21 concentration increased (from 178 [116, 217] to 509 [340, 648] pg/mL). Ingestion of protein, but not leucine, decreased insulin-stimulated glucose disposal (P < 0.05) and prevented both the HECP-mediated decrease in 3-HIB and increase in FGF21 concentration in plasma. Neither protein nor leucine ingestion altered plasma adiponectin or NEFA concentrations. These findings suggest that 3-HIB and FGF21 might be involved in protein-mediated insulin resistance in humans.
Collapse
Affiliation(s)
- Lydia-Ann L S Harris
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Gordon I Smith
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Bruce W Patterson
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Raja S Ramaswamy
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Adewole L Okunade
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Shannon C Kelly
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Lane C Porter
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Samuel Klein
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Jun Yoshino
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Bettina Mittendorfer
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
38
|
Zhang C, Huang Z, Jing H, Fu W, Yuan M, Xia W, Cai L, Gan X, Chen Y, Zou M, Long M, Wang J, Wang M, Xu D. SAK-HV Triggered a Short-period Lipid-lowering Biotherapy Based on the Energy Model of Liver Proliferation via a Novel Pathway. Am J Cancer Res 2017; 7:1749-1769. [PMID: 28529649 PMCID: PMC5436525 DOI: 10.7150/thno.18415] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 03/02/2017] [Indexed: 01/13/2023] Open
Abstract
The accumulations of excess lipids within liver and serum are defined as non-alcoholic fatty liver disease (NAFLD) and hyperlipemia respectively. Both of them are components of metabolic syndrome that greatly threaten human health. Here, a recombinant fusion protein (SAK-HV) effectively treated NAFLD and hyperlipemia in high-fat-fed ApoE-/- mice, quails and rats within just 14 days. Its triglyceride and cholesterol-lowering effects were significantly better than that of atorvastatin during the observation period. We explored the lipid-lowering mechanism of SAK-HV by the hepatic transcriptome analysis and serials of experiments both in vivo and in vitro. Unexpectedly, SAK-HV triggered a moderate energy and material-consuming liver proliferation to dramatically decrease the lipids from both serum and liver. We provided the first evidence that PGC-1α mediated the hepatic synthesis of female hormones during liver proliferation, and proposed the complement system-induced PGC-1α-estrogen axis via the novel STAT3-C/EBPβ-PGC-1α pathway in liver as a new energy model for liver proliferation. In this model, PGC-1α ignited and fueled hepatocyte activation as an “igniter”; PGC-1α-induced estrogen augmented the energy supply of PGC-1α as an “ignition amplifier”, then triggered the hepatocyte state transition from activation to proliferation as a “starter”, causing triglyceride and cholesterol-lowering effects via PPARα-mediated fatty acid oxidation and LDLr-mediated cholesterol uptake, respectively. Collectively, the SAK-HV-triggered distinctive lipid-lowering strategy based on the new energy model of liver proliferation has potential as a novel short-period biotherapy against NAFLD and hyperlipemia.
Collapse
|
39
|
Lin X, Liu YB, Hu H. Metabolic role of fibroblast growth factor 21 in liver, adipose and nervous system tissues. Biomed Rep 2017; 6:495-502. [PMID: 28515909 PMCID: PMC5431415 DOI: 10.3892/br.2017.890] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/27/2017] [Indexed: 01/04/2023] Open
Abstract
The hepatokine fibroblast growth factor 21 (FGF21) is a novel polypeptide ligand, which is involved in glucose and lipid metabolism, and contributes significantly to lowering body weight and enhancing insulin sensitivity. A large number of pre-clinical and clinical results demonstrate that FGF21 is a potential drug target for treating obesity and type 2 diabetes mellitus. In the present review, the tissue specific actions and molecular mechanisms of FGF21 are discussed with a focus on the liver, adipose tissue and nervous system, as well as investigating the outcomes of clinical trials of FGF21, with the aim of interpreting and delineating the complexity physiology of FGF21.
Collapse
Affiliation(s)
- Xiaolong Lin
- Department of Pathology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| | - Yuan Bo Liu
- The Medical Department of Neurology, The Sixth People's Hospital of Huizhou (The People's Hospital of Huiyang), Huizhou, Guangdong 516211, P.R. China
| | - Huijun Hu
- Department of Pathology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| |
Collapse
|
40
|
Morrice N, Mcilroy GD, Tammireddy SR, Reekie J, Shearer KD, Doherty MK, Delibegović M, Whitfield PD, Mody N. Elevated Fibroblast growth factor 21 (FGF21) in obese, insulin resistant states is normalised by the synthetic retinoid Fenretinide in mice. Sci Rep 2017; 7:43782. [PMID: 28256636 PMCID: PMC5335663 DOI: 10.1038/srep43782] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/30/2017] [Indexed: 01/06/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) has emerged as an important beneficial regulator of glucose and lipid homeostasis but its levels are also abnormally increased in insulin-resistant states in rodents and humans. The synthetic retinoid Fenretinide inhibits obesity and improves glucose homeostasis in mice and has pleotropic effects on cellular pathways. To identify Fenretinide target genes, we performed unbiased RNA-seq analysis in liver from mice fed high-fat diet ± Fenretinide. Strikingly, Fgf21 was the most downregulated hepatic gene. Fenretinide normalised elevated levels of FGF21 in both high-fat diet-induced obese mice and in genetically obese-diabetic Leprdbmice. Moreover, Fenretinide-mediated suppression of FGF21 was independent of body weight loss or improved hepatic insulin sensitivity and importantly does not induce unhealthy metabolic complications. In mice which have substantially decreased endogenous retinoic acid biosynthesis, Fgf21 expression was increased, whereas acute pharmacological retinoid treatment decreased FGF21 levels. The repression of FGF21 levels by Fenretinide occurs by reduced binding of RARα and Pol-II at the Fgf21 promoter. We therefore establish Fgf21 as a novel gene target of Fenretinide signalling via a retinoid-dependent mechanism. These results may be of nutritional and therapeutic importance for the treatment of obesity and type-2 diabetes.
Collapse
Affiliation(s)
- Nicola Morrice
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK.,Centre for Genome Enabled Biology and Medicine, University of Aberdeen, 23 St Machar Drive, Old Aberdeen, Aberdeen, Scotland AB24 3UU, UK
| | - George D Mcilroy
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK
| | - Seshu R Tammireddy
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of Highlands and Islands, Old Perth Road, Inverness, Scotland IV2 3JH, UK
| | - Jennifer Reekie
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK
| | - Kirsty D Shearer
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK
| | - Mary K Doherty
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of Highlands and Islands, Old Perth Road, Inverness, Scotland IV2 3JH, UK
| | - Mirela Delibegović
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK
| | - Phillip D Whitfield
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of Highlands and Islands, Old Perth Road, Inverness, Scotland IV2 3JH, UK
| | - Nimesh Mody
- Institute of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Foresterhill Health Campus, Aberdeen, Scotland AB25 2ZD, UK
| |
Collapse
|
41
|
Donepudi AC, Boehme S, Li F, Chiang JY. G-protein-coupled bile acid receptor plays a key role in bile acid metabolism and fasting-induced hepatic steatosis in mice. Hepatology 2017; 65:813-827. [PMID: 27351453 PMCID: PMC5195921 DOI: 10.1002/hep.28707] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 06/12/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
UNLABELLED Bile acids are signaling molecules that play a critical role in regulation of hepatic metabolic homeostasis by activating nuclear farnesoid X receptor (Fxr) and membrane G-protein-coupled receptor (Takeda G-protein-coupled receptor 5; Tgr5). The role of FXR in regulation of bile acid synthesis and hepatic metabolism has been studied extensively. However, the role of TGR5 in hepatic metabolism has not been explored. The liver plays a central role in lipid metabolism, and impaired response to fasting and feeding contributes to steatosis and nonalcoholic fatty liver and obesity. We have performed a detailed analysis of gallbladder bile acid and lipid metabolism in Tgr5-/- mice in both free-fed and fasted conditions. Lipid profiles of serum, liver and adipose tissues, bile acid composition, energy metabolism, and messenger RNA and protein expression of the genes involved in lipid metabolism were analyzed. Results showed that deficiency of the Tgr5 gene in mice alleviated fasting-induced hepatic lipid accumulation. Expression of liver oxysterol 7α-hydroxylase in the alternative bile acid synthesis pathway was reduced. Analysis of gallbladder bile acid composition showed marked increase of taurocholic acid and decrease of tauro-α and β-muricholic acid in Tgr5-/- mice. Tgr5-/- mice had increased hepatic fatty acid oxidation rate and decreased hepatic fatty acid uptake. Interestingly, fasting induction of fibroblast growth factor 21 in liver was attenuated. In addition, fasted Tgr5-/- mice had increased activation of hepatic growth hormone-signal transducer and activator of transcription 5 (GH-Stat5) signaling compared to wild-type mice. CONCLUSION TGR5 may play a role in determining bile acid composition and in fasting-induced hepatic steatosis through a novel mechanism involving activation of the GH-Stat5 signaling pathway. (Hepatology 2017;65:813-827).
Collapse
Affiliation(s)
- Ajay C. Donepudi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Shannon Boehme
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| | - Feng Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - John Y.L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH
| |
Collapse
|
42
|
Chavan R, Preitner N, Okabe T, Strittmatter LM, Xu C, Ripperger JA, Pitteloud N, Albrecht U. REV-ERBα regulates Fgf21 expression in the liver via hepatic nuclear factor 6. Biol Open 2017; 6:1-7. [PMID: 27875243 PMCID: PMC5278426 DOI: 10.1242/bio.021519] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The circadian clock contributes to the timing of many body functions including metabolism and reproduction. The hepatokine fibroblast growth factor 21 (FGF21) is a critical metabolic regulator involved in modulation of fertility. Here we show that lack of the clock component REV-ERBα elevates FGF21 levels in liver and plasma. At the molecular level, REV-ERBα modulates the expression of FGF21 via the liver-specific hepatic nuclear factor 6 (HNF6). We conclude that REV-ERBα regulates metabolism and reproduction, at least in part, via regulation of Fgf21. Summary: The hepatokine Fgf21 is transcriptionally regulated by the nuclear receptor REV-ERBα with the hepatocyte-specific factor HNF6 to regulate metabolism and fertility.
Collapse
Affiliation(s)
- Rohit Chavan
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | - Nadia Preitner
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Takashi Okabe
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | | | - Cheng Xu
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Jürgen A Ripperger
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne CH1011, Switzerland
| | - Urs Albrecht
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg CH1700, Switzerland
| |
Collapse
|
43
|
Besse-Patin A, Léveillé M, Oropeza D, Nguyen BN, Prat A, Estall JL. Estrogen Signals Through Peroxisome Proliferator-Activated Receptor-γ Coactivator 1α to Reduce Oxidative Damage Associated With Diet-Induced Fatty Liver Disease. Gastroenterology 2017; 152:243-256. [PMID: 27658772 DOI: 10.1053/j.gastro.2016.09.017] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Inefficient fatty acid oxidation in mitochondria and increased oxidative damage are features of non-alcoholic fatty liver disease (NAFLD). In rodent models and patients with NAFLD, hepatic expression of peroxisome proliferator-activated receptor-γ (PPARG) coactivator 1α (PPARGC1A or PGC1A) is inversely correlated with liver fat and disease severity. A common polymorphism in this gene (rs8192678, encoding Gly482Ser) has been associated with NAFLD. We investigated whether reduced expression of PGC1A contributes to development of NAFLD using mouse models, primary hepatocytes, and human cell lines. METHODS HepG2 cells were transfected with variants of PPARGC1A and protein and messenger RNA levels were measured. Mice with liver-specific hemizygous or homozygous disruption of Ppargc1a (Ppargc1af/+Alb-cre+/0 and Ppargc1af/f Alb-cre+/0 mice, respectively) were fed regular chow (control) or a high-fat diet supplemented with 30% d-fructose in drinking water (obesogenic diet) for 25-33 weeks. Liver tissues were analyzed by histology and by immunoblotting. Primary hepatocytes were analyzed for insulin signaling, reactive oxygen species, and estrogen response. Luciferase reporter expression was measured in transfected H2.35 cells expressing an estrogen receptor reporter gene, estrogen receptor 1, and/or PGC1A/B. RESULTS The serine 482 variant of the human PGC1A protein had a shorter half-life than the glycine 482 variant when expressed in HepG2 cells. Liver tissues from mice with liver-specific hemizygous disruption of Ppargc1a placed on an obesogenic diet expressed increased markers of inflammation and fibrosis and decreased levels of antioxidant enzymes compared with the Ppargc1a+/+ on the same diet. Oxidative damage was observed in livers from Ppargc1af/+Alb-cre+/0 mice of each sex, in a cell-autonomous manner, but was greater in livers from the female mice. Expression of PGC1A in H2.35 cells coactivated estrogen receptor 1 and was required for estrogen-dependent expression of genes that encode antioxidant proteins. These findings could account for the increased liver damage observed in female Ppargc1af/+Alb-cre+/0 mice; while, compensatory increases in PPARG coactivator 1β could prevent oxidative damage associated with complete loss of PGC1A expression in Ppargc1af/fAlb-cre+/0 female mice. CONCLUSIONS In mice, loss of estrogen signaling contributes to oxidative damage caused by low levels of PGC1A in liver, exacerbating steatohepatitis associated with diets high in fructose and fat.
Collapse
Affiliation(s)
- Aurèle Besse-Patin
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada
| | - Mélissa Léveillé
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada
| | - Daniel Oropeza
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Bich N Nguyen
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Québec, Canada; University of Montreal Health Network, Montreal, Québec, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada
| | - Jennifer L Estall
- Institut de Recherches Cliniques de Montreal, Montreal, Québec, Canada; Department of Medicine, University of Montreal, Montreal, Québec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
44
|
Zarei M, Barroso E, Leiva R, Barniol-Xicota M, Pujol E, Escolano C, Vázquez S, Palomer X, Pardo V, González-Rodríguez Á, Valverde ÁM, Quesada-López T, Villarroya F, Wahli W, Vázquez-Carrera M. Heme-Regulated eIF2α Kinase Modulates Hepatic FGF21 and Is Activated by PPARβ/δ Deficiency. Diabetes 2016; 65:3185-99. [PMID: 27486236 DOI: 10.2337/db16-0155] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/12/2016] [Indexed: 11/13/2022]
Abstract
Fibroblast growth factor 21 (FGF21), a peptide hormone with pleiotropic effects on carbohydrate and lipid metabolism, is considered a target for the treatment of diabetes. We investigated the role of peroxisome proliferator-activated receptor (PPAR) β/δ deficiency in hepatic FGF21 regulation. Increased Fgf21 expression was observed in the livers of PPARβ/δ-null mice and in mouse primary hepatocytes when this receptor was knocked down by small interfering RNA (siRNA). Increased Fgf21 was associated with enhanced protein levels in the heme-regulated eukaryotic translation initiation factor 2α (eIF2α) kinase (HRI). This increase caused enhanced levels of phosphorylated eIF2α and activating transcription factor (ATF) 4, which is essential for Fgf21-induced expression. siRNA analysis demonstrated that HRI regulates Fgf21 expression in primary hepatocytes. Enhanced Fgf21 expression attenuated tunicamycin-induced endoplasmic reticulum stress, as demonstrated by using a neutralizing antibody against FGF21. Of note, increased Fgf21 expression in mice fed a high-fat diet or hepatocytes exposed to palmitate was accompanied by reduced PPARβ/δ and activation of the HRI-eIF2α-ATF4 pathway. Moreover, pharmacological activation of HRI increased Fgf21 expression and reduced lipid-induced hepatic steatosis and glucose intolerance, but these effects were not observed in Fgf21-null mice. Overall, these findings suggest that HRI is a potential target for regulating hepatic FGF21 levels.
Collapse
Affiliation(s)
- Mohammad Zarei
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Rosana Leiva
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
| | - Marta Barniol-Xicota
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
| | - Eugènia Pujol
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
| | - Carmen Escolano
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
| | - Santiago Vázquez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Virginia Pardo
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Águeda González-Rodríguez
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Tania Quesada-López
- Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain Department of Biochemistry and Molecular Biology and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Villarroya
- Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain Department of Biochemistry and Molecular Biology and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore INRA ToxAlim, UMR1331, Chemin de Tournefeuille, Toulouse Cedex, France
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Institute of Biomedicine of the University of Barcelona, Barcelona, Spain CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain Pediatric Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain
| |
Collapse
|
45
|
Rando G, Tan CK, Khaled N, Montagner A, Leuenberger N, Bertrand-Michel J, Paramalingam E, Guillou H, Wahli W. Glucocorticoid receptor-PPARα axis in fetal mouse liver prepares neonates for milk lipid catabolism. eLife 2016; 5. [PMID: 27367842 PMCID: PMC4963200 DOI: 10.7554/elife.11853] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/30/2016] [Indexed: 01/12/2023] Open
Abstract
In mammals, hepatic lipid catabolism is essential for the newborns to efficiently use milk fat as an energy source. However, it is unclear how this critical trait is acquired and regulated. We demonstrate that under the control of PPARα, the genes required for lipid catabolism are transcribed before birth so that the neonatal liver has a prompt capacity to extract energy from milk upon suckling. The mechanism involves a fetal glucocorticoid receptor (GR)-PPARα axis in which GR directly regulates the transcriptional activation of PPARα by binding to its promoter. Certain PPARα target genes such as Fgf21 remain repressed in the fetal liver and become PPARα responsive after birth following an epigenetic switch triggered by β-hydroxybutyrate-mediated inhibition of HDAC3. This study identifies an endocrine developmental axis in which fetal GR primes the activity of PPARα in anticipation of the sudden shifts in postnatal nutrient source and metabolic demands. DOI:http://dx.doi.org/10.7554/eLife.11853.001 Birth is a highly stressful and critical event. In the womb, babies rely on the supply of oxygen and nutrients provided by the placenta. However, once they are born they need to breathe for themselves and gain all their nutrients from suckling milk. The placenta provides a sugar-rich diet, while milk is richer in fat. Failing to cope with this change in diet leads to serious complications and sometimes death. Therefore, a better understanding of how the body adapts to these changes may shed light on pathways that are important for good health in later life. The liver plays a central role in processing the nutrients absorbed by the gut. It uses fats to produce molecules called ketone bodies, such as β-hydroxybutyrate, which are then used as fuel by other tissues and organs including the heart, muscle and the brain. A protein called PPARα controls the production of ketone bodies primarily by regulating genes that are involved in the uptake and breakdown of fat in the liver. However, little is known about how this protein affects the development of the liver. Here, Rando, Tan et al. report that mice start to produce more PPARα in the liver shortly before birth. This ultimately activates several genes that encode enzymes that break down fats. The experiments show that during labor, stress hormones called glucocorticoids directly stimulate the production of PPARα in the liver of the fetus to prepare newborn mice for harnessing energy from fat-rich milk. In the absence of PPARα, mouse liver cells are less able to break down fats after birth and so start to accumulate fat, resulting in fewer ketone bodies being produced. Rando, Tan et al. show that β-hydroxybutyrate regulates some PPARα target genes, including one called Fgf21. The activity of this gene increases only after milk suckling starts and it encodes a protein that enhances the breakdown of fats in the liver. Without PPARα, the expression levels of its target genes, including Fgf21, do not increase after birth, which promotes the build up of fats in liver cells, a condition known as liver steatosis. Overall, the results reported by Rando, Tan et al. highlight how stress during labor plays an important role in priming the body to cope with a fat-rich diet after birth. Future studies will need to determine if stress hormones and ketone bodies could be used as therapies for babies born by caesarean section with liver steatosis. DOI:http://dx.doi.org/10.7554/eLife.11853.002
Collapse
Affiliation(s)
- Gianpaolo Rando
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Chek Kun Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore
| | - Nourhène Khaled
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Alexandra Montagner
- UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| | - Nicolas Leuenberger
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Justine Bertrand-Michel
- IFR 150 Plateforme Metatoul, Institut Fédératif de Recherche Bio-Médicale de Toulouse INSERM U563, Toulouse, France
| | - Eeswari Paramalingam
- Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore
| | - Hervé Guillou
- UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Lee Kong Chian School of Medicine, Nanyang Technological University, , Singapore.,UMR 1331 ToxAlim Research Centre in Food Toxicology, INRA, Université de Toulouse, Toulouse, France
| |
Collapse
|
46
|
Erickson A, Moreau R. The regulation of FGF21 gene expression by metabolic factors and nutrients. Horm Mol Biol Clin Investig 2016; 30:/j/hmbci.ahead-of-print/hmbci-2016-0016/hmbci-2016-0016.xml. [PMID: 27285327 DOI: 10.1515/hmbci-2016-0016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/08/2016] [Indexed: 12/26/2022]
Abstract
Fibroblast growth factor 21 (FGF21) gene expression is altered by a wide array of physiological, metabolic, and environmental factors. Among dietary factors, high dextrose, low protein, methionine restriction, short-chain fatty acids (butyric acid and lipoic acid), and all-trans-retinoic acid were repeatedly shown to induce FGF21 expression and circulating levels. These effects are usually more pronounced in liver or isolated hepatocytes than in adipose tissue or isolated fat cells. Although peroxisome proliferator-activated receptor α (PPARα) is a key mediator of hepatic FGF21 expression and function, including the regulation of gluconeogenesis, ketogenesis, torpor, and growth inhibition, there is increasing evidence of PPARα-independent transactivation of the FGF21 gene by dietary molecules. FGF21 expression is believed to follow the circadian rhythm and be placed under the control of first order clock-controlled transcription factors, retinoic acid receptor-related orphan receptors (RORs) and nuclear receptors subfamily 1 group D (REV-ERBs), with FGF21 rhythm being anti-phase to REV-ERBs. Key metabolic hormones such as glucagon, insulin, and thyroid hormone have presumed or clearly demonstrated roles in regulating FGF21 transcription and secretion. The control of the FGF21 gene by glucagon and insulin appears more complex than first anticipated. Some discrepancies are noted and will need continued studies. The complexity in assessing the significance of FGF21 gene expression resides in the difficulty to ascertain (i) when transcription results in local or systemic increase of FGF21 protein; (ii) if FGF21 is among the first or second order genes upregulated by physiological, metabolic, and environmental stimuli, or merely an epiphenomenon; and (iii) whether FGF21 may have some adverse effects alongside beneficial outcomes.
Collapse
|
47
|
Zhang W, Wang P, Chen S, Zhang Z, Liang T, Liu C. Rhythmic expression of miR-27b-3p targets the clock gene Bmal1 at the posttranscriptional level in the mouse liver. FASEB J 2016; 30:2151-60. [PMID: 26919869 DOI: 10.1096/fj.201500120] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/04/2016] [Indexed: 12/16/2023]
Abstract
Circadian clocks orchestrate daily oscillations in mammalian behaviors, physiology, and gene expression. MicroRNAs (miRNAs) play a crucial role in fine-tuning of the circadian system. However, little is known about the direct regulation of the clock genes by specific miRNAs. In this study, we found that miR-27b-3p exhibits rhythmic expression in the metabolic tissues of the mice subjected to constant darkness. MiR-27b-3p's expression is induced in livers of unfed and ob/ob mice. In addition, the oscillation phases of miR-27b-3p can be reversed by restricted feeding, suggesting a role of peripheral clock in regulating its rhythmicity. Bioinformatics analysis indicated that aryl hydrocarbon receptor nuclear translocator-like (also known as Bmal1) may be a direct target of miR-27b-3p. Luciferase reporter assay showed that miR-27b-3p suppressed Bmal1 3' UTR activity in a dose-dependent manner, and mutagenesis of their binding site abolished this suppression. Furthermore, overexpression of miR-27b-3p dose-dependently reduced the protein expression levels of BMAL1 and impaired the endogenous BMAL1 and gluconeogenic protein rhythmicity. Collectively, our results suggest that miR-27b-3p plays an important role in the posttranscriptional regulation of BMAL1 protein in the liver. MiR-27b-3p may serve as a novel node to integrate the circadian clock and energy metabolism.-Zhang, W., Wang, P., Chen, S., Zhang, Z., Liang, T., Liu, C. Rhythmic expression of miR-27b-3p targets the clock gene Bmal1 at the posttranscriptional level in the mouse liver.
Collapse
Affiliation(s)
- Wenxiang Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Peng Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Siyu Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chang Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
48
|
Jager J, Wang F, Fang B, Lim HW, Peed LC, Steger DJ, Won KJ, Kharitonenkov A, Adams AC, Lazar MA. The Nuclear Receptor Rev-erbα Regulates Adipose Tissue-specific FGF21 Signaling. J Biol Chem 2016; 291:10867-75. [PMID: 27002153 DOI: 10.1074/jbc.m116.719120] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 01/14/2023] Open
Abstract
FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified βKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of βKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.
Collapse
Affiliation(s)
- Jennifer Jager
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Fenfen Wang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Bin Fang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hee-Woong Lim
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Lindsey C Peed
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David J Steger
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kyoung-Jae Won
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Alexei Kharitonenkov
- the Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, and
| | - Andrew C Adams
- the Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Mitchell A Lazar
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,
| |
Collapse
|
49
|
Mang GM, La Spada F, Emmenegger Y, Chappuis S, Ripperger JA, Albrecht U, Franken P. Altered Sleep Homeostasis in Rev-erbα Knockout Mice. Sleep 2016; 39:589-601. [PMID: 26564124 PMCID: PMC4763348 DOI: 10.5665/sleep.5534] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/26/2015] [Indexed: 02/06/2023] Open
Abstract
STUDY OBJECTIVES The nuclear receptor REV-ERBα is a potent, constitutive transcriptional repressor critical for the regulation of key circadian and metabolic genes. Recently, REV-ERBα's involvement in learning, neurogenesis, mood, and dopamine turnover was demonstrated suggesting a specific role in central nervous system functioning. We have previously shown that the brain expression of several core clock genes, including Rev-erbα, is modulated by sleep loss. We here test the consequences of a loss of REV-ERBα on the homeostatic regulation of sleep. METHODS EEG/EMG signals were recorded in Rev-erbα knockout (KO) mice and their wild type (WT) littermates during baseline, sleep deprivation, and recovery. Cortical gene expression measurements after sleep deprivation were contrasted to baseline. RESULTS Although baseline sleep/wake duration was remarkably similar, KO mice showed an advance of the sleep/wake distribution relative to the light-dark cycle. After sleep onset in baseline and after sleep deprivation, both EEG delta power (1-4 Hz) and sleep consolidation were reduced in KO mice indicating a slower increase of homeostatic sleep need during wakefulness. This slower increase might relate to the smaller increase in theta and gamma power observed in the waking EEG prior to sleep onset under both conditions. Indeed, the increased theta activity during wakefulness predicted delta power in subsequent NREM sleep. Lack of Rev-erbα increased Bmal1, Npas2, Clock, and Fabp7 expression, confirming the direct regulation of these genes by REV-ERBα also in the brain. CONCLUSIONS Our results add further proof to the notion that clock genes are involved in sleep homeostasis. Because accumulating evidence directly links REV-ERBα to dopamine signaling the altered homeostatic regulation of sleep reported here are discussed in that context.
Collapse
Affiliation(s)
- Géraldine M. Mang
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Francesco La Spada
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Yann Emmenegger
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Sylvie Chappuis
- Faculty of Science, Department of Biology, Biochemistry, University of Fribourg, Fribourg, Switzerland
| | - Jürgen A. Ripperger
- Faculty of Science, Department of Biology, Biochemistry, University of Fribourg, Fribourg, Switzerland
| | - Urs Albrecht
- Faculty of Science, Department of Biology, Biochemistry, University of Fribourg, Fribourg, Switzerland
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
50
|
Mao S, Ren X, Zhang J. The emerging role of fibroblast growth factor 21 in diabetic nephropathy. J Recept Signal Transduct Res 2016; 36:586-592. [PMID: 26915669 DOI: 10.3109/10799893.2016.1147582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetic nephropathy (DN), an important cause of end-stage renal diseases, brings about great social and economic burden. Due to the variable pathological changes and clinical course, the prognosis of DN is very difficult to predict. DN is also usually associated with enhanced genomic damage and cellular injury. Fibroblast growth factor 21 (FGF21), a nutritionally regulated hormone secreted mainly by the liver, plays a critical role in metabolism. Administration of FGF21 decreases blood glucose, triglyceride, and cholesterol levels, and improves insulin sensitivity, which is closely associated with the development and progression of glomerular diseases. In addition, FGF21 level was associated with renal function. However, the precise role of FGF21 in DN remains unclear. This review will give a comprehensive understanding of the underlying role of FGF21 and its possible interaction with other molecules in DN.
Collapse
Affiliation(s)
- Song Mao
- a Department of Pediatrics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China and
| | - Xianguo Ren
- b Department of Pediatrics , Nanjing Jinling Hospital , Nanjing , China
| | - Jianhua Zhang
- a Department of Pediatrics , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China and
| |
Collapse
|