1
|
Han F, Simeroth S, Zhu J, Gryniuk I, Pranay A, Chen W, Wang Y, Cai Y, Shen Z, Wang G, Griffin CT, Xia L, Yu P. Lymphatic endothelial mTORC1 instructs metabolic and developmental signaling during lymphangiogenesis. Dev Cell 2025:S1534-5807(25)00250-3. [PMID: 40339577 DOI: 10.1016/j.devcel.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 11/10/2024] [Accepted: 04/16/2025] [Indexed: 05/10/2025]
Abstract
The lymphatic vasculature comprises lymphatic capillaries and collecting vessels. To support lymphatic development, lymphatic endothelial cells (LECs) utilize nutrients to fuel lymphangiogenic processes. Meanwhile, LECs maintain constant prospero homeobox 1 (PROX1) expression critical for lymphatic specification. However, molecular mechanisms orchestrating nutrient metabolism while sustaining PROX1 levels in LECs remain unclear. Here, we show that loss of RAPTOR, an indispensable mechanistic target of rapamycin complex 1 (mTORC1) component, downregulates PROX1 and impairs lymphatic capillary growth and differentiation of collecting lymphatics in mice. Mechanistically, mTORC1 inhibition in mouse and human LECs causes Myc reduction, which decreases hexokinase 2 (HK2) and glutaminase (GLS), inhibiting glycolysis and glutaminolysis. Myc or HK2/GLS ablation impedes lymphatic capillary and collecting vessel formation. Interestingly, mTORC1 regulation of PROX1 is independent of Myc-HK2/GLS signaling. Moreover, genetic interaction analysis indicates that Myc and PROX1 play crucial roles in mTORC1-regulated lymphatic development. Collectively, our findings identify mTORC1 as a key regulator of metabolic programs and PROX1 expression during lymphangiogenesis.
Collapse
Affiliation(s)
- Fei Han
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Summer Simeroth
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jie Zhu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Irma Gryniuk
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Weiqing Chen
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA; Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yuan Wang
- Department of Radiation Oncology, Rutgers Cancer Institute and Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Yuanyuan Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Zhiyuan Shen
- Department of Radiation Oncology, Rutgers Cancer Institute and Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA; Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA; Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pengchun Yu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
2
|
Yue Z, Yang Y, Nie L, Sun Y, Wang Q, Lin Y, Gao Y, Cai X. A Binary siRNA-Loaded Tetrahedral DNA Nanobox for Synergetic Anti-Aging Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408323. [PMID: 39690794 DOI: 10.1002/smll.202408323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Indexed: 12/19/2024]
Abstract
Extensive accumulation of senescent cells contributes to organismal aging, and slowing down the process of cellular senescence may ameliorate age-related pathologies. Targeted inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) is found to suppress the conversion of cells to senescence. The regulatory-associated protein of mTOR (Raptor), a key component of mTORC1, has been implicated as important in the aging process, and its druggability deserves to be investigated. Due to high efficiency and high convenience in drug construction, siRNA shows great potential in silencing Raptor expression via RNA interfering therapy. Here, we developed a functionalized anti-aging nanoplatform based on tetrahedral DNA nanostructures (TDNs) encapsulating siRNA targeting Raptor for synergistic anti-aging therapy, named siR-TDNbox. Anti-inflammatory and antioxidant properties of TDN beneficially attenuate age-associated inflammation while serving as siRNA nanocarrier, and thus play a binary role. The results suggest that the siR-TDNbox binary therapeutic nanoplatform has demonstrated an excellent ability to delay aging, inhibit mTORC1 signaling, and extend lifespan. This anti-aging nanoplatform may provide a medium for the combined application of traditional senotherapeutic drugs and promote the upgrading of nanomaterials with anti-aging effects.
Collapse
Affiliation(s)
- Ziqi Yue
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichen Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuezhang Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Uchiyama LF, Nguyen A, Qian K, Cui L, Pham KT, Xiao X, Gao Y, Shimanaka Y, Tol MJ, Vergnes L, Reue K, Tontonoz P. PPARα regulates ER-lipid droplet protein Calsyntenin-3β to promote ketogenesis in hepatocytes. Proc Natl Acad Sci U S A 2025; 122:e2426338122. [PMID: 40258152 PMCID: PMC12054784 DOI: 10.1073/pnas.2426338122] [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: 12/17/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Ketogenesis requires fatty acid flux from intracellular (lipid droplets) and extrahepatic (adipose tissue) lipid stores to hepatocyte mitochondria. However, whether interorganelle contact sites regulate this process is unknown. Recent studies have revealed a role for Calsyntenin-3β (CLSTN3β), an endoplasmic reticulum-lipid droplet contact site protein, in the control of lipid utilization in adipose tissue. Here, we show that Clstn3b expression is induced in the liver by the nuclear receptor PPARα in settings of high lipid utilization, including fasting and ketogenic diet feeding. Hepatocyte-specific loss of CLSTN3β in mice impairs ketogenesis independent of changes in PPARα activation. Conversely, hepatic overexpression of CLSTN3β promotes ketogenesis in mice. Mechanistically, CLSTN3β affects LD-mitochondria crosstalk, as evidenced by changes in fatty acid oxidation, lipid-dependent mitochondrial respiration, and the mitochondrial integrated stress response. These findings define a function for CLSTN3β-dependent membrane contacts in hepatic lipid utilization and ketogenesis.
Collapse
Affiliation(s)
- Lauren F. Uchiyama
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - Alexander Nguyen
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Kevin Qian
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - Liujuan Cui
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
| | - Khoi T. Pham
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
| | - Yajing Gao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
| | - Yuta Shimanaka
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
| | - Marcus J. Tol
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, CA90095
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, CA90095
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Biological Chemistry, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| |
Collapse
|
4
|
Matawali A, Yeap JW, Sulaiman SF, Tan ML. The effects of ketone bodies and ketogenesis on the PI3K/AKT/mTOR signaling pathway: A systematic review. Nutr Res 2025; 139:16-49. [PMID: 40381609 DOI: 10.1016/j.nutres.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Ketogenesis and the PI3K/AKT/mTOR pathway are linked to metabolic imbalance and disease progression. While ketone metabolism and mTOR inhibition are mechanistically connected, their functional relationship across disease models remains unclear. Although ketogenesis can be induced by ketone ingestion, ketogenic diet, or fasting, their individual effects on this pathway require further clarification. This study systematically reviews the relationship between ketogenesis and PI3K/AKT/mTOR signaling, following PRISMA guidelines across 3 databases. Eligible studies that met the selection criteria were evaluated using the risk of bias tools. In most studies involving the ketogenic diet or ketone bodies, suppression of the signaling pathway may lead to positive outcomes in terms of survival rate, lifespan, improved metabolic homeostasis, enhanced neurovascular function and suppressed progression of tumors. By contrast, β-hydroxybutyrate supplementation is associated with the up-regulation of AKT and downstream markers. It may exert an anabolic activity by activating the mTOR signaling pathway in muscle atrophy models and is associated with muscle recovery. Although fasting increases p-AKT expression, this may not necessarily indicate activation of the downstream mTOR signaling cascade, as it could result from an insulin response or regulatory feedback mechanisms. Regulation of the mTOR signaling by ketogenesis may be tissue-specific. Inhibition of PI3K/AKT/mTOR in ketogenesis-induced circumstances may justify the importance of a ketogenic-based diet regimen in combating metabolic diseases. However, future studies should consider standardizing factors such as the duration of fasting, timing, composition of the ketogenic diet and target tissues as these factors may affect study outcomes.
Collapse
Affiliation(s)
- Azlinah Matawali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, Malaysia; Preparatory Centre for Science and Technology, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Jia Wen Yeap
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | | | - Mei Lan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, Malaysia; Preparatory Centre for Science and Technology, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia.
| |
Collapse
|
5
|
Régnier M, Polizzi A, Fougeray T, Fougerat A, Perrier P, Anderson K, Lippi Y, Smati S, Lukowicz C, Lasserre F, Fouche E, Huillet M, Rives C, Tramunt B, Naylies C, Garcia G, Rousseau-Bacquié E, Bertrand-Michel J, Canlet C, Chevolleau-Mege S, Debrauwer L, Heymes C, Burcelin R, Levade T, Gourdy P, Wahli W, Blum Y, Gamet-Payrastre L, Ellero-Simatos S, Guillermet-Guibert J, Hawkins P, Stephens L, Postic C, Montagner A, Loiseau N, Guillou H. Liver gene expression and its rewiring in hepatic steatosis are controlled by PI3Kα-dependent hepatocyte signaling. PLoS Biol 2025; 23:e3003112. [PMID: 40228209 PMCID: PMC12021288 DOI: 10.1371/journal.pbio.3003112] [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: 11/02/2024] [Revised: 04/24/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Insulin and other growth factors are key regulators of liver gene expression, including in metabolic diseases. Most of the phosphoinositide 3-kinase (PI3K) activity induced by insulin is considered to be dependent on PI3Kα. We used mice lacking p110α, the catalytic subunit of PI3Kα, to investigate its role in the regulation of liver gene expression in health and in metabolic dysfunction-associated steatotic liver disease (MASLD). The absence of hepatocyte PI3Kα reduced maximal insulin-induced PI3K activity and signaling, promoted glucose intolerance in lean mice and significantly regulated liver gene expression, including insulin-sensitive genes, in ad libitum feeding. Some of the defective regulation of gene expression in response to hepatocyte-restricted insulin receptor deletion was related to PI3Kα signaling. In addition, though PI3Kα deletion in hepatocytes promoted insulin resistance, it was protective against steatotic liver disease in diet-induced obesity. In the absence of hepatocyte PI3Kα, the effect of diet-induced obesity on liver gene expression was significantly altered, with changes in rhythmic gene expression in liver. Altogether, this study highlights the specific role of p110α in the control of liver gene expression in physiology and in the metabolic rewiring that occurs during MASLD.
Collapse
Affiliation(s)
- Marion Régnier
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
| | - Arnaud Polizzi
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Tiffany Fougeray
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Prunelle Perrier
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Karen Anderson
- The Signaling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Yannick Lippi
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Sarra Smati
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Céline Lukowicz
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Frédéric Lasserre
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Edwin Fouche
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Marine Huillet
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Clémence Rives
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Blandine Tramunt
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
- Diabetology Department, CHU de Toulouse, Toulouse, France
| | - Claire Naylies
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Géraldine Garcia
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Elodie Rousseau-Bacquié
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Justine Bertrand-Michel
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
- Metatoul-Lipidomic Facility, MetaboHUB, Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Cécile Canlet
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Sylvie Chevolleau-Mege
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Laurent Debrauwer
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Rémy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Thierry Levade
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm U1037, CNRS U5071, Université de Toulouse, Toulouse, France
- Laboratoire de Biochimie, CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
- Diabetology Department, CHU de Toulouse, Toulouse, France
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Center for Integrative Genomics, Université de Lausanne, Lausanne, Switzerland
| | - Yuna Blum
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305, Rennes, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Julie Guillermet-Guibert
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm U1037, CNRS U5071, Université de Toulouse, Toulouse, France
| | - Phillip Hawkins
- The Signaling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Len Stephens
- The Signaling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Catherine Postic
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR1331, Université de Toulouse, Toulouse, France
| |
Collapse
|
6
|
Li W, Yang T, Wang N, Li B, Meng C, Yu K, Zhou X, Cao R, Cui S. Maladaptive Peripheral Ketogenesis in Schwann Cells Mediated by CB 1R Contributes to Diabetic Neuropathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414547. [PMID: 39887953 PMCID: PMC11967812 DOI: 10.1002/advs.202414547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/08/2025] [Indexed: 02/01/2025]
Abstract
Diabetic peripheral neuropathy (DPN) is the most common complication of diabetes. Although studies have previously investigated metabolic disruptions in the peripheral nervous system (PNS), the exact metabolic mechanisms underlying DPN remain largely unknown. Herein, a specific form of metabolic remodeling involving aberrant ketogenesis within Schwann cells (SCs) in streptozotocin (STZ)-induced type I diabetes mellitus is identified. The PNS adapts poorly to such aberrant ketogenesis, resulting in disrupted energy metabolism, mitochondrial damage, and homeostatic decompensation, ultimately contributing to DPN. Additionally, the maladaptive peripheral ketogenesis is highly dependent on the cannabinoid type-1 receptor (CB1R)-Hmgcs2 axis. Silencing CB1R reprogrammed the metabolism of SCs by blocking maladaptive ketogenesis, resulting in rebalanced energy metabolism, reduced histopathological changes, and improved neuropathic symptoms. Moreover, this metabolic reprogramming can be induced pharmacologically using JD5037, a peripheral CB1R blocker. These findings revealed a new metabolic mechanism underlying DPN, and promoted CB1R as a promising therapeutic target for DPN.
Collapse
Affiliation(s)
- Weizhen Li
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Tuo Yang
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Ningning Wang
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Baolong Li
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Chuikai Meng
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Kaiming Yu
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Xiongyao Zhou
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Rangjuan Cao
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| | - Shusen Cui
- Department of Hand and Foot SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchun130033China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin ProvinceChangchun130033China
| |
Collapse
|
7
|
Saito S, Cao DY, Bernstein EA, Shibata T, Jones AE, Rios A, Hoshi AO, Stotland AB, Nishi EE, Van Eyk JE, Divakaruni A, Khan Z, Bernstein KE. Peroxisome proliferator-activated receptor alpha is an essential factor in enhanced macrophage immune function induced by angiotensin-converting enzyme. Cell Mol Immunol 2025; 22:243-259. [PMID: 39910334 PMCID: PMC11868401 DOI: 10.1038/s41423-025-01257-y] [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: 04/11/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025] Open
Abstract
Increased expression of angiotensin-converting enzyme (ACE) by myeloid lineage cells strongly increases the immune activity of these cells, as observed in ACE10/10 mice, which exhibit a marked increase in antitumor and antibactericidal immunity. We report that peroxisome proliferator-activated receptor alpha (PPARα), a transcription factor that regulates genes critical for lipid metabolism, is a key molecule in the enhanced macrophage function induced by ACE. Here, we used a Cre-LoxP approach with LysM-Cre to create a modified ACE10/10 mouse line in which macrophages continue to generate abundant ACE but in which monocyte and macrophage PPARα expression is selectively suppressed. These mice, termed A10-PPARα-Cre, have significantly increased growth of B16-F10 tumors compared with ACE10/10 mice with Cre expression. PPARα depletion impaired cytokine production and antigen-presenting activity in ACE-expressing macrophages, resulting in reduced tumor antigen-specific CD8+ T-cell generation. Additionally, the elevated bactericidal resistance typical of ACE10/10 mice was significantly reduced in A10-PPARα-Cre mice, such that these mice resembled WT mice in their resistance to methicillin-resistant Staphylococcus aureus (MRSA) infection. THP-1 cells expressing increased ACE (termed THP-1-ACE) constitute a human macrophage model with increased PPARα that shows enhanced cytotoxicity against tumor cells and better phagocytosis and killing of MRSA. RNA silencing of PPARα in THP-1-ACE cells reduced both tumor cell death and bacterial phagocytosis and clearance. In contrast, the in vivo administration of pemafibrate, a specific agonist of PPARα, to WT and A10-PPARα-Cre mice reduced B16-F10 tumor growth by 24.5% and 25.8%, respectively, but pemafibrate reduced tumors by 57.8% in ACE10/10 mice. With pemafibrate, the number of antitumor CD8+ T cells was significantly lower in A10-PPARα-Cre mice than in ACE10/10 mice. We conclude that PPARα is important in the immune system of myeloid cells, including wild-type cells, and that its increased expression by ACE-expressing macrophages in ACE10/10 mice is indispensable for ACE-dependent functional upregulation of macrophages in both mice and human cells.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tomohiro Shibata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anthony E Jones
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amy Rios
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aoi O Hoshi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Graduate School of Comprehensive Human Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aleksandr B Stotland
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erika E Nishi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Physiology, São Paulo School of Medicine, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Jennifer E Van Eyk
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ajit Divakaruni
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zakir Khan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [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: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
9
|
Saunders RA, Allen WE, Pan X, Sandhu J, Lu J, Lau TK, Smolyar K, Sullivan ZA, Dulac C, Weissman JS, Zhuang X. A platform for multimodal in vivo pooled genetic screens reveals regulators of liver function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.624217. [PMID: 39605605 PMCID: PMC11601512 DOI: 10.1101/2024.11.18.624217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Organ function requires coordinated activities of thousands of genes in distinct, spatially organized cell types. Understanding the basis of emergent tissue function requires approaches to dissect the genetic control of diverse cellular and tissue phenotypes in vivo. Here, we develop paired imaging and sequencing methods to construct large-scale, multi-modal genotype-phenotypes maps in tissue with pooled genetic perturbations. Using imaging, we identify genetic perturbations in individual cells while simultaneously measuring their gene expression and subcellular morphology. Using single-cell sequencing, we measure transcriptomic responses to the same genetic perturbations. We apply this approach to study hundreds of genetic perturbations in the mouse liver. Our study reveals regulators of hepatocyte zonation and liver unfolded protein response, as well as distinct pathways that cause hepatocyte steatosis. Our approach enables new ways of interrogating the genetic basis of complex cellular and organismal physiology and provides crucial training data for emerging machine-learning models of cellular function.
Collapse
Affiliation(s)
- Reuben A. Saunders
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
- Present address: Society of Fellows, Harvard University, MA 02138, USA
- These authors contributed equally
| | - William E. Allen
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Society of Fellows, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Present address: Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305; Arc Institute, Palo Alto, CA 94304
- These authors contributed equally
- Lead contact
| | - Xingjie Pan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Lead AI Scientist
| | - Jaspreet Sandhu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jiaqi Lu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas K. Lau
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Karina Smolyar
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Zuri A. Sullivan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Catherine Dulac
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jonathan S. Weissman
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
10
|
Wicks TR, Shalaurova I, Wolska A, Browne RW, Weinstock-Guttman B, Zivadinov R, Remaley AT, Otvos JD, Ramanathan M. Endogenous Ketone Bodies Are Associated with Metabolic Vulnerability and Disability in Multiple Sclerosis. Nutrients 2025; 17:640. [PMID: 40004969 PMCID: PMC11858685 DOI: 10.3390/nu17040640] [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: 01/13/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose: Ketone bodies could be useful biomarkers in multiple sclerosis (MS) because the pathophysiological processes underlying MS disease progression induce metabolic stress. The purpose was to assess the relationships of ketone bodies with biomarkers of metabolic, inflammatory, and oxidative stress in MS. Methods: Blood samples and neurological assessments were obtained from 153 healthy controls (HC), 187 relapsing-remitting (RRMS), and 91 progressive MS (PMS) patients. AcAc, BHB, and acetone were measured using proton nuclear magnetic resonance spectroscopy. Indices of inflammatory vulnerability (IVX), metabolic malnutrition (MMX), and metabolic vulnerability (MVX) were computed from the NMR profiles. Cholesterol, apolipoprotein, lipid peroxidation, and antioxidant profiles were obtained. Regression analysis adjusted for age, sex, body mass index, and HC, RRMS, or PMS disease status. Results: AcAc and BHB levels were greater in MS compared to HC. BHB and ketone bodies were positively associated with disability on the MS Severity Scale and ambulation time. BHB was positively associated with IVX, MMX, and MVX. AcAc was positively associated with MMX and negatively associated with IVX and MVX. Total ketone body concentration was positively associated with MMX and MVX. BHB and AcAc levels were negatively associated with the amino acids alanine, valine, and leucine. Conclusions: Ketone bodies are associated with inflammatory vulnerability, metabolic vulnerability, and ambulatory disability measures in MS.
Collapse
Affiliation(s)
- Taylor R. Wicks
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA;
| | | | - Anna Wolska
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard W. Browne
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA
| | | | - James D. Otvos
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA;
- Department of Neurology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA
| |
Collapse
|
11
|
Tagne AM, Fotio Y, Lee HL, Jung KM, Katz J, Ahmed F, Le J, Bazinet R, Jang C, Piomelli D. Metabolic reprogramming in the spinal cord drives the transition to pain chronicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635746. [PMID: 39975205 PMCID: PMC11838349 DOI: 10.1101/2025.01.30.635746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Acute injuries can progress into painful states that endure long after healing. The mechanism underlying this transition remains unclear, but metabolic adaptations to the bioenergy demands imposed by injury are plausible contributors. Here we show that peripheral injury activates AKT/mTORC1 in afferent segments of the mouse spinal cord, redirecting local core metabolism toward biomass production while simultaneously suppressing autophagy-mediated biomass reclamation. This metabolic shift supports neuroplasticity, but creates a resource bottleneck that depletes critical spinal cord nutrients. Preventing this depletion with a modified diet normalizes biomass generation and autophagy and halts the transition to chronic pain. This effect, observed across multiple pain models, requires activation of the nutrient sensors, sirtuin-1 and AMPK, as well as restoration of autophagy. The findings identify metabolic reprogramming as a key driver of the progression to pain chronicity and point to nutritional and pharmacological interventions that could prevent this progression after surgery or other physical traumas.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Jean Katz
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Richard Bazinet
- Department of Nutritional Sciences, University of Toronto, ON, Canada
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Hernandez CA, Verzeroli C, Roca-Suarez AA, Farca-Luna AJ, Tonon L, Esteban-Fabró R, Pinyol R, Plissonnier ML, Chicherova I, Dubois A, Bellaud P, Seffals M, Turlin B, Fautrel A, Ichim G, Rivoire M, Passot G, Macek-Jilkova Z, Decaens T, Viari A, Testoni B, Rebouissou S, Llovet JM, Zoulim F, Parent R. Hepatocellular carcinoma hosts cholinergic neural cells and tumoral hepatocytes harboring targetable muscarinic receptors. JHEP Rep 2025; 7:101245. [PMID: 39717507 PMCID: PMC11663970 DOI: 10.1016/j.jhepr.2024.101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 12/25/2024] Open
Abstract
Background & Aims Owing to unexplained interpatient variation and treatment failure in hepatocellular carcinoma (HCC), novel therapeutic approaches remain an urgent clinical need. Hepatic neurons, belonging to the autonomic nervous system (ANS), mediate liver/whole body crosstalk. Pathological innervation of the ANS has been identified in cancer, nurturing tumor stroma and conferring stronger carcinogenic properties. Methods We characterized the innervation of liver tumors from the French Liver Biobank, then applied bioinformatics to TCGA (The Cancer Genome Atlas), several other datasets and a European validation cohort, to re-evaluate patient stratification. Cell biology and pharmacology studies were also performed. Results Densely packed nucleated DCX+, synaptophysin+, NeuN+, VAChT+, TH-, CD31-, CD45- clusters, to date undetected, were identified in human HCCs, and independently confirmed by single-cell RNA sequencing data. Using the new concept of a neuronal score, human and rat HCCs displayed tightly netrin-1-associated neural reconfiguration towards cholinergic polarity, which was associated with chronic liver disease progression, cancer onset and many features of aggressive (proliferative class) HCC, including shortened survival. This score was conditioned by tumoral hepatocytes, and predicted sorafenib efficacy in the STORM HCC phase III trial. Conversely, intratumoral adrenergic lymphocytes were enriched in TEMRA and cytotoxic phenotypes. Amongst all cholinergic transcripts, the medically targeted CHRM3 receptor was enriched and associated with pathogenic traits in HCC, as well as poor prognosis in HCC stages 1-2, while its level dropped upon experimental re-differentiation. Its pharmacological inhibition with low concentrations of anticholinergic drugs, but not cholinomimetics, decreased anchorage-independent growth and anoikis, synergized with sorafenib and lenvatinib in HCC class 1 to 3 lines, yet not in primary human hepatocytes, and preserved mature hepatocyte functions. Conclusion These data identify cholinergic processes as instrumental in liver carcinogenesis and support the use of EMA/FDA-approved cholinergic drugs in HCC research. Impact and implications Hepatocellular carcinoma (HCC) care has long been hampered by the enigmatic nature of disease evolution, as well as of response or resistance to treatment. Hepatic neurons are likely the least studied liver cell type and mediate patients singularities from the ANS to the organ in real-time. Cholinergic inputs identified in this study as pathogenic may be targeted with the well charted pharmacopoeia of neurotropic drugs already available, for basic or clinical research purposes, with an expected high level of safety.
Collapse
Affiliation(s)
- Charlotte A. Hernandez
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | - Claire Verzeroli
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | - Armando Andres Roca-Suarez
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | - Abud-José Farca-Luna
- Fondation Synergie Lyon Cancer, Gilles Thomas Bioinformatics Plateform, Centre Léon Bérard, F-69008 Lyon, France
| | - Laurie Tonon
- Fondation Synergie Lyon Cancer, Gilles Thomas Bioinformatics Plateform, Centre Léon Bérard, F-69008 Lyon, France
| | - Roger Esteban-Fabró
- Translational Research in Hepatic Oncology Group, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Roser Pinyol
- Translational Research in Hepatic Oncology Group, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Marie-Laure Plissonnier
- Epigenetics, Microenvironment, and Liver Cancer, U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon – IHU EVEREST, University of Lyon 1, ISPB, CNRS UMR5286, F-69083 Lyon, France, Centre Léon Bérard, Lyon, France
| | - Ievgeniia Chicherova
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | - Anaëlle Dubois
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | | | | | - Bruno Turlin
- H2P2 platform, University of Rennes, Rennes, France
| | | | - Gabriel Ichim
- Cancer Cell Death team – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon, F-69003 Lyon, France, University of Lyon, F-69003 Lyon, University of Lyon 1, ISPB, Lyon, F-69622, France, CNRS UMR5286, F-69083 Lyon, France, Centre Léon Bérard, F-69008 Lyon, France
| | - Michel Rivoire
- Department of Surgical Oncology, Centre Léon Bérard, F-69008 Lyon, France
| | - Guillaume Passot
- Hospices Civils de Lyon, Service of Gastroenterology, F-69600 Oullins, France
| | - Zuzana Macek-Jilkova
- Institute for Advanced Biosciences, Inserm U1209, University of Grenoble-Alpes, F-38700 La Tronche, France
- Service d’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble-Alpes, 38700 La Tronche, France
| | - Thomas Decaens
- Institute for Advanced Biosciences, Inserm U1209, University of Grenoble-Alpes, F-38700 La Tronche, France
- Service d’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble-Alpes, 38700 La Tronche, France
| | - Alain Viari
- Fondation Synergie Lyon Cancer, Gilles Thomas Bioinformatics Plateform, Centre Léon Bérard, F-69008 Lyon, France
| | - Barbara Testoni
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| | - Sandra Rebouissou
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Josep M. Llovet
- Translational Research in Hepatic Oncology Group, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Fabien Zoulim
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
- Hospices Civils de Lyon, Service of Hepato-Gastroenterology, F-69001 Lyon, France
| | - Romain Parent
- Hepatitis Viruses and Pathobiology of Chronic Liver Diseases – LabEx DEVweCAN, Inserm U1052, Cancer Research Centre of Lyon – Hepatology Institute of Lyon F – IHU EVEREST, University of Lyon 1, ISPB, France, CNRS UMR5286, Centre Léon, Lyon, France
| |
Collapse
|
13
|
Kowalczyk M, Lee YJ, Huang WH. TSC2-mTORC1 axis regulates morphogenesis and neurological function of Gli1 + adult-born dentate granule cells. Mol Biol Cell 2025; 36:br1. [PMID: 39602293 PMCID: PMC11742115 DOI: 10.1091/mbc.e24-08-0366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Aberrant adult hippocampal neurogenesis is implicated in neurological and mood disorders associated with dysregulation of the mechanistic target of rapamycin (mTOR). Understanding how the mTOR pathway shapes the functional development of different subpopulations of adult-born hippocampal neural stem cells will enable insight into potential therapeutic pathways for these disorders. Here we study how loss of TSC2, a regulator of mTOR pathway and a causal gene for tuberous sclerosis complex (TSC), affects dentate gyrus granule cell morphogenesis and hippocampal-dependent function. We found that Tsc2KO mice with TSC2 specifically ablated from Gli1+ adult-born neural stem cells showed neuronal hypertrophy, reduced NEUN expression, increased dendritic arborization, premature cellular senescence, and hypervascularization of the dentate gyrus. Neurologically, Tsc2KO mice showed altered exploratory behavior, impaired spatial learning, abnormal contextual recall, and hypersensitivity to kainic acid-induced seizures. Importantly, genetic reduction of Raptor, essential for mTORC1 signaling, rebalanced mTORC1 signaling and mitigated molecular, cellular, and neurological deficits in Tsc2KO mice. This study uncovered functions of TSC2 in Gli1+ adult-born neural stem cells and highlights RAPTOR as a potential therapeutic target for reversing disease features associated with TSC2 mutations.
Collapse
Affiliation(s)
- Max Kowalczyk
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Montréal, Québec H3G 1A3, Canada
| | - Yu-Ju Lee
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Montréal, Québec H3G 1A3, Canada
| | - Wei-Hsiang Huang
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University, Montréal, Québec H3G 1A3, Canada
| |
Collapse
|
14
|
Lee HY, Min KJ. Dietary Restriction and Lipid Metabolism: Unveiling Pathways to Extended Healthspan. Nutrients 2024; 16:4424. [PMID: 39771045 PMCID: PMC11678862 DOI: 10.3390/nu16244424] [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] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Dietary restriction (DR) has been reported to be a significant intervention that influences lipid metabolism and potentially modulates the aging process in a wide range of organisms. Lipid metabolism plays a pivotal role in the regulation of aging and longevity. In this review, we summarize studies on the significant role of lipid metabolism in aging in relation to DR. As a potent intervention to slow down aging, DR has demonstrated promising effects on lipid metabolism, influencing the aging processes across various species. The current review focuses on the relationships among DR-related molecular signaling proteins such as the sirtuins, signaling pathways such as the target of rapamycin and the insulin/insulin-like growth factor (IGF)-1, lipid metabolism, and aging. Furthermore, the review presents research results on diet-associated changes in cell membrane lipids and alterations in lipid metabolism caused by commensal bacteria, highlighting the importance of lipid metabolism in aging. Overall, the review explores the interplay between diet, lipid metabolism, and aging, while presenting untapped areas for further understanding of the aging process.
Collapse
Affiliation(s)
| | - Kyung-Jin Min
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea;
| |
Collapse
|
15
|
Behzadi P, Cuevas RA, Crane A, Wendling AA, Chu CC, Moorhead WJ, Wong R, Brown M, Tamakloe J, Suresh S, Salehi P, Jaffe IZ, Kuipers AL, Lukashova L, Verdelis K, St Hilaire C. Rapamycin Reduces Arterial Mineral Density and Promotes Beneficial Vascular Remodeling in a Murine Model of Severe Medial Arterial Calcification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606196. [PMID: 39149364 PMCID: PMC11326142 DOI: 10.1101/2024.08.01.606196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Peripheral artery disease (PAD) is the narrowing of the arteries that carry blood to the lower extremities. PAD has been traditionally associated with atherosclerosis. However, recent studies have found that medial arterial calcification (MAC) is the primary cause of chronic limb ischemia below the knee. MAC involves calcification of the elastic fibers surrounding smooth muscle cells (SMCs) in arteries. Matrix GLA protein (MGP) inhibits vascular calcification by binding circulating calcium and preventing hydroxyapatite crystal deposition, while also modulating osteogenic signaling by blocking BMP-2 activation of RUNX2. Mgp -/- mice develop severe MAC and die around 8 weeks after birth due to aortic rupture or heart failure. We previously discovered a rare genetic disease Arterial Calcification due to Deficiency in CD73 (ACDC) in which patients present with extensive MAC in their lower extremity arteries. Using a patient-specific induced pluripotent stem cell model we found that rapamycin inhibited calcification. Here we investigated whether rapamycin could reduce MAC in vivo using the Mgp -/- murine model. Mgp +/+ and Mgp -/- mice received 5mg/kg rapamycin or vehicle. Calcification content was assessed via microCT, and vascular morphology and extracellular matrix content assessed histologically. Immunostaining and western blot analysis were used to examine SMC phenotype and extracellular matrix content. Rapamycin prolonged Mgp -/- mice lifespan, decreased mineral density in the arteries, maintained SMC contractile phenotype, and improved vessel structure, however, calcification volume was unchanged. Mgp -/- mice with SMC-specific deletion of Raptor or Rictor, did not recapitulate treatment with rapamycin. These findings suggest rapamycin promotes beneficial vascular remodeling in vessels with MAC.
Collapse
Affiliation(s)
- Parya Behzadi
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rolando A Cuevas
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Crane
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A Wendling
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Claire C Chu
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William J Moorhead
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Wong
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark Brown
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joshua Tamakloe
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Swathi Suresh
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Payam Salehi
- CardioVascular Center, Vascular Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Allison L Kuipers
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lyudmila Lukashova
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Konstantinos Verdelis
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia St Hilaire
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Queiro-Palou A, Jin Y, Jakobsson L. Genetic and pharmacological targeting of mTORC1 in mouse models of arteriovenous malformation expose non-cell autonomous signalling in HHT. Angiogenesis 2024; 28:6. [PMID: 39661206 PMCID: PMC11634917 DOI: 10.1007/s10456-024-09961-5] [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: 09/01/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
Arteriovenous malformations (AVMs) are abnormal high flow shunts between arteries and veins with major negative impact on the cardiovascular system. Inherited loss-of-function (LOF) mutations in endoglin, encoding an endothelial cell (EC) expressed co-receptor for BMP9/10, causes the disease HHT1/Osler-Weber-Rendu, characterized by bleeding and AVMs. Here we observe increased activity of the downstream signalling complex mTORC1 within the retinal vasculature of HHT mouse models. To investigate its importance in AVM biology, concerning subvascular action, cell specificity, signalling strength and kinetics we combine timed genetic and antibody-based models of HHT with genetic mTORC1 inhibition or activation through EC specific deletion of Rptor or Tsc1. Results demonstrate that EC mTORC1 activation is secondary to endoglin LOF and mainly a consequence of systemic effects following AVM. While genetic EC inhibition of mTORC1 only showed tendencies towards reduced AVM severity, EC overactivation counterintuitively reduced it, implying that mTORC1 must be within a certain range to facilitate AVM. Complete inhibition of mTORC1 signalling by rapamycin provided the strongest therapeutic effect, pointing to potential involvement of RAPTOR-independent pathways or AVM-promoting effects of non-ECs in this pathology.
Collapse
Affiliation(s)
- Antonio Queiro-Palou
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Yi Jin
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Lars Jakobsson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden.
| |
Collapse
|
17
|
Mishra A, Sobha D, Patel D, Suresh PS. Intermittent fasting in health and disease. Arch Physiol Biochem 2024; 130:755-767. [PMID: 37828854 DOI: 10.1080/13813455.2023.2268301] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
CONTEXT Intermittent fasting, a new-age dietary concept derived from an age-old tradition, involves repetitive cycles of fasting/calorie restriction and eating. OBJECTIVE We aim to take a deep dive into the biological responses to intermittent fasting, delineate the disease-modifying and cognitive effects of intermittent fasting, and also shed light on the possible side effects. METHODS Numerous in vitro and in vivo studies were reviewed, followed by an in-depth analysis, and compilation of their implications in health and disease. RESULTS Intermittent fasting improves the body's stress tolerance, which is further amplified with exercise. It impacts various pathological conditions like cancer, obesity, diabetes, cardiovascular disease, and neurodegenerative diseases. CONCLUSION During dietary restriction, the human body experiences a metabolic switch due to the depletion of liver glycogen, which promotes a shift towards utilising fatty acids and ketones in the system, thereby significantly impacting adiposity, ageing and the immune response to various diseases.
Collapse
Affiliation(s)
- Anubhav Mishra
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Devika Sobha
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Dimple Patel
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| |
Collapse
|
18
|
Jindal J, Hill J, Harte J, Dunachie SJ, Kronsteiner B. Starvation and infection: The role of sickness-associated anorexia in metabolic adaptation during acute infection. Metabolism 2024; 161:156035. [PMID: 39326837 DOI: 10.1016/j.metabol.2024.156035] [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: 05/17/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Sickness-associated anorexia, the reduction in appetite seen during infection, is a widely conserved and well-recognized symptom of acute infection, yet there is very little understanding of its functional role in recovery. Anorexic sickness behaviours can be understood as an evolutionary strategy to increase tolerance to pathogen-mediated illness. In this review we explore the evidence for mechanisms and potential metabolic benefits of sickness-associated anorexia. Energy intake can impact on the immune response, control of inflammation and tissue stress, and on pathogen fitness. Fasting mediators including hormone-sensitive lipase, peroxisome proliferator-activated receptor-alpha (PPAR-α) and ketone bodies are potential facilitators of infection recovery through multiple pathways including suppression of inflammation, adaptation to lipid utilising pathways, and resistance to pathogen-induced cellular stress. However, the effect and benefit of calorie restriction is highly heterogeneous depending on both the infection and the metabolic status of the host, which has implications regarding clinical recommendations for feeding during different infections.
Collapse
Affiliation(s)
- Jessy Jindal
- The Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Jennifer Hill
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Jodie Harte
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.
| | - Barbara Kronsteiner
- NDM Centre for Global Health Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Kaade E, Mausbach S, Erps N, Sylvester M, Shakeri F, Jachimowicz RD, Gieselmann V, Thelen M. Starvation-induced metabolic rewiring affects mTORC1 composition in vivo. Sci Rep 2024; 14:28296. [PMID: 39550382 PMCID: PMC11569187 DOI: 10.1038/s41598-024-78873-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Lysosomes play a crucial role in metabolic adaptation to starvation, but detailed in vivo studies are scarce. Therefore, we investigated the changes of the proteome of liver lysosomes in mice starved short-term for 6h or long-term for 24h. We verified starvation-induced catabolism by weight loss, ketone body production, drop in blood glucose and an increase of 3-methylhistidine. Deactivation of mTORC1 in vivo after short-term starvation causes a depletion of mTORC1 and the associated Ragulator complex in hepatic lysosomes, resulting in diminished phosphorylation of mTORC1 target proteins. While mTORC1 lysosomal protein levels and activity in liver were restored after long-term starvation, the lysosomal levels of Ragulator remained constantly reduced. To determine whether this mTORC1 activity pattern may be organ-specific, we further investigated the key metabolic organs muscle and brain. mTORC1 inactivation, but not re-activation, occurred in muscle after a starvation of 12 h or longer. In brain, mTORC1 activity remained unchanged during starvation. As mTORC1 deactivation is known to induce autophagy, we further investigated the more than 150 non-lysosomal proteins enriched in the lysosomal fraction upon starvation. Proteasomal, cytosolic and peroxisomal proteins dominated after short-term starvation, while after long-term starvation, mainly proteasomal and mitochondrial proteins accumulated, indicating ordered autophagic protein degradation.
Collapse
Affiliation(s)
- Edgar Kaade
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Simone Mausbach
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Nina Erps
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany
| | - Marc Sylvester
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
- Core Facility Analytical Proteomics, Medical Faculty , Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Farhad Shakeri
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Ron D Jachimowicz
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Volkmar Gieselmann
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Melanie Thelen
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany.
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany.
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany.
| |
Collapse
|
20
|
Vaezi MA, Nekoufar S, Robati AK, Salimi V, Tavakoli-Yaraki M. Therapeutic potential of β-hydroxybutyrate in the management of pancreatic neoplasms: exploring novel diagnostic and treatment strategies. Lipids Health Dis 2024; 23:376. [PMID: 39543582 PMCID: PMC11562866 DOI: 10.1186/s12944-024-02368-7] [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: 08/22/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Pancreatic neoplasm, a highly aggressive and often fatal cancer, poses challenges due to late detection and nonspecific symptoms. Therefore, both early diagnosis and appropriate therapeutic approaches are necessary to augment the condition of these patients. Cancer cells undergo metabolic deregulation, which enables their proliferation, survival, and invasion. As a result, it is crucial to focus on the metabolic pathways in prevalent cancers and explore treatment strategies that target these pathways to control tumor growth effectively. This is particularly relevant in cancers like pancreatic cancer, which undergo numerous metabolic alterations. The ketogenic regimen, characterized by low carbohydrate and protein contents and high-fat sources, does not involve caloric restriction. This allows for the induction of ketogenesis and an increase in ketone bodies, while insulin and glucose levels remain low even after meals. This unique metabolic state may influence the tumor microenvironment. Given the lack of unanimous agreement on the precise role and mechanism of the ketogenic diet, this review aims to clarify the diagnostic value and accuracy of ketone bodies in various types of pancreatic tumors and explore the potential anti-cancer effects of the ketogenic diet when used alone or in conjunction with chemotherapy, also to determine the potential of the ketogenic diet to be used as adjuvant therapy. The outcomes of this study are instrumental in enhancing our understanding of the benefits and drawbacks associated with employing this diet for the management and diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Mohammad Amin Vaezi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Samira Nekoufar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Ali Karami Robati
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran.
- Finetech in Medicine Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Fougerat A, Bruse J, Polizzi A, Montagner A, Guillou H, Wahli W. Lipid sensing by PPARα: Role in controlling hepatocyte gene regulatory networks and the metabolic response to fasting. Prog Lipid Res 2024; 96:101303. [PMID: 39521352 DOI: 10.1016/j.plipres.2024.101303] [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: 07/17/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Peroxisome proliferator-activated receptors (PPARs) constitute a small family of three nuclear receptors that act as lipid sensors, and thereby regulate the transcription of genes having key roles in hepatic and whole-body energy homeostasis, and in other processes (e.g., inflammation), which have far-reaching health consequences. Peroxisome proliferator-activated receptor isotype α (PPARα) is expressed in oxidative tissues, particularly in the liver, carrying out critical functions during the adaptive fasting response. Advanced omics technologies have provided insight into the vast complexity of the regulation of PPAR expression and activity, as well as their downstream effects on the physiology of the liver and its associated metabolic organs. Here, we provide an overview of the gene regulatory networks controlled by PPARα in the liver in response to fasting. We discuss impacts on liver metabolism, the systemic repercussions and benefits of PPARα-regulated ketogenesis and production of fibroblast growth factor 21 (FGF21), a fasting- and stress-inducible metabolic hormone. We also highlight current challenges in using novel methods to further improve our knowledge of PPARα in health and disease.
Collapse
Affiliation(s)
- Anne Fougerat
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France.
| | - Justine Bruse
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Alexandra Montagner
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM UMR1297, Toulouse III University, University Paul Sabatier (UPS), Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France
| | - Walter Wahli
- Toxalim (Research Centre in Toxicology), INRAE, ENVT, INP-Purpan, UPS, Toulouse University, Toulouse, France; Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
22
|
Elmansi AM, Miller RA. Oxidative phosphorylation and fatty acid oxidation in slow-aging mice. Free Radic Biol Med 2024; 224:246-255. [PMID: 39153667 DOI: 10.1016/j.freeradbiomed.2024.08.018] [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: 03/25/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Oxidative metabolism declines with aging in humans leading to multiple metabolic ailments and subsequent inflammation. In mice, there is evidence of age-related suppression of fatty acid oxidation and oxidative phosphorylation in the liver, heart, and muscles. Many interventions that extend healthy lifespan of mice have been developed, including genetic, pharmacological, and dietary interventions. In this article, we review the literature on oxidative metabolism changes in response to those interventions. We also discuss the molecular pathways that mediate those changes, and their potential as targets for future longevity interventions.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA; University of Michigan Geriatrics Center, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Fulghum K, Salathe SF, Davis X, Thyfault JP, Puchalska P, Crawford PA. Ketone body metabolism and cardiometabolic implications for cognitive health. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:29. [PMID: 40093558 PMCID: PMC11908690 DOI: 10.1038/s44324-024-00029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/05/2024] [Indexed: 03/19/2025]
Abstract
Cardiometabolic complications of obesity present a growing public health concern and are associated with poor outcomes, mediated in part by an increased risk for cardiovascular disease, metabolic dysfunction-associated fatty liver disease, and systemic insulin resistance. Recent studies support that both insulin resistance and obesity are also associated with aberrant brain metabolism and cognitive impairment similar to what is observed in neurodegenerative diseases. Central to these pathological outcomes are adverse changes in tissue glucose and ketone body metabolism, suggesting that regulation of substrate utilization could be a mechanistic link between the cardiometabolic outcomes of obesity and the progression of cognitive decline. Here, we review ketone body metabolism in physiological and pathological conditions with an emphasis on the therapeutic potential of ketone bodies in treating cardiometabolic diseases and neurodegenerative diseases that lead to cognitive decline. We highlight recent findings in the associations among cardiometabolic disease, ketone body metabolism, and cognitive health while providing a theoretical framework by which ketone bodies may promote positive health outcomes and preserve cognitive function.
Collapse
Affiliation(s)
- Kyle Fulghum
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sebastian F. Salathe
- Departments of Cell Biology and Physiology and Internal Medicine – Division of Endocrinology and Metabolism, Kansas University Medical Center, Kansas City, KS, USA
| | - Xin Davis
- Departments of Cell Biology and Physiology and Internal Medicine – Division of Endocrinology and Metabolism, Kansas University Medical Center, Kansas City, KS, USA
| | - John P. Thyfault
- Departments of Cell Biology and Physiology and Internal Medicine – Division of Endocrinology and Metabolism, Kansas University Medical Center, Kansas City, KS, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
24
|
Hwang D, Baek S, Chang J, Seol T, Ku B, Ha H, Lee H, Cho S, Roh TY, Kim YK, Lim DS. YAP promotes global mRNA translation to fuel oncogenic growth despite starvation. Exp Mol Med 2024; 56:2202-2215. [PMID: 39349825 PMCID: PMC11542038 DOI: 10.1038/s12276-024-01316-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 11/08/2024] Open
Abstract
Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play fundamental roles in stem/progenitor cell expansion during homeostasis, and their dysregulation often leads to tissue overgrowth. Here, we show that YAP activation is sufficient to overcome the restriction of global protein synthesis induced by serum starvation, enabling cells to sustain proliferation and survival despite an unfavorable environment. Mechanistically, YAP/TAZ selectively promoted the mTORC1-dependent translation of mRNAs containing 5' terminal oligopyrimidine (5'TOP) motifs, ultimately increasing the cellular polysome content. Interestingly, DNA damage-inducible transcript 4 (DDIT4), a negative regulator of mTORC1, was upregulated by serum starvation but repressed by YAP/TAZ. DDIT4 was sufficient to suppress the translation and transformative potential of uveal melanoma cells, which are often serum unresponsive due to G protein mutations. Our findings reveal a vital role for protein synthesis as a key modality of YAP/TAZ-induced oncogenic transformation and indicate the potential for targeting mTORC1 or translation to treat YAP/TAZ-driven malignancies.
Collapse
Affiliation(s)
- Daehee Hwang
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seonguk Baek
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jeeyoon Chang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Taejun Seol
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Bomin Ku
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hongseok Ha
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Hyeonji Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Suhyeon Cho
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Tae-Young Roh
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Yoon Ki Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dae-Sik Lim
- National Creative Research Initiatives Center for Cell Plasticity, KAIST Stem Cell Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
25
|
Ruan M, Xu F, Li N, Yu J, Teng F, Tang J, Huang C, Zhu H. Free long-chain fatty acids trigger early postembryonic development in starved Caenorhabditis elegans by suppressing mTORC1. PLoS Biol 2024; 22:e3002841. [PMID: 39436954 PMCID: PMC11530034 DOI: 10.1371/journal.pbio.3002841] [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: 11/09/2023] [Revised: 11/01/2024] [Accepted: 09/14/2024] [Indexed: 10/25/2024] Open
Abstract
Postembryonic development of animals has long been considered an internally predetermined program, while macronutrients were believed to be essential solely for providing biomatters and energy to support this process. However, in this study, by using a nematode Caenorhabditis elegans (abbreviated as C. elegans hereafter) model, we surprisingly discovered that dietary supplementation of palmitic acid alone, rather than other abundant essential nutrients such as glucose or amino acid mixture, was sufficient to initiate early postembryonic development even under complete macronutrient deprivation. Such a development was evidenced by changes in morphology, cellular markers in multiple tissues, behaviors, and the global transcription pattern and it occurred earlier than the well-known early L1 nutrient checkpoint. Mechanistically, palmitic acid did not function as a biomatter/energy provider, but rather as a ligand to activate the nuclear hormone receptor NHR-49/80, leading to the production of an unknown peroxisome-derived secretive hormone in the intestine. This hormonal signal was received by chemosensory neurons in the head, regulating the insulin-like neuropeptide secretion and its downstream nuclear receptor to orchestrate global development. Additionally, the nutrient-sensing hub mTORC1 played a negative role in this process. In conclusion, our data indicate that free fatty acids act as a primary nutrient signal to launch the early development in C. elegans, which suggests that specific nutrients, rather than the internal genetic program, serve as the first impetus for postembryonic development.
Collapse
Affiliation(s)
- Meiyu Ruan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Fan Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Na Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing Yu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fukang Teng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiawei Tang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huanhu Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
26
|
Nord C, Jones I, Garcia-Maestre M, Hägglund AC, Carlsson L. Reduced mTORC1-signaling in progenitor cells leads to retinal lamination deficits. Dev Dyn 2024; 253:922-939. [PMID: 38546215 DOI: 10.1002/dvdy.707] [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: 09/20/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 10/02/2024] Open
Abstract
BACKGROUND Neuronal lamination is a hallmark of the mammalian central nervous system (CNS) and underlies connectivity and function. Initial formation of this tissue architecture involves the integration of various signaling pathways that regulate the differentiation and migration of neural progenitor cells. RESULTS Here, we demonstrate that mTORC1 mediates critical roles during neuronal lamination using the mouse retina as a model system. Down-regulation of mTORC1-signaling in retinal progenitor cells by conditional deletion of Rptor led to decreases in proliferation and increased apoptosis during embryogenesis. These developmental deficits preceded aberrant lamination in adult animals which was best exemplified by the fusion of the outer and inner nuclear layer and the absence of an outer plexiform layer. Moreover, ganglion cell axons originating from each Rptor-ablated retina appeared to segregate to an equal degree at the optic chiasm with both contralateral and ipsilateral projections displaying overlapping termination topographies within several retinorecipient nuclei. In combination, these visual pathway defects led to visually mediated behavioral deficits. CONCLUSIONS This study establishes a critical role for mTORC1-signaling during retinal lamination and demonstrates that this pathway regulates diverse developmental mechanisms involved in driving the stratified arrangement of neurons during CNS development.
Collapse
Affiliation(s)
- Christoffer Nord
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | - Iwan Jones
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | | | | | - Leif Carlsson
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| |
Collapse
|
27
|
Zheng K, Qian Y, Wang H, Song D, You H, Hou B, Han F, Zhu Y, Feng F, Lam SM, Shui G, Li X. Withdrawn: Combinatorial lipidomics and proteomics underscore erythrocyte lipid membrane aberrations in the development of adverse cardio-cerebrovascular complications in maintenance hemodialysis patients. Redox Biol 2024; 76:103295. [PMID: 39159596 PMCID: PMC11378344 DOI: 10.1016/j.redox.2024.103295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/21/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
This article has been withdrawn: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/policies/article-withdrawal). The authors reached out to the Publisher to alert the Publisher to incorrect text published in the article. After investigating the situation, the journal came to the conclusion that the wrong version of the file was sent by the authors to the production team during the proof stage and the misplaced text was not noticed by the authors when they approved the final version. After consulting with the Editor-in-Chief of the journal, the decision was made to withdraw the current version of the article.
Collapse
Affiliation(s)
- Ke Zheng
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujun Qian
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China; Department of Nephrology, Jiangsu Province Hospital/The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyun Wang
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dan Song
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Xuemei Li
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
28
|
Pak HH, Grossberg AN, Sanderfoot RR, Babygirija R, Green CL, Koller M, Dzieciatkowska M, Paredes DA, Lamming DW. Non-canonical metabolic and molecular effects of calorie restriction are revealed by varying temporal conditions. Cell Rep 2024; 43:114663. [PMID: 39167490 PMCID: PMC11427179 DOI: 10.1016/j.celrep.2024.114663] [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: 02/21/2024] [Revised: 06/27/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Calorie restriction (CR) extends lifespan and healthspan in diverse species. Comparing ad libitum- and CR-fed mice is challenging due to their significantly different feeding patterns, with CR-fed mice consuming their daily meal in 2 h and then subjecting themselves to a prolonged daily fast. Here, we examine how ad libitum- and CR-fed mice respond to tests performed at various times and fasting durations and find that the effects of CR-insulin sensitivity, circulating metabolite levels, and mechanistic target of rapamycin 1 (mTORC1) activity-result from the specific temporal conditions chosen, with CR-induced improvements in insulin sensitivity observed only after a prolonged fast, and the observed differences in mTORC1 activity between ad libitum- and CR-fed mice dependent upon both fasting duration and the specific tissue examined. Our results demonstrate that much of our understanding of the effects of CR are related to when, relative to feeding, we choose to examine the mice.
Collapse
Affiliation(s)
- Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Allison N Grossberg
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA; Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Rachel R Sanderfoot
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mikaela Koller
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Daniel A Paredes
- Department of Biological Sciences, University of Denver, Denver, CO, USA; Department of Electrical and Computer Engineering, University of Denver, Denver, CO, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
29
|
Cormerais Y, Lapp SC, Kalafut KC, Cissé MY, Shin J, Stefadu B, Personnaz J, Schrotter S, D’Amore A, Martin ER, Salussolia CL, Sahin M, Menon S, Byles V, Manning BD. AKT-mediated phosphorylation of TSC2 controls stimulus- and tissue-specific mTORC1 signaling and organ growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614519. [PMID: 39386441 PMCID: PMC11463511 DOI: 10.1101/2024.09.23.614519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) integrates diverse intracellular and extracellular growth signals to regulate cell and tissue growth. How the molecular mechanisms regulating mTORC1 signaling established through biochemical and cell biological studies function under physiological states in specific mammalian tissues are unknown. Here, we characterize a genetic mouse model lacking the 5 phosphorylation sites on the tuberous sclerosis complex 2 (TSC2) protein through which the growth factor-stimulated protein kinase AKT can active mTORC1 signaling in cell culture models. These phospho-mutant mice (TSC2-5A) are developmentally normal but exhibit reduced body weight and the weight of specific organs, such as brain and skeletal muscle, associated with cell intrinsic decreases in growth factor-stimulated mTORC1 signaling. The TSC2-5A mouse model demonstrates that TSC2 phosphorylation is a primary mechanism of mTORC1 activation in some, but not all, tissues and provides a genetic tool to facilitate studies on the physiological regulation of mTORC1.
Collapse
Affiliation(s)
- Yann Cormerais
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Samuel C. Lapp
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Krystle C. Kalafut
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Madi Y. Cissé
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Jong Shin
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Benjamin Stefadu
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jean Personnaz
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Sandra Schrotter
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Cell Signaling Technologies, Inc, Beverly, MA, 01915, USA
| | - Angelica D’Amore
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Emma R. Martin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Catherine L. Salussolia
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Suchithra Menon
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Vanessa Byles
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Brendan D. Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
30
|
Bilkei-Gorzo A, Schurmann B, Schneider M, Kraemer M, Nidadavolu P, Beins EC, Müller CE, Dvir-Ginzberg M, Zimmer A. Bidirectional Effect of Long-Term Δ 9-Tetrahydrocannabinol Treatment on mTOR Activity and Metabolome. ACS Pharmacol Transl Sci 2024; 7:2637-2649. [PMID: 39296258 PMCID: PMC11406684 DOI: 10.1021/acsptsci.4c00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/15/2024] [Accepted: 06/18/2024] [Indexed: 09/21/2024]
Abstract
Brain aging is associated with cognitive decline, reduced synaptic plasticity, and altered metabolism. The activity of mechanistic target of rapamycin (mTOR) has a major impact on aging by regulating cellular metabolism. Although reduced mTOR signaling has a general antiaging effect, it can negatively affect the aging brain by reducing synaptogenesis and thus cognitive functions. Increased mTOR activity facilitates aging and is responsible for the amnestic effect of the cannabinoid receptor 1 agonist Δ9-tetrahydrocannabinol (THC) in higher doses. Long-term low-dose Δ9-THC had an antiaging effect on the brain by restoring cognitive abilities and synapse densities in old mice. Whether changes in mTOR signaling and metabolome are associated with its positive effects on the aging brain is an open question. Here, we show that Δ9-THC treatment has a tissue-dependent and dual effect on mTOR signaling and the metabolome. In the brain, Δ9-THC treatment induced a transient increase in mTOR activity and in the levels of amino acids and metabolites involved in energy production, followed by an increased synthesis of synaptic proteins. Unexpectedly, we found a similar reduction in the mTOR activity in adipose tissue and in the level of amino acids and carbohydrate metabolites in blood plasma as in animals on a low-calorie diet. Thus, long-term Δ9-THC treatment first increases the level of energy and synaptic protein production in the brain, followed by a reduction in mTOR activity and metabolic processes in the periphery. Our study suggests that a dual effect on mTOR activity and the metabolome could be the basis for an effective antiaging and pro-cognitive medication.
Collapse
Affiliation(s)
- Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Britta Schurmann
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Marion Schneider
- Pharmaceutical Institute, University of Bonn, Bonn 53121, Germany
| | - Michael Kraemer
- Institute of Forensic Medicine, Medical Faculty, University of Bonn, Bonn 53111, Germany
| | - Prakash Nidadavolu
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Eva C Beins
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| | - Christa E Müller
- Pharmaceutical Institute, University of Bonn, Bonn 53121, Germany
| | - Mona Dvir-Ginzberg
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn 53125, Germany
| |
Collapse
|
31
|
Yang H, Zingaro VA, Lincoff J, Tom H, Oikawa S, Oses-Prieto JA, Edmondson Q, Seiple I, Shah H, Kajimura S, Burlingame AL, Grabe M, Ruggero D. Remodelling of the translatome controls diet and its impact on tumorigenesis. Nature 2024; 633:189-197. [PMID: 39143206 DOI: 10.1038/s41586-024-07781-7] [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: 04/15/2023] [Accepted: 07/03/2024] [Indexed: 08/16/2024]
Abstract
Fasting is associated with a range of health benefits1-6. How fasting signals elicit changes in the proteome to establish metabolic programmes remains poorly understood. Here we show that hepatocytes selectively remodel the translatome while global translation is paradoxically downregulated during fasting7,8. We discover that phosphorylation of eukaryotic translation initiation factor 4E (P-eIF4E) is induced during fasting. We show that P-eIF4E is responsible for controlling the translation of genes involved in lipid catabolism and the production of ketone bodies. Inhibiting P-eIF4E impairs ketogenesis in response to fasting and a ketogenic diet. P-eIF4E regulates those messenger RNAs through a specific translation regulatory element within their 5' untranslated regions (5' UTRs). Our findings reveal a new signalling property of fatty acids, which are elevated during fasting. We found that fatty acids bind and induce AMP-activated protein kinase (AMPK) kinase activity that in turn enhances the phosphorylation of MAP kinase-interacting protein kinase (MNK), the kinase that phosphorylates eIF4E. The AMPK-MNK-eIF4E axis controls ketogenesis, revealing a new lipid-mediated kinase signalling pathway that links ketogenesis to translation control. Certain types of cancer use ketone bodies as an energy source9,10 that may rely on P-eIF4E. Our findings reveal that on a ketogenic diet, treatment with eFT508 (also known as tomivosertib; a P-eIF4E inhibitor) restrains pancreatic tumour growth. Thus, our findings unveil a new fatty acid-induced signalling pathway that activates selective translation, which underlies ketogenesis and provides a tailored diet intervention therapy for cancer.
Collapse
Affiliation(s)
- Haojun Yang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- School of Medicine and Department of Urology, UCSF, San Francisco, CA, USA
| | - Vincenzo Andrea Zingaro
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- School of Medicine and Department of Urology, UCSF, San Francisco, CA, USA
| | - James Lincoff
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Harrison Tom
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- School of Medicine and Department of Urology, UCSF, San Francisco, CA, USA
| | - Satoshi Oikawa
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Quinn Edmondson
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Ian Seiple
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Hardik Shah
- Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Alma L Burlingame
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
| | - Michael Grabe
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA.
- School of Medicine and Department of Urology, UCSF, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA.
| |
Collapse
|
32
|
Min W, Qin L, Zhang H, López-Giráldez F, Jiang N, Kim Y, Mohan VK, Su M, Murray KN, Grutzendler J, Zhou JH. mTORC1 Signaling in Brain Endothelial Progenitors Contributes to CCM Pathogenesis. Circ Res 2024; 135:e94-e113. [PMID: 38957991 PMCID: PMC11293987 DOI: 10.1161/circresaha.123.324015] [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: 11/18/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Cerebral vascular malformations (CCMs) are primarily found within the brain, where they result in increased risk for stroke, seizures, and focal neurological deficits. The unique feature of the brain vasculature is the blood-brain barrier formed by the brain neurovascular unit. Recent studies suggest that loss of CCM genes causes disruptions of blood-brain barrier integrity as the inciting events for CCM development. CCM lesions are proposed to be initially derived from a single clonal expansion of a subset of angiogenic venous capillary endothelial cells (ECs) and respective resident endothelial progenitor cells (EPCs). However, the critical signaling events in the subclass of brain ECs/EPCs for CCM lesion initiation and progression are unclear. METHODS Brain EC-specific CCM3-deficient (Pdcd10BECKO) mice were generated by crossing Pdcd10fl/fl mice with Mfsd2a-CreERT2 mice. Single-cell RNA-sequencing analyses were performed by the chromium single-cell platform (10× genomics). Cell clusters were annotated into EC subtypes based on visual inspection and GO analyses. Cerebral vessels were visualized by 2-photon in vivo imaging and tissue immunofluorescence analyses. Regulation of mTOR (mechanistic target of rapamycin) signaling by CCM3 and Cav1 (caveolin-1) was performed by cell biology and biochemical approaches. RESULTS Single-cell RNA-sequencing analyses from P10 Pdcd10BECKO mice harboring visible CCM lesions identified upregulated CCM lesion signature and mitotic EC clusters but decreased blood-brain barrier-associated EC clusters. However, a unique EPC cluster with high expression levels of stem cell markers enriched with mTOR signaling was identified from early stages of the P6 Pdcd10BECKO brain. Indeed, mTOR signaling was upregulated in both mouse and human CCM lesions. Genetic deficiency of Raptor (regulatory-associated protein of mTOR), but not of Rictor (rapamycin-insensitive companion of mTOR), prevented CCM lesion formation in the Pdcd10BECKO model. Importantly, the mTORC1 (mTOR complex 1) pharmacological inhibitor rapamycin suppressed EPC proliferation and ameliorated CCM pathogenesis in Pdcd10BECKO mice. Mechanistic studies suggested that Cav1/caveolae increased in CCM3-depleted EPC-mediated intracellular trafficking and complex formation of the mTORC1 signaling proteins. CONCLUSIONS CCM3 is critical for maintaining blood-brain barrier integrity and CCM3 loss-induced mTORC1 signaling in brain EPCs initiates and facilitates CCM pathogenesis.
Collapse
Affiliation(s)
- Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lingfeng Qin
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Francesc López-Giráldez
- Yale Center for Genomic Analysis, Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Ning Jiang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Yeaji Kim
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Varsha K. Mohan
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Minhong Su
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Katie N Murray
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jaime Grutzendler
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jenny Huanjiao Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
33
|
Ortega-Molina A, Lebrero-Fernández C, Sanz A, Calvo-Rubio M, Deleyto-Seldas N, de Prado-Rivas L, Plata-Gómez AB, Fernández-Florido E, González-García P, Vivas-García Y, Sánchez García E, Graña-Castro O, Price NL, Aroca-Crevillén A, Caleiras E, Monleón D, Borrás C, Casanova-Acebes M, de Cabo R, Efeyan A. A mild increase in nutrient signaling to mTORC1 in mice leads to parenchymal damage, myeloid inflammation and shortened lifespan. NATURE AGING 2024; 4:1102-1120. [PMID: 38849535 PMCID: PMC11333293 DOI: 10.1038/s43587-024-00635-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024]
Abstract
The mechanistic target of rapamycin complex 1 controls cellular anabolism in response to growth factor signaling and to nutrient sufficiency signaled through the Rag GTPases. Inhibition of mTOR reproducibly extends longevity across eukaryotes. Here we report that mice that endogenously express active mutant variants of RagC exhibit multiple features of parenchymal damage that include senescence, expression of inflammatory molecules, increased myeloid inflammation with extensive features of inflammaging and a ~30% reduction in lifespan. Through bone marrow transplantation experiments, we show that myeloid cells are abnormally activated by signals emanating from dysfunctional RagC-mutant parenchyma, causing neutrophil extravasation that inflicts additional inflammatory damage. Therapeutic suppression of myeloid inflammation in aged RagC-mutant mice attenuates parenchymal damage and extends survival. Together, our findings link mildly increased nutrient signaling to limited lifespan in mammals, and support a two-component process of parenchymal damage and myeloid inflammation that together precipitate a time-dependent organ deterioration that limits longevity.
Collapse
Affiliation(s)
- Ana Ortega-Molina
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Metabolism in cancer and aging Laboratory, Immune System Development And Function Department, Centro de Biología Molecular Severo Ochoa (CBM), Madrid, Spain.
| | - Cristina Lebrero-Fernández
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Metabolism in cancer and aging Laboratory, Immune System Development And Function Department, Centro de Biología Molecular Severo Ochoa (CBM), Madrid, Spain
| | - Alba Sanz
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Miguel Calvo-Rubio
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Nerea Deleyto-Seldas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Lucía de Prado-Rivas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Fernández-Florido
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Yurena Vivas-García
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Sánchez García
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA-Nemesio Díez), Department of Basic Medical Sciences, School of Medicine, San Pablo-CEU University, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Alejandra Aroca-Crevillén
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniel Monleón
- Department of Pathology, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), MiniAging Research Group, Institute of Health Research-INCLIVA, Valencia, Spain
| | - María Casanova-Acebes
- Cancer Immunity Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
34
|
Sima J, Zhang Y, Farriday D, Ahn AYE, Lopez ER, Jin C, Harrell J, Darmohray D, Silverman D, Dan Y. Restoration of locus coeruleus noradrenergic transmission during sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601820. [PMID: 39005471 PMCID: PMC11244971 DOI: 10.1101/2024.07.03.601820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Sleep is indispensable for health and wellbeing, but its basic function remains elusive. The locus coeruleus (LC) powerfully promotes arousal by releasing noradrenaline. We found that noradrenaline transmission is reduced by prolonged wakefulness and restored during sleep. Fiber-photometry imaging of noradrenaline using its biosensor showed that its release evoked by optogenetic LC neuron activation was strongly attenuated by three hours of sleep deprivation and restored during subsequent sleep. This is accompanied by the reduction and recovery of the wake-promoting effect of the LC neurons. The reduction of both LC evoked noradrenaline release and wake-inducing potency is activity dependent, and the rate of noradrenaline transmission recovery depends on mammalian target of rapamycin (mTOR) signaling. The decline and recovery of noradrenaline transmission also occur in spontaneous sleep-wake cycles on a timescale of minutes. Together, these results reveal an essential role of sleep in restoring transmission of a key arousal-promoting neuromodulator.
Collapse
Affiliation(s)
- Jiao Sima
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Yuchen Zhang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Declan Farriday
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Andy Young-Eon Ahn
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Eduardo Ramirez Lopez
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Chennan Jin
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Jade Harrell
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Dana Darmohray
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Daniel Silverman
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
35
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [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: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
36
|
Shi R, Wang B. Nutrient metabolism in regulating intestinal stem cell homeostasis. Cell Prolif 2024; 57:e13602. [PMID: 38386338 PMCID: PMC11150145 DOI: 10.1111/cpr.13602] [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: 09/28/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/23/2024] Open
Abstract
Intestinal stem cells (ISCs) are known for their remarkable proliferative capacity, making them one of the most active cell populations in the body. However, a high turnover rate of intestinal epithelium raises the likelihood of dysregulated homeostasis, which is known to cause various diseases, including cancer. Maintaining precise control over the homeostasis of ISCs is crucial to preserve the intestinal epithelium's integrity during homeostasis or stressed conditions. Recent research has indicated that nutrients and metabolic pathways can extensively modulate the fate of ISCs. This review will explore recent findings concerning the influence of various nutrients, including lipids, carbohydrates, and vitamin D, on the delicate balance between ISC proliferation and differentiation.
Collapse
Affiliation(s)
- Ruicheng Shi
- Department of Comparative Biosciences, College of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Bo Wang
- Department of Comparative Biosciences, College of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional Sciences, College of Agricultural, Consumer and Environmental SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
37
|
Samson N, Bosoi CR, Roy C, Turcotte L, Tribouillard L, Mouchiroud M, Berthiaume L, Trottier J, Silva HCG, Guerbette T, Plata-Gómez AB, Besse-Patin A, Montoni A, Ilacqua N, Lamothe J, Citron YR, Gélinas Y, Gobeil S, Zoncu R, Caron A, Morissette M, Pellegrini L, Rochette PJ, Estall JL, Efeyan A, Shum M, Audet-Walsh É, Barbier O, Marette A, Laplante M. HSDL2 links nutritional cues to bile acid and cholesterol homeostasis. SCIENCE ADVANCES 2024; 10:eadk9681. [PMID: 38820148 PMCID: PMC11141617 DOI: 10.1126/sciadv.adk9681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
In response to energy and nutrient shortage, the liver triggers several catabolic processes to promote survival. Despite recent progress, the precise molecular mechanisms regulating the hepatic adaptation to fasting remain incompletely characterized. Here, we report the identification of hydroxysteroid dehydrogenase-like 2 (HSDL2) as a mitochondrial protein highly induced by fasting. We show that the activation of PGC1α-PPARα and the inhibition of the PI3K-mTORC1 axis stimulate HSDL2 expression in hepatocytes. We found that HSDL2 depletion decreases cholesterol conversion to bile acids (BAs) and impairs FXR activity. HSDL2 knockdown also reduces mitochondrial respiration, fatty acid oxidation, and TCA cycle activity. Bioinformatics analyses revealed that hepatic Hsdl2 expression positively associates with the postprandial excursion of various BA species in mice. We show that liver-specific HSDL2 depletion affects BA metabolism and decreases circulating cholesterol levels upon refeeding. Overall, our report identifies HSDL2 as a fasting-induced mitochondrial protein that links nutritional signals to BAs and cholesterol homeostasis.
Collapse
Affiliation(s)
- Nolwenn Samson
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Cristina R. Bosoi
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Christian Roy
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laurie Turcotte
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laura Tribouillard
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Line Berthiaume
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Jocelyn Trottier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Heitor C. G. Silva
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Thomas Guerbette
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Aurèle Besse-Patin
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Alicia Montoni
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
| | - Nicolò Ilacqua
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Centre de recherche CERVO, Québec, QC, Canada
| | - Jennifer Lamothe
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yemima R. Citron
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Yves Gélinas
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | | | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Alexandre Caron
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Mathieu Morissette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Luca Pellegrini
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, QC, Canada
| | - Patrick J. Rochette
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
- Département d’Ophtalmologie et ORL – chirurgie cervico-faciale, Université Laval, Québec, QC, Canada
| | - Jennifer L. Estall
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
- Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Michael Shum
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Olivier Barbier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - André Marette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
38
|
Onaka GM, de Carvalho MR, Onaka PK, Barbosa CM, Martinez PF, de Oliveira-Junior SA. Exercise, mTOR Activation, and Potential Impacts on the Liver in Rodents. BIOLOGY 2024; 13:362. [PMID: 38927242 PMCID: PMC11201249 DOI: 10.3390/biology13060362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024]
Abstract
The literature offers a consensus on the association between exercise training (ET) protocols based on the adequate parameters of intensity and frequency, and several adaptive alterations in the liver. Indeed, regular ET can reverse glucose and lipid metabolism disorders, especially from aerobic modalities, which can decrease intrahepatic fat formation. In terms of molecular mechanisms, the regulation of hepatic fat formation would be directly related to the modulation of the mechanistic target of rapamycin (mTOR), which would be stimulated by insulin signaling and Akt activation, from the following three different primary signaling pathways: (I) growth factor, (II) energy/ATP-sensitive, and (III) amino acid-sensitive signaling pathways, respectively. Hyperactivation of the Akt/mTORC1 pathway induces lipogenesis by regulating the action of sterol regulatory element binding protein-1 (SREBP-1). Exercise training interventions have been associated with multiple metabolic and tissue benefits. However, it is worth highlighting that the mTOR signaling in the liver in response to exercise interventions remains unclear. Hepatic adaptive alterations seem to be most outstanding when sustained by chronic interventions or high-intensity exercise protocols.
Collapse
Affiliation(s)
- Giuliano Moreto Onaka
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
| | - Marianna Rabelo de Carvalho
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
| | - Patricia Kubalaki Onaka
- Graduate Program in Education and Health, State University of Mato Grosso do Sul, Dourados 79804-970, MS, Brazil
| | - Claudiane Maria Barbosa
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| | - Paula Felippe Martinez
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| | - Silvio Assis de Oliveira-Junior
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| |
Collapse
|
39
|
Bo T, Gao L, Yao Z, Shao S, Wang X, Proud CG, Zhao J. Hepatic selective insulin resistance at the intersection of insulin signaling and metabolic dysfunction-associated steatotic liver disease. Cell Metab 2024; 36:947-968. [PMID: 38718757 DOI: 10.1016/j.cmet.2024.04.006] [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: 10/03/2023] [Revised: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 06/26/2024]
Abstract
Insulin resistance (IR) is a major pathogenic factor in the progression of MASLD. In the liver, insulin suppresses gluconeogenesis and enhances de novo lipogenesis (DNL). During IR, there is a defect in insulin-mediated suppression of gluconeogenesis, but an unrestrained increase in hepatic lipogenesis persists. The mechanism of increased hepatic steatosis in IR is unclear and remains controversial. The key discrepancy is whether insulin retains its ability to directly regulate hepatic lipogenesis. Blocking insulin/IRS/AKT signaling reduces liver lipid deposition in IR, suggesting insulin can still regulate lipid metabolism; hepatic glucose metabolism that bypasses insulin's action may contribute to lipogenesis; and due to peripheral IR, other tissues are likely to impact liver lipid deposition. We here review the current understanding of insulin's action in governing different aspects of hepatic lipid metabolism under normal and IR states, with the purpose of highlighting the essential issues that remain unsettled.
Collapse
Affiliation(s)
- Tao Bo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Zhenyu Yao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia.
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China.
| |
Collapse
|
40
|
Willmann K, Moita LF. Physiologic disruption and metabolic reprogramming in infection and sepsis. Cell Metab 2024; 36:927-946. [PMID: 38513649 DOI: 10.1016/j.cmet.2024.02.013] [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: 11/05/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024]
Abstract
Effective responses against severe systemic infection require coordination between two complementary defense strategies that minimize the negative impact of infection on the host: resistance, aimed at pathogen elimination, and disease tolerance, which limits tissue damage and preserves organ function. Resistance and disease tolerance mostly rely on divergent metabolic programs that may not operate simultaneously in time and space. Due to evolutionary reasons, the host initially prioritizes the elimination of the pathogen, leading to dominant resistance mechanisms at the potential expense of disease tolerance, which can contribute to organ failure. Here, we summarize our current understanding of the role of physiological perturbations resulting from infection in immune response dynamics and the metabolic program requirements associated with resistance and disease tolerance mechanisms. We then discuss how insight into the interplay of these mechanisms could inform future research aimed at improving sepsis outcomes and the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Katharina Willmann
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Luis F Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal; Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
41
|
Feng R, Liu F, Li R, Zhou Z, Lin Z, Lin S, Deng S, Li Y, Nong B, Xia Y, Li Z, Zhong X, Yang S, Wan G, Ma W, Wu S, Songyang Z. The rapid proximity labeling system PhastID identifies ATP6AP1 as an unconventional GEF for Rheb. Cell Res 2024; 34:355-369. [PMID: 38448650 PMCID: PMC11061317 DOI: 10.1038/s41422-024-00938-z] [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: 09/10/2023] [Accepted: 02/02/2024] [Indexed: 03/08/2024] Open
Abstract
Rheb is a small G protein that functions as the direct activator of the mechanistic target of rapamycin complex 1 (mTORC1) to coordinate signaling cascades in response to nutrients and growth factors. Despite extensive studies, the guanine nucleotide exchange factor (GEF) that directly activates Rheb remains unclear, at least in part due to the dynamic and transient nature of protein-protein interactions (PPIs) that are the hallmarks of signal transduction. Here, we report the development of a rapid and robust proximity labeling system named Pyrococcus horikoshii biotin protein ligase (PhBPL)-assisted biotin identification (PhastID) and detail the insulin-stimulated changes in Rheb-proximity protein networks that were identified using PhastID. In particular, we found that the lysosomal V-ATPase subunit ATP6AP1 could dynamically interact with Rheb. ATP6AP1 could directly bind to Rheb through its last 12 amino acids and utilizes a tri-aspartate motif in its highly conserved C-tail to enhance Rheb GTP loading. In fact, targeting the ATP6AP1 C-tail could block Rheb activation and inhibit cancer cell proliferation and migration. Our findings highlight the versatility of PhastID in mapping transient PPIs in live cells, reveal ATP6AP1's role as an unconventional GEF for Rheb, and underscore the importance of ATP6AP1 in integrating mTORC1 activation signals through Rheb, filling in the missing link in Rheb/mTORC1 activation.
Collapse
Affiliation(s)
- Ran Feng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Ruofei Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhifen Zhou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhuoheng Lin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Song Lin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shengcheng Deng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingying Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Baoting Nong
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Xia
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhiyi Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoqin Zhong
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuhan Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gang Wan
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenbin Ma
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Su Wu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
42
|
Deans-Fielder K, Wu T, Nguyen T, To S, Huang YZ, Bark SJ, Mills JC, Shroyer NF. Mechanisms driving fasting-induced protection from genotoxic injury in the small intestine. Am J Physiol Gastrointest Liver Physiol 2024; 326:G504-G524. [PMID: 38349111 PMCID: PMC11376978 DOI: 10.1152/ajpgi.00126.2023] [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: 06/23/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 04/05/2024]
Abstract
Genotoxic agents such as doxorubicin (DXR) can cause damage to the intestines that can be ameliorated by fasting. How fasting is protective and the optimal timing of fasting and refeeding remain unclear. Here, our analysis of fasting/refeeding-induced global intestinal transcriptional changes revealed metabolic shifts and implicated the cellular energetic hub mechanistic target of rapamycin complex 1 (mTORC1) in protecting from DXR-induced DNA damage. Our analysis of specific transcripts and proteins in intestinal tissue and tissue extracts showed that fasting followed by refeeding at the time of DXR administration reduced damage and caused a spike in mTORC1 activity. However, continued fasting after DXR prevented the mTORC1 spike and damage reduction. Surprisingly, the mTORC1 inhibitor, rapamycin, did not block fasting/refeeding-induced reduction in DNA damage, suggesting that increased mTORC1 is dispensable for protection against the initial DNA damage response. In Ddit4-/- mice [DDIT4 (DNA-damage-inducible transcript 4) functions to regulate mTORC1 activity], fasting reduced DNA damage and increased intestinal crypt viability vs. ad libitum-fed Ddit4-/- mice. Fasted/refed Ddit4-/- mice maintained body weight, with increased crypt proliferation by 5 days post-DXR, whereas ad libitum-fed Ddit4-/- mice continued to lose weight and displayed limited crypt proliferation. Genes encoding epithelial stem cell and DNA repair proteins were elevated in DXR-injured, fasted vs. ad libitum Ddit4-/- intestines. Thus, fasting strongly reduced intestinal damage when normal dynamic regulation of mTORC1 was lost. Overall, the results confirm that fasting protects the intestines against DXR and suggests that fasting works by pleiotropic - including both mTORC1-dependent and independent - mechanisms across the temporally dynamic injury response.NEW & NOTEWORTHY New findings are 1) DNA damage reduction following a 24-h fast depends on the timing of postfast refeeding in relation to chemotherapy initiation; 2) fasting/refeeding-induced upregulation of mTORC1 activity is not required for early (6 h) protection against DXR-induced DNA damage; and 3) fasting increases expression of intestinal stem cell and DNA damage repair genes, even when mTORC1 is dysregulated, highlighting fasting's crucial role in regulating mTORC1-dependent and independent mechanisms in the dynamic recovery process.
Collapse
Affiliation(s)
- Kali Deans-Fielder
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Translational Biology and Molecular Medicine Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, United States
| | - Timothy Wu
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Thanh Nguyen
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Cancer and Cell Biology Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
| | - Sarah To
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Yang-Zhe Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Cancer and Cell Biology Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
| | - Steven J Bark
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Jason C Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Noah F Shroyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Translational Biology and Molecular Medicine Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
43
|
Zhang P, Catterson JH, Grönke S, Partridge L. Inhibition of S6K lowers age-related inflammation and increases lifespan through the endolysosomal system. NATURE AGING 2024; 4:491-509. [PMID: 38413780 PMCID: PMC11031405 DOI: 10.1038/s43587-024-00578-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Suppression of target of rapamycin complex 1 (TORC1) by rapamycin ameliorates aging in diverse species. S6 kinase (S6K) is an essential mediator, but the mechanisms involved are unclear. Here we show that activation of S6K specifically in Drosophila fat-body blocked extension of lifespan by rapamycin, induced accumulation of multilamellar lysosomes and blocked age-associated hyperactivation of the NF-κB-like immune deficiency (IMD) pathway, indicative of reduced inflammaging. Syntaxin 13 mediated the effects of TORC1-S6K signaling on lysosome morphology and inflammaging, suggesting they may be linked. Inflammaging depended on the IMD receptor regulatory isoform PGRP-LC, and repression of the IMD pathway from midlife extended lifespan. Age-related inflammaging was higher in females than in males and was not lowered in males by rapamycin treatment or lowered S6K. Rapamycin treatment also elevated Syntaxin 12/13 levels in mouse liver and prevented age-related increase in noncanonical NF-κB signaling, suggesting that the effect of TORC1 on inflammaging is conserved from flies to mammals.
Collapse
Affiliation(s)
- Pingze Zhang
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - James H Catterson
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK.
| |
Collapse
|
44
|
Xie M, Kaiser M, Gershtein Y, Schnyder D, Deviatiiarov R, Gazizova G, Shagimardanova E, Zikmund T, Kerckhofs G, Ivashkin E, Batkovskyte D, Newton PT, Andersson O, Fried K, Gusev O, Zeberg H, Kaiser J, Adameyko I, Chagin AS. The level of protein in the maternal murine diet modulates the facial appearance of the offspring via mTORC1 signaling. Nat Commun 2024; 15:2367. [PMID: 38531868 DOI: 10.1038/s41467-024-46030-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 02/09/2024] [Indexed: 03/28/2024] Open
Abstract
The development of craniofacial skeletal structures is fascinatingly complex and elucidation of the underlying mechanisms will not only provide novel scientific insights, but also help develop more effective clinical approaches to the treatment and/or prevention of the numerous congenital craniofacial malformations. To this end, we performed a genome-wide analysis of RNA transcription from non-coding regulatory elements by CAGE-sequencing of the facial mesenchyme of human embryos and cross-checked the active enhancers thus identified against genes, identified by GWAS for the normal range human facial appearance. Among the identified active cis-enhancers, several belonged to the components of the PI3/AKT/mTORC1/autophagy pathway. To assess the functional role of this pathway, we manipulated it both genetically and pharmacologically in mice and zebrafish. These experiments revealed that mTORC1 signaling modulates craniofacial shaping at the stage of skeletal mesenchymal condensations, with subsequent fine-tuning during clonal intercalation. This ability of mTORC1 pathway to modulate facial shaping, along with its evolutionary conservation and ability to sense external stimuli, in particular dietary amino acids, indicate that the mTORC1 pathway may play a role in facial phenotypic plasticity. Indeed, the level of protein in the diet of pregnant female mice influenced the activity of mTORC1 in fetal craniofacial structures and altered the size of skeletogenic clones, thus exerting an impact on the local geometry and craniofacial shaping. Overall, our findings indicate that the mTORC1 signaling pathway is involved in the effect of environmental conditions on the shaping of craniofacial structures.
Collapse
Affiliation(s)
- Meng Xie
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Flemingsberg, Sweden
- School of Psychological and Cognitive Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Markéta Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Yaakov Gershtein
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela Schnyder
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ruslan Deviatiiarov
- Regulatory Genomics Research Center, Kazan Federal University, Kazan, Russia
- Endocrinology Research Center, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Intractable Disease Research Center, Juntendo University, Tokyo, Japan
| | - Guzel Gazizova
- Regulatory Genomics Research Center, Kazan Federal University, Kazan, Russia
| | - Elena Shagimardanova
- Regulatory Genomics Research Center, Kazan Federal University, Kazan, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Tomáš Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Greet Kerckhofs
- Biomechanics Lab, Institute of Mechanics, Materials, and Civil Engineering (iMMC), UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research (IREC), UCLouvain, Woluwe, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- Prometheus, Division for Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Evgeny Ivashkin
- A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
- Department of Developmental and Comparative Physiology, N.K. Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dominyka Batkovskyte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Phillip T Newton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's hospital, Stockholm, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Oleg Gusev
- Regulatory Genomics Research Center, Kazan Federal University, Kazan, Russia
- Endocrinology Research Center, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Intractable Disease Research Center, Juntendo University, Tokyo, Japan
| | - Hugo Zeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
45
|
Plata-Gómez AB, de Prado-Rivas L, Sanz A, Deleyto-Seldas N, García F, de la Calle Arregui C, Silva C, Caleiras E, Graña-Castro O, Piñeiro-Yáñez E, Krebs J, Leiva-Vega L, Muñoz J, Jain A, Sabio G, Efeyan A. Hepatic nutrient and hormone signaling to mTORC1 instructs the postnatal metabolic zonation of the liver. Nat Commun 2024; 15:1878. [PMID: 38499523 PMCID: PMC10948770 DOI: 10.1038/s41467-024-46032-1] [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: 04/20/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024] Open
Abstract
The metabolic functions of the liver are spatially organized in a phenomenon called zonation, linked to the differential exposure of portal and central hepatocytes to nutrient-rich blood. The mTORC1 signaling pathway controls cellular metabolism in response to nutrients and insulin fluctuations. Here we show that simultaneous genetic activation of nutrient and hormone signaling to mTORC1 in hepatocytes results in impaired establishment of postnatal metabolic and zonal identity of hepatocytes. Mutant hepatocytes fail to upregulate postnatally the expression of Frizzled receptors 1 and 8, and show reduced Wnt/β-catenin activation. This defect, alongside diminished paracrine Wnt2 ligand expression by endothelial cells, underlies impaired postnatal maturation. Impaired zonation is recapitulated in a model of constant supply of nutrients by parenteral nutrition to piglets. Our work shows the role of hepatocyte sensing of fluctuations in nutrients and hormones for triggering a latent metabolic zonation program.
Collapse
Affiliation(s)
- Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Lucía de Prado-Rivas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Alba Sanz
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Nerea Deleyto-Seldas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Fernando García
- Proteomics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Celia de la Calle Arregui
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Camila Silva
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Eduardo Caleiras
- Histopathology Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA-Nemesio Díez), School of Medicine, San Pablo-CEU University, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Joseph Krebs
- Department of Pediatrics, Saint Louis University, Saint Louis, MO, USA
| | - Luis Leiva-Vega
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Muñoz
- Proteomics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Cell Signalling and Clinical Proteomics Group, Biocruces Bizkaia Health Research Institute & Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Ajay Jain
- Department of Pediatrics, Saint Louis University, Saint Louis, MO, USA
| | - Guadalupe Sabio
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
46
|
Iyer SH, Yeh MY, Netzel L, Lindsey MG, Wallace M, Simeone KA, Simeone TA. Dietary and Metabolic Approaches for Treating Autism Spectrum Disorders, Affective Disorders and Cognitive Impairment Comorbid with Epilepsy: A Review of Clinical and Preclinical Evidence. Nutrients 2024; 16:553. [PMID: 38398876 PMCID: PMC10893388 DOI: 10.3390/nu16040553] [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: 12/24/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Epilepsy often occurs with other neurological disorders, such as autism, affective disorders, and cognitive impairment. Research indicates that many neurological disorders share a common pathophysiology of dysfunctional energy metabolism, neuroinflammation, oxidative stress, and gut dysbiosis. The past decade has witnessed a growing interest in the use of metabolic therapies for these disorders with or without the context of epilepsy. Over one hundred years ago, the high-fat, low-carbohydrate ketogenic diet (KD) was formulated as a treatment for epilepsy. For those who cannot tolerate the KD, other diets have been developed to provide similar seizure control, presumably through similar mechanisms. These include, but are not limited to, the medium-chain triglyceride diet, low glycemic index diet, and calorie restriction. In addition, dietary supplementation with ketone bodies, polyunsaturated fatty acids, or triheptanoin may also be beneficial. The proposed mechanisms through which these diets and supplements work to reduce neuronal hyperexcitability involve normalization of aberrant energy metabolism, dampening of inflammation, promotion of endogenous antioxidants, and reduction of gut dysbiosis. This raises the possibility that these dietary and metabolic therapies may not only exert anti-seizure effects, but also reduce comorbid disorders in people with epilepsy. Here, we explore this possibility and review the clinical and preclinical evidence where available.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Timothy A. Simeone
- Department of Pharmacology & Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (S.H.I.); (K.A.S.)
| |
Collapse
|
47
|
Ruppert PMM, Kersten S. Mechanisms of hepatic fatty acid oxidation and ketogenesis during fasting. Trends Endocrinol Metab 2024; 35:107-124. [PMID: 37940485 DOI: 10.1016/j.tem.2023.10.002] [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: 08/31/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 11/10/2023]
Abstract
Fasting is part of many weight management and health-boosting regimens. Fasting causes substantial metabolic adaptations in the liver that include the stimulation of fatty acid oxidation and ketogenesis. The induction of fatty acid oxidation and ketogenesis during fasting is mainly driven by interrelated changes in plasma levels of various hormones and an increase in plasma nonesterified fatty acid (NEFA) levels and is mediated transcriptionally by the peroxisome proliferator-activated receptor (PPAR)α, supported by CREB3L3 (cyclic AMP-responsive element-binding protein 3 like 3). Compared with men, women exhibit higher ketone levels during fasting, likely due to higher NEFA availability, suggesting that the metabolic response to fasting shows sexual dimorphism. Here, we synthesize the current molecular knowledge on the impact of fasting on hepatic fatty acid oxidation and ketogenesis.
Collapse
Affiliation(s)
- Philip M M Ruppert
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5000 C Odense, Denmark
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708 WE Wageningen, The Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
48
|
Uehara K, Lee WD, Stefkovich M, Biswas D, Santoleri D, Garcia Whitlock A, Quinn W, Coopersmith T, Creasy KT, Rader DJ, Sakamoto K, Rabinowitz JD, Titchenell PM. mTORC1 controls murine postprandial hepatic glycogen synthesis via Ppp1r3b. J Clin Invest 2024; 134:e173782. [PMID: 38290087 PMCID: PMC10977990 DOI: 10.1172/jci173782] [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: 07/07/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024] Open
Abstract
In response to a meal, insulin drives hepatic glycogen synthesis to help regulate systemic glucose homeostasis. The mechanistic target of rapamycin complex 1 (mTORC1) is a well-established insulin target and contributes to the postprandial control of liver lipid metabolism, autophagy, and protein synthesis. However, its role in hepatic glucose metabolism is less understood. Here, we used metabolomics, isotope tracing, and mouse genetics to define a role for liver mTORC1 signaling in the control of postprandial glycolytic intermediates and glycogen deposition. We show that mTORC1 is required for glycogen synthase activity and glycogenesis. Mechanistically, hepatic mTORC1 activity promotes the feeding-dependent induction of Ppp1r3b, a gene encoding a phosphatase important for glycogen synthase activity whose polymorphisms are linked to human diabetes. Reexpression of Ppp1r3b in livers lacking mTORC1 signaling enhances glycogen synthase activity and restores postprandial glycogen content. mTORC1-dependent transcriptional control of Ppp1r3b is facilitated by FOXO1, a well characterized transcriptional regulator involved in the hepatic response to nutrient intake. Collectively, we identify a role for mTORC1 signaling in the transcriptional regulation of Ppp1r3b and the subsequent induction of postprandial hepatic glycogen synthesis.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute for Diabetes, Obesity, and Metabolism
- Biochemistry and Molecular Biophysics Graduate Group, and
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Won Dong Lee
- Lewis Sigler Institute for Integrative Genomics
- Department of Chemistry, and
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, New Jersey, USA
| | | | - Dipsikha Biswas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dominic Santoleri
- Institute for Diabetes, Obesity, and Metabolism
- Biochemistry and Molecular Biophysics Graduate Group, and
| | | | | | | | - Kate Townsend Creasy
- Institute for Diabetes, Obesity, and Metabolism
- Department of Medicine, Division of Translational Medicine and Human Genetics, and
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J. Rader
- Institute for Diabetes, Obesity, and Metabolism
- Department of Medicine, Division of Translational Medicine and Human Genetics, and
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics
- Department of Chemistry, and
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, New Jersey, USA
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity, and Metabolism
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Zhang L, Lu Y, An J, Wu Y, Liu Z, Zou MH. AMPKα2 regulates fasting-induced hyperketonemia by suppressing SCOT ubiquitination and degradation. Sci Rep 2024; 14:1713. [PMID: 38242911 PMCID: PMC10798978 DOI: 10.1038/s41598-023-49991-5] [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: 07/27/2023] [Accepted: 12/14/2023] [Indexed: 01/21/2024] Open
Abstract
Ketone bodies serve as an energy source, especially in the absence of carbohydrates or in the extended exercise. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a crucial energy sensor that regulates lipid and glucose metabolism. However, whether AMPK regulates ketone metabolism in whole body is unclear even though AMPK regulates ketogenesis in liver. Prolonged resulted in a significant increase in blood and urine levels of ketone bodies in wild-type (WT) mice. Interestingly, fasting AMPKα2-/- and AMPKα1-/- mice exhibited significantly higher levels of ketone bodies in both blood and urine compared to fasting WT mice. BHB tolerance assays revealed that both AMPKα2-/- and AMPKα1-/- mice exhibited slower ketone consumption compared to WT mice, as indicated by higher blood BHB or urine BHB levels in the AMPKα2-/- and AMPKα1-/- mice even after the peak. Interestingly, fasting AMPKα2-/- and AMPKα1-/- mice exhibited significantly higher levels of ketone bodies in both blood and urine compared to fasting WT mice. . Specifically, AMPKα2ΔMusc mice showed approximately a twofold increase in blood BHB levels, and AMPKα2ΔMyo mice exhibited a 1.5-fold increase compared to their WT littermates after a 48-h fasting. However, blood BHB levels in AMPKα1ΔMusc and AMPKα1ΔMyo mice were as same as in WT mice. Notably, AMPKα2ΔMusc mice demonstrated a slower rate of BHB consumption in the BHB tolerance assay, whereas AMPKα1ΔMusc mice did not show such an effect. Declining rates of body weights and blood glucoses were similar among all the mice. Protein levels of SCOT, the rate-limiting enzyme of ketolysis, decreased in skeletal muscle of AMPKα2-/- mice. Moreover, SCOT protein ubiquitination increased in C2C12 cells either transfected with kinase-dead AMPKα2 or subjected to AMPKα2 inhibition. AMPKα2 physiologically binds and stabilizes SCOT, which is dependent on AMPKα2 activity.
Collapse
Affiliation(s)
- Lingxue Zhang
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA
| | - Yanqiao Lu
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA
| | - Junqing An
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA
| | - Yin Wu
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA
| | - Zhixue Liu
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA.
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, USA
| |
Collapse
|
50
|
Liu Y, Zhang M, Jang H, Nussinov R. The allosteric mechanism of mTOR activation can inform bitopic inhibitor optimization. Chem Sci 2024; 15:1003-1017. [PMID: 38239681 PMCID: PMC10793652 DOI: 10.1039/d3sc04690g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
mTOR serine/threonine kinase is a cornerstone in the PI3K/AKT/mTOR pathway. Yet, the detailed mechanism of activation of its catalytic core is still unresolved, likely due to mTOR complexes' complexity. Its dysregulation was implicated in cancer and neurodevelopmental disorders. Using extensive molecular dynamics (MD) simulations and compiled published experimental data, we determine exactly how mTOR's inherent motifs can control the conformational changes in the kinase domain, thus kinase activity. We also chronicle the critical regulation by the unstructured negative regulator domain (NRD). When positioned inside the catalytic cleft (NRD IN state), mTOR tends to adopt a deep and closed catalytic cleft. This is primarily due to the direct interaction with the FKBP-rapamycin binding (FRB) domain which restricts it, preventing substrate access. Conversely, when outside the catalytic cleft (NRD OUT state), mTOR favors an open conformation, exposing the substrate-binding site on the FRB domain. We further show how an oncogenic mutation (L2427R) promotes shifting the mTOR ensemble toward the catalysis-favored state. Collectively, we extend mTOR's "active-site restriction" mechanism and clarify mutation action. In particular, our mechanism suggests that RMC-5552 (RMC-6272) bitopic inhibitors may benefit from adjustment of the (PEG8) linker length when targeting certain mTOR variants. In the cryo-EM mTOR/RMC-5552 structure, the distance between the allosteric and orthosteric inhibitors is ∼22.7 Å. With a closed catalytic cleft, this linker bridges the sites. However, in our activation mechanism, in the open cleft it expands to ∼24.7 Å, offering what we believe to be the first direct example of how discovering an activation mechanism can potentially increase the affinity of inhibitors targeting mutants.
Collapse
Affiliation(s)
- Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Mingzhen Zhang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA +1-301-846-5579
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA +1-301-846-5579
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA +1-301-846-5579
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University Tel Aviv 69978 Israel
| |
Collapse
|