1
|
Yan L, Li X, Xu J, Tang S, Wang G, Shi M, Liu P. The CNC-family transcription factor NRF3: A crucial therapeutic target for cancer treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167794. [PMID: 40081618 DOI: 10.1016/j.bbadis.2025.167794] [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: 11/20/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
The CNC-bZIP family member NRF3 (NFE2L3) has received limited attention since its discovery. However, recent research has gradually revealed its biological functions, such as involvement in the regulation of cell differentiation, lipid metabolism, and malignant cell proliferation. Under physiological conditions, NRF3 is anchored to the endoplasmic reticulum within the cytoplasm and is biologically inactive. Upon cellular exposure to microenvironmental stresses such as oxidative stress, NRF3 translocates to the nucleus, binds to DNA, and acts as a transcription factor by inducing or repressing the expression of various genes. In terms of tumor regulation, NRF3 exhibits a dual role. It can function as a tumor suppressor to prevent the malignant progression of tumor tissues, protecting the organism from harm. Conversely, current research indicates that NRF3 plays a tumor-promoting role in most tumor tissues. NRF3 enhances the proliferation, migration and invasion of tumor cells by regulating cell cycle-related proteins and enhancing proteasome assembly to degrade tumor suppressors. Studies correlating NRF3 expression with clinical tumor features have found that elevated NRF3 expression is often associated with poor prognoses in various cancers, with patients exhibiting higher NRF3 expression typically having lower survival rates. Several studies suggest that NRF3 could serve as a clinical diagnostic and prognostic marker for tumors. Finally, from the clinical perspective, exploring the feasibility of inhibiting NRF3 activity in tumor treatment provides new insights for the development of NRF3-targeted oncological therapies.
Collapse
Affiliation(s)
- Liangwen Yan
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shenkang Tang
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Gang Wang
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| | - Mengjiao Shi
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| |
Collapse
|
2
|
Sun CL, Xu C, Itani O, Christensen EL, Vijay H, Ho J, Correa-Medina A, Klingler CB, Mathew ND, Flibotte S, Humphreys IR, Rubalcaba DD, Ritter AE, Desbois M, Grill B, Crowder CM. Biased regulation of protein synthesis and hypoxic death by a conditional raptor mutation. Curr Biol 2025:S0960-9822(25)00504-4. [PMID: 40339571 DOI: 10.1016/j.cub.2025.04.040] [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: 11/25/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
Mechanistic target of rapamycin (mTOR) functions in mTOR complex 1 (mTORC1) with raptor to match metazoan metabolism to available nutrients to regulate multiple cellular, physiological, and pathological processes. Hypoxic cellular injury is influenced by the mTORC1 pathway, but whether its activity promotes or prevents injury is unclear, and which mTORC1-regulated mechanisms control hypoxic injury are obscure. Here, we report the discovery of a hypoxia-resistant, temperature-sensitive raptor mutant in an unbiased forward mutagenesis screen in C. elegans. This raptor mutant is both hypoxia resistant and long lived at intermediate temperatures, while unable to develop at higher temperatures. Temperature-shift experiments show that the conditional hypoxia resistance can be induced in the raptor mutant immediately prior to the hypoxic insult. At these intermediate temperatures, the raptor mutation selectively reduces protein synthesis without affecting autophagy, and epistasis experiments implicate mTOR-targeted translation regulators as components of the hypoxia resistance mechanism. Using the conditional developmental arrest phenotype in a selection for suppressors of raptor loss of function, we isolated multiple second-site raptor missense mutants, whose mutated residue is predicted to interact with RagA, a raptor-binding protein. These suppressor mutations restore normal protein synthesis, hypoxic sensitivity, and lifespan and thereby implicate raptor-RagA interactions as critical to these biological processes.
Collapse
Affiliation(s)
- Chun-Ling Sun
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Cong Xu
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Omar Itani
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Elyse L Christensen
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Harshitha Vijay
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Jessica Ho
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Abraham Correa-Medina
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Christian B Klingler
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Neal D Mathew
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Stephane Flibotte
- Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Ian R Humphreys
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Box 357350, Seattle, WA 98105, USA; Institute for Protein Design, University of Washington, 3946 W Stevens Way NE, Box 351655, Seattle, WA 98105, USA
| | - Diego Delgadillo Rubalcaba
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Alison E Ritter
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Muriel Desbois
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA; Departments of Pediatrics and Pharmacology, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - C Michael Crowder
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Box 355065, 3720 15th Avenue NE, Seattle, WA 98105, USA.
| |
Collapse
|
3
|
Kalenta H, Kilroe SP, Romsdahl TB, Marchant ED, Maroto R, Linares JJ, Russell WK, Rasmussen BB. Constitutively active mTORC1 signaling modifies the skeletal muscle metabolome and lipidome response to exercise. J Appl Physiol (1985) 2025; 138:1173-1186. [PMID: 40215109 DOI: 10.1152/japplphysiol.00987.2024] [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] [Revised: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 05/01/2025] Open
Abstract
A chronic increase in the Mammalian Target of Rapamycin Complex 1 (mTORC1) signaling is implicated in reduced longevity, altered metabolism, and mitochondrial dysfunction. Abnormal mTORC1 signaling may also be involved in the etiology of sarcopenia. To better understand the role of mTORC1 signaling in the regulation of muscle metabolism, we developed an inducible muscle-specific knockout model of DEP domain-containing 5 protein (DEPDC5 mKO), which results in constitutively active mTORC1 signaling. We hypothesized that constitutively active mTORC1 signaling in skeletal muscle would alter the metabolomic and lipidomic response to an acute bout of exercise. Wild-type (WT) and DEPDC5 muscle-specific knockout (KO) mice were studied at rest and following a 1 h bout of treadmill exercise. Acute exercise induced an increased reliance on glycolytic and pentose phosphate pathway (PPP) metabolites in the muscle of mice with hyperactive mTORC1. Lipidomic analysis showed an increase in triglycerides (TGs) in KO mice. Although exercise had a pronounced effect on muscle metabolism, the genotype effect was larger, indicating that constitutively active mTORC1 signaling exerts a dominant influence on metabolic and lipidomic regulation. We conclude that increased mTORC1 signaling shifts muscle metabolism toward greater reliance on nonoxidative energy sources in response to exercise. Understanding the mechanisms responsible for these effects may lead to the development of strategies for restoring proper mTORC1 signaling in conditions such as aging and sarcopenia.NEW & NOTEWORTHY This study demonstrates that hyperactive mTORC1 alters the muscle metabolomic and lipidomic response to exercise, with genotype having a larger effect than exercise. Knockout mice exhibited an increase in reliance on glycolysis and pentose phosphate pathway and an increase in triglyceride turnover. Wild-type mice primarily showed an increase in utilization of TCA cycle and lipid metabolism intermediates.
Collapse
Affiliation(s)
- Hanna Kalenta
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Sean P Kilroe
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Trevor B Romsdahl
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Erik D Marchant
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Rosario Maroto
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
| | - Jennifer J Linares
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Mass Spectrometry Facility, University of Texas Medical Branch, Galveston, Texas, United States
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Mass Spectrometry Facility, University of Texas Medical Branch, Galveston, Texas, United States
| | - Blake B Rasmussen
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| |
Collapse
|
4
|
Rieber J, Wolint P, Meier-Bürgisser G, Ongini E, Giovanoli P, Calcagni M, Snedeker JG, Buschmann J. Synergistic Effects of Insulin-like Growth Factor-1 and Platelet-Derived Growth Factor-BB in Tendon Healing. Int J Mol Sci 2025; 26:4039. [PMID: 40362278 PMCID: PMC12072114 DOI: 10.3390/ijms26094039] [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: 03/31/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Tendon ruptures are common musculoskeletal injuries associated with prolonged healing and complications such as adhesion formation and rerupture. Despite advancements in treatment strategies, full functional recovery remains a challenge. Growth factors (GFs) like insulin-like growth factor-1 (IGF-1) and platelet-derived growth factor-BB (PDGF-BB) play key roles in tendon repair and may have synergistic effects when applied together. To support tendon healing, a bioactive electrospun polymer scaffold made of Degrapol® (DP) was developed, incorporating IGF-1, PDGF-BB, or both. A range of in vitro and in vivo analyses were performed to assess scaffold structure, cell behavior, gene expression, metabolism, and biomechanical and adhesion outcomes three weeks post-surgery. Interestingly, the combined application of IGF-1 and PDGF-BB did not simply amplify individual effects but showed a complex interaction. Depending on the parameter and time point, the combination led to either enhanced or reduced responses compared to single-factor treatments, indicating a synergistic modulation rather than a purely additive effect. These findings suggest that the combination of IGF-1 and PDGF-BB can modulate key cellular and molecular processes in tendon regeneration, making this approach a promising strategy to improve tendon healing.
Collapse
Affiliation(s)
- Julia Rieber
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| | - Petra Wolint
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| | - Gabriella Meier-Bürgisser
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| | - Esteban Ongini
- Institute for Biomechanics, ETH Zurich, 8092 Zurich, Switzerland; (E.O.); (J.G.S.)
- Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Pietro Giovanoli
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| | - Maurizio Calcagni
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| | - Jess G. Snedeker
- Institute for Biomechanics, ETH Zurich, 8092 Zurich, Switzerland; (E.O.); (J.G.S.)
- Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Johanna Buschmann
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (P.W.); (G.M.-B.); (P.G.); (M.C.)
| |
Collapse
|
5
|
Chang L, Čok Z, Yu L. Protein Kinases as Mediators for miRNA Modulation of Neuropathic Pain. Cells 2025; 14:577. [PMID: 40277902 PMCID: PMC12025903 DOI: 10.3390/cells14080577] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Neuropathic pain is a chronic condition resulting from injury or dysfunction in the somatosensory nervous system, which leads to persistent pain and a significant impairment of quality of life. Research has highlighted the complex molecular mechanisms that underlie neuropathic pain and has begun to delineate the roles of microRNAs (miRNAs) in modulating pain pathways. miRNAs, which are small non-coding RNAs that regulate gene expression post-transcriptionally, have been shown to influence key cellular processes, including neuroinflammation, neuronal excitability, and synaptic plasticity. These processes contribute to the persistence of neuropathic pain, and miRNAs have emerged as critical regulators of pain behaviors by modulating signaling pathways that control pain sensitivity. miRNAs can influence neuropathic pain by targeting genes that encode protein kinases involved in pain signaling. This review focuses on miRNAs that have been demonstrated to modulate neuropathic pain behavior through their effects on protein kinases or their immediate upstream regulators. The relationship between miRNAs and neuropathic pain behaviors is characterized as either an upregulation or a downregulation of miRNA levels that leads to a reduction in neuropathic pain. In the case of miRNA upregulation resulting in an alleviation of neuropathic pain behaviors, protein kinases exhibit a positive correlation with neuropathic pain, whereas decreased protein kinase levels correlate with diminished neuropathic pain behaviors. The only exception is GRK2, which shows an inverse correlation with neuropathic pain. In the case of miRNA downregulation resulting in a reduction in neuropathic pain behaviors, protein kinases display mixed relationships to neuropathic pain, with some kinases exhibiting positive correlation, while others exhibit negative correlation. By exploring how protein kinases mediate miRNA modulation of neuropathic pain, valuable insight may be gained into the pathophysiology of neuropathic pain, offering potential therapeutic targets for developing more effective strategies for pain management.
Collapse
Affiliation(s)
| | | | - Lei Yu
- Department of Genetics, Center of Alcohol & Substance Use Studies, Rutgers University, Piscataway, NJ 08854, USA; (L.C.)
| |
Collapse
|
6
|
Bi J, Sun Y, Guo M, Sun X, Sun J, Jiang R, Wang N, Huang G. Lysosomes: guardians and healers within cells- multifaceted perspective and outlook from injury repair to disease treatment. Cancer Cell Int 2025; 25:136. [PMID: 40205430 PMCID: PMC11984033 DOI: 10.1186/s12935-025-03771-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Lysosomes, as crucial organelles within cells, carry out diverse biological functions such as waste degradation, regulation of the cellular environment, and precise control of cell signaling. This paper reviews the core functions and structural characteristics of lysosomes, and delves into the current research status of lysosomes damage repair mechanisms. Subsequently, we explore in depth the close association between lysosomes and various diseases, including but not limited to age-related chronic diseases, neuro-degenerative diseases, tumors, inflammation, and immune imbalance. Additionally, we also provide a detailed discussion of the application of lysosome-targeted substances in the field of regenerative medicine, especially the enormous potential demonstrated in key areas such as stem cell regulation and therapy, and myocardial cell repair. Though the integration of multidisciplinary research efforts, we believe that lysosomes damage repair mechanisms will demonstrate even greater application value in disease treatment and regenerative medicine.
Collapse
Affiliation(s)
- Jianlei Bi
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China
| | - Yincong Sun
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Meihua Guo
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiaoxin Sun
- College of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, P.R. China
| | - Jie Sun
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Rujiao Jiang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Gena Huang
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China.
| |
Collapse
|
7
|
Sonsalla MM, Babygirija R, Johnson M, Cai S, Cole M, Yeh CY, Grunow I, Liu Y, Vertein D, Calubag MF, Trautman ME, Green CL, Rigby MJ, Puglielli L, Lamming DW. Acarbose ameliorates Western diet-induced metabolic and cognitive impairments in the 3xTg mouse model of Alzheimer's disease. GeroScience 2025; 47:1569-1591. [PMID: 39271570 PMCID: PMC11978593 DOI: 10.1007/s11357-024-01337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Age is the greatest risk factor for Alzheimer's disease (AD) as well as for other disorders that increase the risk of AD such as diabetes and obesity. There is growing interest in determining if interventions that promote metabolic health can prevent or delay AD. Acarbose is an anti-diabetic drug that not only improves glucose homeostasis, but also extends the lifespan of wild-type mice. Here, we test the hypothesis that acarbose will not only preserve metabolic health, but also slow or prevent AD pathology and cognitive deficits in 3xTg mice, a model of AD, fed either a Control diet or a high-fat, high-sucrose Western diet (WD). We find that acarbose decreases the body weight and adiposity of WD-fed 3xTg mice, increasing energy expenditure while also stimulating food consumption, and improves glycemic control. Both male and female WD-fed 3xTg mice have worsened cognitive deficits than Control-fed mice, and these deficits are ameliorated by acarbose treatment. Molecular and histological analysis of tau and amyloid pathology identified sex-specific effects of acarbose which are uncoupled from the dramatic improvements in cognition in females, suggesting that the benefits of acarbose on AD may be largely driven by improved metabolic health. In conclusion, our results suggest that acarbose may be a promising intervention to prevent, delay, or even treat AD, especially in individuals consuming a WD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Madeline Johnson
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Samuel Cai
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mari Cole
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Yang Liu
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Diana Vertein
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael J Rigby
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, 53705, USA.
| |
Collapse
|
8
|
Ren B, Chen Y, Liu J, Zhou Z, He Y, Wan S, Chen Y, Wu X, Du M, Gao H, Liu L, Shen H. DNA methylation of genes that mediate autophagosome formation contributes to iodine-induced autoimmune thyroiditis: A population-based study conducted at regions with different iodine levels in China. Int J Hyg Environ Health 2025; 265:114537. [PMID: 39965511 DOI: 10.1016/j.ijheh.2025.114537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Autoimmune thyroiditis (AIT) results from a combination of genetic predispositions and environmental factors. This study aims to examine the methylation patterns of genes that mediate autophagosome formation (MAF) between AIT patients and controls from areas with different water iodine levels. METHODS A protein-protein interaction (PPI) network was constructed to explore the interactions of autophagy-related genes (ARGs). The MCODE plugin in Cytoscape software identified two functional epigenetic modules. We included 176 AIT cases from regions with varying water iodine concentrations and matched controls at a 1:1 ratio. The MethylTarget™ assay was employed to assess DNA methylation changes in six MAF-related genes and analyzed the association between iodine levels and epigenetic modifications. RESULTS Significant methylation differences were observed in 11 targets with 75 CpG sites on MAF-related genes between AIT patients and controls (P < 0.05). Methylation levels were generally lower in AIT patients. A negative correlation was found between the methylation status of PRKAB1_36 and urinary iodine concentration (UIC) (rs = -0.208, P = 0.006). In iodine-fortification areas (IFA), AIT patients exhibited significantly higher mRNA expression levels of six MAF-related genes in whole blood compared to controls (P < 0.05). CONCLUSION Environmental water iodine concentration influences the DNA methylation status of MAF-related genes. Hypomethylated targets and sites in these genes were identified, and the relative mRNA expression levels of these genes were significantly increased in AIT cases from regions with a history of prolonged iodine deficiency followed by iodine supplementation.
Collapse
Affiliation(s)
- Bingxuan Ren
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo City, Zhejiang Province 315211, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Yun Chen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Jinjin Liu
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Zheng Zhou
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Yanhong He
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Siyuan Wan
- Department of Preventive Medicine, Qiqihar Medical University, Qiqihar City, Heilongjiang Province 161006, People's Republic of China
| | - Yao Chen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Xianhao Wu
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo City, Zhejiang Province 315000, People's Republic of China
| | - Mengxue Du
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Haiyan Gao
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; Department of Clinical Laboratory, The Sixth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province 150023, People's Republic of China
| | - Lixiang Liu
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Hongmei Shen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China.
| |
Collapse
|
9
|
Chen B, Zhang C, Zhou M, Deng H, Xu J, Yin J, Chen C, Zhang D, Pu Y, Zheng L, Wang B, Fu J. CD4+ T-cell metabolism in the pathogenesis of Sjogren's syndrome. Int Immunopharmacol 2025; 150:114320. [PMID: 39970711 DOI: 10.1016/j.intimp.2025.114320] [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: 08/30/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
The abnormal effector function of CD4+ T cells plays a key role in the pathogenesis of Sjogren's syndrome (SS) and its associated systematic autoimmune response. Cellular metabolism, including glucose metabolism, lipid metabolism and amino acid metabolism, supports proliferation, migration, survival and differentiation into distinct CD4+ T-cell subsets. Different subtypes of T cells have significantly different demands for related metabolic processes, which enables us to finely regulate CD4+ T cells through different metabolic processes in autoimmune diseases such as SS. In this review, we summarize the effects of disturbances in distinct metabolic processes, such as glycolysis, fatty acid metabolism, glutamine decomposition, mitochondrial dynamics, and ferroptosis, on how to support the effector functions of CD4+ T cells in the SS. We also discuss potential drugs with high value in the treatment of SS through metabolic normalization in CD4+ T cells. Finally, we propose possible directions for future targeted therapy for immunometabolism in SS.
Collapse
Affiliation(s)
- Baixi Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Chenji Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Mengyuan Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Hongyu Deng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Jiabao Xu
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians University of Würzburg, Würzburg 97255, Germany
| | - Junhao Yin
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prothodontics, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Changyu Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai 200001, China
| | - Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Yiping Pu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China
| | - Baoli Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology & National Clinical Research Center of Oral Disease, Shanghai Key Laboratory of Stomatology, Shanghai 200001, China.
| | - Jiayao Fu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prothodontics, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China.
| |
Collapse
|
10
|
Haws SA, Liu Y, Green CL, Chaiyakul K, Mishra P, Babygirija R, Armstrong EA, Mehendale AT, Ong IM, Lamming DW, Denu JM. Individual dietary amino acid restrictions induce distinct metabolic and chromatin states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.06.570456. [PMID: 38106163 PMCID: PMC10723491 DOI: 10.1101/2023.12.06.570456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Dietary protein and essential amino acid (EAA) restriction promote favorable metabolic reprogramming, although the extent to which shared or EAA-specific mechanisms facilitate diet-associated phenotypes remains unclear. Here, we compared the physiological and molecular effects of dietary methionine, leucine, or isoleucine depletion (Met-D, Leu-D, and Ile-D) in C57BL/6J mice. Each diet elicited responses not phenocopied by mTORC1 inhibition, including reduced fat mass and hepatic amino acid catabolism. Ile-D yielded additional distinct responses, highlighted by histone H2A/H4 hypoacetylation and maintained hepatic acetyl-CoA levels despite downregulated FA β-oxidation. Multi-Omics Factor Analysis of 14,139 data points objectively affirmed Ile-D phenotypes are distinct from Met-D or Leu-D and identified several metabolic and chromatin features as primary discriminators. Metabolic and epigenetic responses to Ile-D were recapitulated in vitro , suggesting underlying mechanisms represent fundamental cellular properties. Together, these results demonstrate EAAs can stimulate unique phenotypes and highlight distinct molecular mechanisms by which EAAs may inform metabolic health.
Collapse
|
11
|
Minton DM, Ailiani AR, Focht MD, Kersh ME, Li A, Elliehausen CJ, Sonsalla MM, Lamming DW, Marolf AJ, Santangelo KS, Salmon AB, Konopka AR. Inhibition of mTORC1 by rapamycin results in feedback activation of Akt S473 and aggravates hallmarks of osteoarthritis in female mice and non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.14.594256. [PMID: 38798488 PMCID: PMC11118493 DOI: 10.1101/2024.05.14.594256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Purpose Genetic deletion of mTOR has protected against post-traumatic osteoarthritis (OA) in male mice, however, effects of pharmacological mTOR-inhibition are equivocal and have not been tested in aging models nor in female subjects. Therefore, the goal of this study was to determine if mTOR-inhibition by rapamycin can modify OA pathology in aging non-human primates and female mice. Methods Common marmosets were administered oral rapamycin (1mg/kg/day) or vehicle starting near mid-life until death. Five-month-old, female C57BL/6J mice were treated with vehicle or rapamycin (IP, 2mg/kg, 3x/week) for 8-weeks following non-invasive ACL rupture. Knee OA pathology was assessed via microCT and histology. Phosphorylation of mTORC1 (p-RPS6S235/36) and mTORC2 (p-AktS473, p-NDRG1T638, p-PKCαT348) substrates were evaluated via western blot in articular cartilage, meniscus, and/or infrapatellar fat pad. ATDC5 cells were cultured with rapamycin to determine time and dose effects on mTORC1/2 signaling. Results In marmosets, rapamycin did not impact age-related radiographic OA severity or cartilage pathology but increased medial meniscus calcification and lowered lateral tibia subchondral thickness, particularly in females. In female mice, rapamycin worsened ACLR-induced meniscus calcification and cartilage pathology. In marmoset and mouse joint tissues, rapamycin inhibited mTORC1 and increased p-AktS473 but not p-NDRG1T638 or p-PKCαT348. This mTOR signaling pattern was replicated in ATDC5 cells during exposure to low concentrations of rapamycin. Conclusions Rapamycin attenuated mTORC1 signaling with feedback activation of AktS473 in articular cartilage, meniscus, and/or infrapatellar fat pad and was accompanied by deleterious effects on meniscus calcification and/or cartilage pathology in female mice and common marmosets.
Collapse
Affiliation(s)
- Dennis M. Minton
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Aditya R. Ailiani
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Michael D.K. Focht
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Mariana E. Kersh
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Champaign, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Amy Li
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Christian J. Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Michelle M. Sonsalla
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Dudley W. Lamming
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Angela J. Marolf
- Department of Veterinary Clinical Sciences, Ohio State University, Columbus, OH, USA
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Adam B. Salmon
- Barshop Institute for Longevity and Aging Studies and Departments of Molecular Medicine and Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, TX, USA
- Geriatric Research, Education, and Clinical Center, South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - Adam R. Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
12
|
Li T, Huang N, Chen H, Yang Y, Zhang J, Xu W, Gong H, Gong C, Yang M, Zhao T, Wang F, Xiao H. Daytime-Restricted Feeding Alleviates D-Galactose-Induced Aging in Mice and Regulates the AMPK and mTORC1 Activities. J Cell Physiol 2025; 240:e70020. [PMID: 40070151 DOI: 10.1002/jcp.70020] [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] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/17/2025]
Abstract
Time-restricted feeding (TRF) is a distinct regimen of intermittent fasting advocated for health improving. Although nighttime TRF (NRF) in rodents is analogous to daytime TRF (DRF) in humans and has health benefits, the effects of DRF on rodent's health remain uncertain. The adverse health effects of DRF in rodents are primarily attributed to its implementation-induced temporal shift in the expression of circadian rhythm-related genes. However, studies also demonstrate the health-beneficial effect of restricted feeding itself on metabolic homeostasis, particularly in periphery tissues. Moreover, the direct effects of DRF on aging progression in rodents are underexplored, highlighting a gap in current research. To explore the overall health effects of long-term DRF in rodents, especially its influence on aging progression, we investigated the impact of long-term DRF on mice under a progeric aging condition. Results showed that both 4-h and 8-h DRF regimens exerted positive effects on aging retardation; these effects were manifested as improved physical and memory capacities, enhanced liver and kidney functions, and reduced oxidative damage and inflammatory response. These DRF regimens also lowered the manifestation of aging-related markers in peripheral tissues, with decreased SA-β-gal staining and p16 expression. Mechanistically, DRF regimens, especially DRF8, upregulated AMPK signaling and downregulated mTORC1 signaling. Interestingly, the health benefits of DRF are similar to those of metformin intervention. In conclusion, our study demonstrates for the first time that DRF effectively counteracts oxidative stress-induced aging progression in mice, supporting the viewpoint that TRF as a promising strategy for preventing aging and aging-related disorders.
Collapse
Affiliation(s)
- Tiepeng Li
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Huang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Honghan Chen
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weitong Xu
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Gong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chuhui Gong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Yang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Zhao
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [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: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
14
|
Jefcoate CR, Larsen MC, Song YS, Maguire M, Sheibani N. Defined Diets Link Iron and α-Linolenic Acid to Cyp1b1 Regulation of Neonatal Liver Development Through Srebp Forms and LncRNA H19. Int J Mol Sci 2025; 26:2011. [PMID: 40076634 PMCID: PMC11901102 DOI: 10.3390/ijms26052011] [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: 11/02/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 03/14/2025] Open
Abstract
Cyp1b1 substantially affects hepatic vascular and stellate cells (HSC) with linkage to liver fibrosis. Despite minimal hepatocyte expression, Cyp1b1 deletion substantially impacts liver gene expression at birth and weaning. The appreciable Cyp1b1 expression in surrounding embryo mesenchyme, during early organogenesis, provides a likely source for Cyp1b1. Here defined breeder diets established major interconnected effects on neonatal liver of α-linolenic acid (ALA), vitamin A deficiency (VAD) and suboptimal iron fed mice. At birth Cyp1b1 deletion and VAD each activated perinatal HSC, while suppressing iron control by hepcidin. Cyp1b1 deletion also advanced the expression of diverse genes linked to iron regulation. Postnatal stimulations of Srebp-regulated genes in the fatty acid and cholesterol biosynthesis pathways were suppressed by Cyp1b1-deficiency. LncRNA H19 and the neutrophil alarmin S100a9 expression increased due to slower postnatal decline with Cyp1b1 deficiency. VAD reversed each of Cyp1b1 effect, probably due to enhanced HSC release of Apo-Rbp4. At birth, Cyp1b1 deletion enhanced H19 participation. Notably, a suppressor (Cnot3) decreased while an activity partner (Ezh2/H3K methylation) increased H19 expression. ALA elevated hepcidin mRNA and countered the inhibitory effects of Cyp1b1 deletion on hepcidin expression. Oxylipin metabolites of ALA from highly expressed hepatic Cyps are potential mediators. Cyp expression patterns demonstrated female dimorphism for neonatal liver. Mothers followed one of three fetal growth support programs probably linked to maturity at conception.
Collapse
Affiliation(s)
- Colin R. Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Michele C. Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Meghan Maguire
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| |
Collapse
|
15
|
Jiang X, Wang T, Zhao B, Sun H, Dong Y, Ma Y, Li Z, Wu Y, Wang K, Guan X, Long B, Qin L, Shi W, Shi L, He Q, Liu W, Li M, Xiao L, Zhou C, Sun H, Yang J, Guan J, Zhou H, Yu Z, Jiao Z. KRAS G12D-driven pentose phosphate pathway remodeling imparts a targetable vulnerability synergizing with MRTX1133 for durable remissions in PDAC. Cell Rep Med 2025; 6:101966. [PMID: 39970873 PMCID: PMC11866490 DOI: 10.1016/j.xcrm.2025.101966] [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: 06/24/2024] [Revised: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
The KRASG12D inhibitor MRTX1133 shows the potential to revolutionize the treatment paradigm for pancreatic ductal adenocarcinoma (PDAC), yet presents challenges. Our findings indicate that KRASG12D remodels a pentose phosphate pathway (PPP)-dominant central carbon metabolism pattern, facilitating malignant progression and resistance to MRTX1133 in PDAC. Mechanistically, KRASG12D drives excessive degradation of p53 and glucose-6-phosphate dehydrogenase (G6PD)-mediated PPP reprogramming through retinoblastoma (Rb)/E2F1/p53 axis-regulated feedback loops that amplify ubiquitin-conjugating enzyme E2T (UBE2T) transcription. Genetic ablation or pharmacological inhibition of UBE2T significantly suppresses PDAC progression and potentiates MRTX1133 efficacy. Leveraging structure advantages of the UBE2T inhibitor pentagalloylglucose (PGG), we develop a self-assembling nano co-delivery system with F-127, PGG, and MRTX1133. This system enhances the efficacy of PGG and MRTX1133, achieving durable remissions (85% overall response rate) and long-term survival (100% progression-free survival) in patient-derived xenografts and spontaneous PDAC mice. This study reveals the role of KRASG12D-preferred PPP reprogramming in MRTX1133 resistance and proposes a potentially therapeutic strategy for KRASG12D-mutated PDAC.
Collapse
Affiliation(s)
- Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Tao Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Bin Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Haonan Sun
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuman Dong
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Zhigang Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuxia Wu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Keshen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Xiaoying Guan
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Bo Long
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Long Qin
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Wengui Shi
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Lei Shi
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qichen He
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Wenbo Liu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Mingdou Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Lixia Xiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Chengliang Zhou
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Hui Sun
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jing Yang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Junhong Guan
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Huinian Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China.
| |
Collapse
|
16
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Glorieux C, Enríquez C, Buc Calderon P. The complex interplay between redox dysregulation and mTOR signaling pathway in cancer: A rationale for cancer treatment. Biochem Pharmacol 2025; 232:116729. [PMID: 39709038 DOI: 10.1016/j.bcp.2024.116729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that plays a critical role in regulating cellular processes such as growth, proliferation, and metabolism in healthy cells. Dysregulation of mTOR signaling and oxidative stress have been implicated in various diseases including cancer. This review aims to provide an overview of the current understanding of mTOR and its involvement in cell survival and the regulation of cancer cell metabolism as well as its complex interplay with reactive oxygen species (ROS). On the one hand, ROS can inhibit or activate mTOR pathway in cancer cells through various mechanisms. Conversely, mTOR signaling can induce oxidative stress in tumor cells notably due to the inhibition in the expression of antioxidant enzyme genes. Since mTOR is often activated and plays crucial role in cancer cell survival, the use of mTOR inhibitors, which often induce ROS accumulation, could be an interesting approach for cancer treatment. This review will address the advantages, disadvantages, combination strategies, and limitations associated with therapeutic modulation of mTOR signaling pathway in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, China.
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Programa de Doctorado en Química Medicinal, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000 Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Brussels, Belgium.
| |
Collapse
|
18
|
Sun X, Cui H, Li J, An B, Liu R, Guo Z, Chu D, Geng X, Cui B, Zhu L, Li J, Li Z. An injectable shape-adaptive hydrogel system for subconjunctival injuries: In situ and permanently releases rapamycin to prevent fibrosis via promoting autophagy. Mater Today Bio 2025; 30:101380. [PMID: 39790484 PMCID: PMC11713510 DOI: 10.1016/j.mtbio.2024.101380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/27/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
Subconjunctival fibrosis (SCF) is a common and refractory eye disease that is a serious threat to vision. The severe side effects of existing drugs and low drug bioavailability due to the ocular barrier are major challenges in SCF treatment. Hence, there is an urgent need to explore safer and more effective strategies for administering anti-SCF drugs. Herein, an injectable and adaptable hydrogel system containing the antifibrotic drug rapamycin was fabricated to address this complex need. This system possesses moderate mechanical properties, self-healing and shape-adaptive capabilities, injectability, and biosafety. It is designed to promote autophagy by modulating the PI3K/AKT/mTOR/WIPI2 pathway, thereby inhibiting SCF. In vivo experiments utilizing a rat subconjunctival injury model indicated that in situ administration of this hydrogel system effectively inhibited SCF. This system constitutes a promising method for promoting autophagy to protect against SCF, which will foster its widespread application for other fibrotic diseases.
Collapse
Affiliation(s)
| | | | - Jingfan Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Boyuan An
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Ruixing Liu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhihua Guo
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Dandan Chu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Xingchen Geng
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Bingbing Cui
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lei Zhu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jingguo Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhanrong Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| |
Collapse
|
19
|
Calubag MF, Ademi I, Grunow I, Breuer L, Babygirija R, Lialios P, Le S, Minton D, Sonsalla MM, Illiano J, Knopf BA, Xiao F, Konopka AR, Harris DA, Lamming DW. Tissue-specific effects of dietary protein on cellular senescence are mediated by branched-chain amino acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632607. [PMID: 39868338 PMCID: PMC11761368 DOI: 10.1101/2025.01.13.632607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Dietary protein is a key regulator of healthy aging in both mice and humans. In mice, reducing dietary levels of the branched-chain amino acids (BCAAs) recapitulates many of the benefits of a low protein diet; BCAA-restricted diets extend lifespan, reduce frailty, and improve metabolic health, while BCAA supplementation shortens lifespan, promotes obesity, and impairs glycemic control. Recently, high protein diets have been shown to promote cellular senescence, a hallmark of aging implicated in many age-related diseases, in the liver of mice. Here, we test the hypothesis that the effects of high protein diets on metabolic health and on cell senescence are mediated by BCAAs. We find that reducing dietary levels of BCAAs protects male and female mice from the negative metabolic consequences of both normal and high protein diets. Further, we identify tissue-specific effects of BCAAs on cellular senescence, with restriction of all three BCAAs - but not individual BCAAs - protecting from hepatic cellular senescence while potentiating cell senescence in white adipose tissue. We find that the effects of BCAAs on hepatic cellular senescence are cell-autonomous, with lower levels of BCAAs protecting cultured cells from antimycin-A induced senescence. Our results demonstrate a direct effect of a specific dietary component on a hallmark of aging and suggest that cellular senescence may be highly susceptible to dietary interventions.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ismail Ademi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Lucia Breuer
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Penelope Lialios
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Sandra Le
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Dennis Minton
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia Illiano
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Bailey A Knopf
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Fan Xiao
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David A Harris
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
20
|
Sousa MDC, Cavalcanti CM, Conde AJH, Alves BVDF, Cesar LFB, de Sena JN, Miguel YH, Fernandes CCL, Alves JPM, Teixeira DÍA, Rondina D. Short Supply of High Levels of Guanidine Acetic Acid, Alters Ovarian Artery Flow and Improves Intraovarian Blood Perfusion Area Associated with Follicular Growth in Sheep. Animals (Basel) 2025; 15:143. [PMID: 39858143 PMCID: PMC11758299 DOI: 10.3390/ani15020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Guanidinoacetic acid (GAA), a precursor of creatine, has a recognized effect on ruminant performance when used as a dietary supplement. However, its impact on reproductive response remains to be elucidated. Therefore, this study aimed to contribute initially to this area by supplementing the diets of ewes with a high dose of GAA, evaluating its effects on reproductive response. Twenty adult sheep had their estrus synchronized using an MPA sponge, eCG, and PGF2α. After estrus detection ewes were mated. For 10 days until mating, ewes were grouped in groups of baseline diet (BSD; n = 10) and GAA diet (GAAD; n = 10), which was the BSD with daily 0.9 g/kg DM of GAA. After the eCG + PGF2α dose, the GAAD group exhibited an increase in the peak diastolic and pulsatility of the ovarian artery, a reduction in the systolic/diastolic peaks ratio, and a larger intraovarian blood perfusion area. A greater depletion of follicles with <3 mm was observed in the GAAD group and a higher number of follicles ≥3 mm. No differences were observed between the diets respect to pregnancy, and twin rates. Thus, a high GAA supply before mating significantly alters ovarian vasculature and improves follicular growth in ewes but does not affect the pregnancy rate.
Collapse
Affiliation(s)
- Marta da Costa Sousa
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Camila Muniz Cavalcanti
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Alfredo José Herrera Conde
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Bruna Vitória de Freitas Alves
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Larissa Fernandes Baia Cesar
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Jhennyfe Nobre de Sena
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Yohana Huicho Miguel
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | | | - Juliana Paula Martins Alves
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Dárcio Ítalo Alves Teixeira
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| | - Davide Rondina
- School of Veterinary Medicine, Ceará State University (UECE), Fortaleza 60714-903, Ceará, Brazil; (M.d.C.S.); (C.M.C.); (A.J.H.C.); (B.V.d.F.A.); (L.F.B.C.); (J.N.d.S.); (Y.H.M.); (J.P.M.A.); (D.Í.A.T.)
| |
Collapse
|
21
|
Chen J, Li H, Zhuo J, Lin Z, Hu Z, He C, Wu X, Jin Y, Lin Z, Su R, Sun Y, Wang R, Sun J, Wei X, Zheng S, Lu D, Xu X. Impact of immunosuppressants on tumor pulmonary metastasis: new insight into transplantation for hepatocellular carcinoma. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0267. [PMID: 39718153 PMCID: PMC11667780 DOI: 10.20892/j.issn.2095-3941.2024.0267] [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: 07/28/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Pulmonary metastasis is a life-threatening complication for patients with hepatocellular carcinoma (HCC) undergoing liver transplantation (LT). In addition to the common mechanisms underlying tumor metastasis, another inevitable factor is that the application of immunosuppressive agents, including calcineurin inhibitors (CNIs) and rapamycin inhibitors (mTORis), after transplantation could influence tumor recurrence and metastasis. In recent years, several studies have reported that mTORis, unlike CNIs, have the capacity to modulate the tumorigenic landscape post-liver transplantation by targeting metastasis-initiating cells and reshaping the pulmonary microenvironment. Therefore, we focused on the effects of immunosuppressive agents on the lung metastatic microenvironment and how mTORis impact tumor growth in distant organs. This revelation has provided profound insights into transplant oncology, leading to a renewed understanding of the use of immunosuppressants after LT for HCC.
Collapse
Affiliation(s)
- Jinyan Chen
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huigang Li
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Zuyuan Lin
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310053, China
| | - Zhihang Hu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chiyu He
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiang Wu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiru Jin
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhanyi Lin
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Renyi Su
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiyang Sun
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310063, China
| | - Rongsen Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310053, China
| | - Jiancai Sun
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou 310022, China
| | - Di Lu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310014, China
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310014, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| |
Collapse
|
22
|
Yao Z, Chen H. Everolimus in pituitary tumor: a review of preclinical and clinical evidence. Front Endocrinol (Lausanne) 2024; 15:1456922. [PMID: 39736867 PMCID: PMC11682973 DOI: 10.3389/fendo.2024.1456922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Although pituitary tumors (PTs) are mostly benign, some PTs are characterized by low surgical resection rates, high recurrence rates, and poor response to conventional treatments and profoundly affect patients' quality of life. Everolimus (EVE) is the only FDA-approved mTOR inhibitor, which can be used for oral treatment. It effectively inhibits tumor cell proliferation and angiogenesis. It has been administered for various neuroendocrine tumors of the digestive tract, lungs, and pancreas. EVE not only suppresses the growth and proliferation of APT cells but also enhances their sensitivity to radiotherapy and chemotherapy. This review introduces the role of the PI3K/AKT/mTOR pathway in the development of APTs, comprehensively explores the current status of preclinical and clinical research of EVE in APTs, and discusses the blood-brain barrier permeability and safety of EVE.
Collapse
Affiliation(s)
- Zihong Yao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hui Chen
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
23
|
Hochmuth L, Hirrlinger J. Physiological and Pathological Role of mTOR Signaling in Astrocytes. Neurochem Res 2024; 50:53. [PMID: 39652154 PMCID: PMC11628441 DOI: 10.1007/s11064-024-04306-6] [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: 09/06/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is one of the key regulators of cellular energy metabolism. It senses diverse alterations in the extracellular environment such as availability of nutrients and growth factors, and mediates the corresponding intracellular response. In the brain, astrocytes crucially contribute to energy and neurotransmitter metabolism, and numerous other functions. However, the relevance of physiological, astrocytic mTOR signaling in maintaining brain homeostasis and function is not well understood. Pathophysiological mTOR signaling is involved in manifold diseases in the central nervous system and most of the knowledge about astrocytic mTOR signaling has been derived from observations on these disorders. Dysregulation of the mTOR signaling pathway impairs important functions of astrocytes including neurotransmitter uptake and -signaling as well as energy metabolism. Some of these alterations could trigger neuropathological conditions such as epilepsy. This review focuses on how mTOR signaling regulates properties of astrocytes, and how these signaling events might contribute to the physiological function of the brain.
Collapse
Affiliation(s)
- Luise Hochmuth
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, D- 04103, Leipzig, Germany.
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, D- 04103, Leipzig, Germany.
- Department of Neurogenetics, Max-Planck-Institute for Multidisciplinary Sciences, D- 37075, Göttingen, Germany.
| |
Collapse
|
24
|
Qian M, Wan Z, Liang X, Jing L, Zhang H, Qin H, Duan W, Chen R, Zhang T, He Q, Lu M, Jiang J. Targeting autophagy in HCC treatment: exploiting the CD147 internalization pathway. Cell Commun Signal 2024; 22:583. [PMID: 39627812 PMCID: PMC11616386 DOI: 10.1186/s12964-024-01956-5] [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: 07/22/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND/AIMS Chemotherapy resistance in liver cancer is a major clinical issue, with CD147 playing a vital role in this process. However, the specific mechanisms underlying these processes remain largely unknown. This study investigates how CD147 internalization leads to cytoprotective autophagy, contributing to chemotherapy resistance in hepatocellular carcinoma (HCC). METHODS Utilizing bioinformatics methods for KEGG pathways enrichment and screening key molecules associated with chemotherapy resistance through analyses of GEO and TCGA databases. An overexpression/knockdown system was used to study how CD147 internalization leads to autophagy in vitro and in vivo. The process was observed using microscopes, and molecular interactions and autophagy flux were analyzed. Analyzing the internalization of CD147 intracellular domains and the interaction with G3BP1 in clinical chemotherapy recurrence HCC tissues by immunohistochemistry, tissue immunofluorescence, and mass spectrometry. A tumor xenograft mice model was used to study cytoprotective autophagy induced by CD147 and test the effectiveness of combining cisplatin with an autophagy inhibitor in nude mice models. RESULTS In our study, we identified the tumor-associated membrane protein CD147, which implicated in chemoresistance lysosome pathways, by evaluating its protein degree value and betweenness centrality using Cytoscape. Our findings revealed that CD147 undergoes internalization and interacts with G3BP1 following treatment with cisplatin and methyl-β-cyclodextrin, forming a complex that is transported to lysosomes via Rab7A. Notably, higher doses of cisplatin enhanced CD147-mediated lysosomal transport while concurrently inhibiting SG assembly. The CD147-G3BP1 complex additionally inhibits mTOR activity, promoting autophagy and augmenting chemoresistance in hepatoma cells. In vivo studies investigations and analyses of clinical samples revealed that elevated internalization of CD147 is associated with chemotherapy recurrence in liver cancer and the maintenance of stem cells. Mice experiments found that the combined administration of cisplatin and hydroxychloroquine enhanced the efficacy of treatment. CONCLUSIONS This study reveals that CD147 internalization and CD147-G3BP1 complex translocation to lysosomes induce cytoprotective autophagy, reducing chemotherapy sensitivity by suppressing mTOR activity. It is also shown that chemotherapy drugs combined with autophagy inhibitors can improve the therapeutic effect of cancer, providing new insights into potential targeted therapeutic approaches in treating HCC.
Collapse
Affiliation(s)
- Meirui Qian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Ziyu Wan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Jing
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Huijie Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Heyao Qin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenli Duan
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ruo Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, 710032, China
| | - Tianjiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Qian He
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Meng Lu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianli Jiang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
25
|
Yeh CY, Chini LCS, Davidson JW, Garcia GG, Gallagher MS, Freichels IT, Calubag MF, Rodgers AC, Green CL, Babygirija R, Sonsalla MM, Pak HH, Trautman ME, Hacker TA, Miller RA, Simcox JA, Lamming DW. Late-life protein or isoleucine restriction impacts physiological and molecular signatures of aging. NATURE AGING 2024; 4:1760-1771. [PMID: 39604703 PMCID: PMC11672203 DOI: 10.1038/s43587-024-00744-7] [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: 01/09/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024]
Abstract
Restricting the intake of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan in young or adult mice. However, their effects when initiated in aged animals are unknown. Here we investigate the consequences of consuming a diet with 67% reduction of all amino acids (low AA) or of isoleucine alone (low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. Both dietary regimens effectively promote overall metabolic health without reducing calorie intake. Both low AA and low Ile diets improve aspects of frailty and slow multiple molecular indicators of aging rate; however, the low Ile diet reduces grip strength in both sexes and has mixed, sexually dimorphic effects on the heart. These results demonstrate that low AA and low Ile diets can promote aspects of healthy aging in aged mice and suggest that similar interventions might promote healthy aging in older adults.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Lucas C S Chini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Gonzalo G Garcia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Meredith S Gallagher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac T Freichels
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- 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
| | - Allison C Rodgers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, 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
| | - 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
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- 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.
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
26
|
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
|
27
|
Bettedi L, Zhang Y, Yang S, Lilly MA. Unveiling GATOR2 Function: Novel Insights from Drosophila Research. Cells 2024; 13:1795. [PMID: 39513902 PMCID: PMC11545208 DOI: 10.3390/cells13211795] [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: 08/09/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The multiprotein Target of Rapamycin (TOR) Complex 1 (TORC1) is a serine/threonine kinase that stimulates anabolic metabolism and suppresses catabolism. Deregulation of TORC1 is implicated in various human pathologies, including cancer, epilepsy, and neurodegenerative disorders. The Gap Activity Towards Rags (GATOR) complex contains two subcomplexes: GATOR1, which inhibits TORC1 activity; and GATOR2, which counteracts GATOR1s function. Structural and biochemical studies have elucidated how GATOR1 regulates TORC1 activity by acting as a GTPase activating protein for Rag GTPase. However, while cryogenic electron microscopy has determined that the structure of the multi-protein GATOR2 complex is conserved from yeast to humans, how GATOR2 inhibits GATOR1 remains unclear. Here, we describe recent whole-animal studies in Drosophila that have yielded novel insights into GATOR2 function, including identifying a novel role for the GATOR2 subunit WDR59, redefining the core proteins sufficient for GATOR2 activity, and defining a TORC1-independent role for GATOR2 in the regulation of the lysosomal autophagic endomembrane system. Additionally, the recent characterization of a novel methionine receptor in Drosophila that acts through the GATOR2 complex suggests an attractive model for the evolution of species-specific nutrient sensors. Research on GATOR2 function in Drosophila highlights how whole-animal genetic models can be used to dissect intracellular signaling pathways to identify tissue-specific functions and functional redundancies that may be missed in studies confined to rapidly proliferating cell lines.
Collapse
Affiliation(s)
- Lucia Bettedi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| | - Yingbiao Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China;
| | - Shu Yang
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| | - Mary A. Lilly
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| |
Collapse
|
28
|
Huang Q, Wen C, Gu S, Jie Y, Li G, Yan Y, Tian C, Wu G, Yang N. Synergy of gut microbiota and host genome in driving heterosis expression of chickens. J Genet Genomics 2024; 51:1121-1134. [PMID: 38950856 DOI: 10.1016/j.jgg.2024.06.011] [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: 04/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Heterosis has been widely utilized in agricultural production. Despite over a century of extensive research, the underlying mechanisms of heterosis remain elusive. Most hypotheses and research have focused on the genetic basis of heterosis. However, the potential role of gut microbiota in heterosis has been largely ignored. Here, we carefully design a crossbreeding experiment with two distinct broiler breeds and conduct 16S rRNA amplicon and transcriptome sequencing to investigate the synergistic role of gut microbiota and host genes in driving heterosis. We find that the breast muscle weight of hybrids exhibits a high heterosis, 6.28% higher than the mid-parent value. A notable difference is observed in the composition and potential function of cecal microbiota between hybrids and their parents. Over 90% of differentially colonized microbiota and differentially expressed genes exhibit nonadditive patterns. Integrative analyses uncover associations between nonadditive genes and nonadditive microbiota, including a connection between the expression of cellular signaling pathways and metabolism-related genes and the abundance of Odoribacter, Oscillibacter, and Alistipes in hybrids. Moreover, higher abundances of these microbiota are related to better meat yield. In summary, these findings highlight the importance of gut microbiota in heterosis, serving as crucial factors that modulate heterosis expression in chickens.
Collapse
Affiliation(s)
- Qiang Huang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangqi Li
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Yiyuan Yan
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Chuanyao Tian
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Guiqin Wu
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| |
Collapse
|
29
|
Babygirija R, Han JH, Sonsalla MM, Matoska R, Calubag MF, Green CL, Tobon A, Yeh CY, Vertein D, Schlorf S, Illiano J, Liu Y, Grunow I, Rigby MJ, Puglielli L, Harris DA, Denu JM, Lamming DW. Fasting is required for many of the benefits of calorie restriction in the 3xTg mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613904. [PMID: 39386545 PMCID: PMC11463641 DOI: 10.1101/2024.09.19.613904] [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: 10/12/2024]
Abstract
Caloric restriction (CR) is a widely recognized geroprotective intervention that slows or prevents Alzheimer's disease (AD) in animal models. CR is typically implemented via feeding mice a single meal per day; as CR mice rapidly consume their food, they are subject to a prolonged fast between meals. While CR has been shown to improve metabolic and cognitive functions and suppress pathological markers in AD mouse models, the specific contributions of fasting versus calorie reduction remains unclear. Here, we investigated the contribution of fasting and energy restriction to the beneficial effects of CR on AD progression. To test this, we placed 6-month-old 3xTg mice on one of several diet regimens, allowing us to dissect the effects of calories and fasting on metabolism, AD pathology, and cognition. We find that energy restriction alone, without fasting, was sufficient to improve glucose tolerance and reduce adiposity in both sexes, and to reduce Aβ plaques and improve aspects of cognitive performance in females. However, we find that a prolonged fast between meals is necessary for many of the benefits of CR, including improved insulin sensitivity, reduced phosphorylation of tau, decreased neuroinflammation, inhibition of mTORC1 signaling, and activation of autophagy, as well as for the full cognitive benefits of CR. Finally, we find that fasting is essential for the benefits of CR on survival in male 3xTg mice. Overall, our results demonstrate that fasting is required for the full benefits of a CR diet on the development and progression of AD in 3xTg mice, and suggest that both when and how much we eat influences the development and progress of AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Diana Vertein
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sophia Schlorf
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Julia Illiano
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yang Liu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Endocrinology and Reproductive Physiology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael J. Rigby
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David A. Harris
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, Madison, WI 53705, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
30
|
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: 3] [Impact Index Per Article: 3.0] [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
|
31
|
Al-Diab O, Sünkel C, Blanc E, Catar RA, Ashraf MI, Zhao H, Wang P, Rinschen MM, Fritsche-Guenther R, Grahammer F, Bachmann S, Beule D, Kirwan JA, Rajewsky N, Huber TB, Gürgen D, Kusch A. Sex-specific molecular signature of mouse podocytes in homeostasis and in response to pharmacological challenge with rapamycin. Biol Sex Differ 2024; 15:72. [PMID: 39278930 PMCID: PMC11404044 DOI: 10.1186/s13293-024-00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/30/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Sex differences exist in the prevalence and progression of major glomerular diseases. Podocytes are the essential cell-type in the kidney which maintain the physiological blood-urine barrier, and pathological changes in podocyte homeostasis are critical accelerators of impairment of kidney function. However, sex-specific molecular signatures of podocytes under physiological and stress conditions remain unknown. This work aimed at identifying sexual dimorphic molecular signatures of podocytes under physiological condition and pharmacologically challenged homeostasis with mechanistic target of rapamycin (mTOR) inhibition. mTOR is a crucial regulator involved in a variety of physiological and pathological stress responses in the kidney and inhibition of this pathway may therefore serve as a general stress challenger to get fundamental insights into sex differences in podocytes. METHODS The genomic ROSAmT/mG-NPHS2 Cre mouse model was used which allows obtaining highly pure podocyte fractions for cell-specific molecular analyses, and vehicle or pharmacologic treatment with the mTOR inhibitor rapamycin was performed for 3 weeks. Subsequently, deep RNA sequencing and proteomics were performed of the isolated podocytes to identify intrinsic sex differences. Studies were supplemented with metabolomics from kidney cortex tissues. RESULTS Although kidney function and morphology remained normal in all experimental groups, RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways. Interestingly, rapamycin abolished prominent sex-specific clustering of podocyte gene expression and induced major changes only in male transcriptome. Several sex-biased transcription factors could be identified as possible upstream regulators of these sexually dimorphic responses. Concordant to transcriptomics, metabolomic changes were more prominent in males. Remarkably, high number of previously reported kidney disease genes showed intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. CONCLUSIONS Our results highlight remarkable intrinsic sex-differences and sex-specific response patterns towards pharmacological challenged podocyte homeostasis which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. This work provides rationale and an in-depth database for novel targets to be tested in specific kidney disease models to advance with sex-specific treatment strategies.
Collapse
Affiliation(s)
- Ola Al-Diab
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christin Sünkel
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Rusan Ali Catar
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pinchao Wang
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Markus M Rinschen
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Raphaela Fritsche-Guenther
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jennifer A Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Dennis Gürgen
- Experimental Pharmacology & Oncology Berlin-Buch GmbH, 13125 Berlin-Buch, Germany
| | - Angelika Kusch
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- BIH Biomedical Innovation Academy (BIA), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
32
|
Calubag MF, Robbins PD, Lamming DW. A nutrigeroscience approach: Dietary macronutrients and cellular senescence. Cell Metab 2024; 36:1914-1944. [PMID: 39178854 PMCID: PMC11386599 DOI: 10.1016/j.cmet.2024.07.025] [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: 03/22/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Cellular senescence, a process in which a cell exits the cell cycle in response to stressors, is one of the hallmarks of aging. Senescence and the senescence-associated secretory phenotype (SASP)-a heterogeneous set of secreted factors that disrupt tissue homeostasis and promote the accumulation of senescent cells-reprogram metabolism and can lead to metabolic dysfunction. Dietary interventions have long been studied as methods to combat age-associated metabolic dysfunction, promote health, and increase lifespan. A growing body of literature suggests that senescence is responsive to diet, both to calories and specific dietary macronutrients, and that the metabolic benefits of dietary interventions may arise in part through reducing senescence. Here, we review what is currently known about dietary macronutrients' effect on senescence and the SASP, the nutrient-responsive molecular mechanisms that may mediate these effects, and the potential for these findings to inform the development of a nutrigeroscience approach to healthy aging.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul D Robbins
- Institute On the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
33
|
Zhang R, Chen S, Zhao F, Wang W, Liu D, Chen L, Bai T, Wu Z, Ji L, Zhang J. Sulforaphane enhanced muscle growth by promoting lipid oxidation through modulating key signaling pathways. Biosci Rep 2024; 44:BSR20240084. [PMID: 38868980 PMCID: PMC11224001 DOI: 10.1042/bsr20240084] [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: 01/24/2024] [Revised: 05/29/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024] Open
Abstract
Sulforaphane (SFN) has shown diverse effects on human health and diseases. SFN was administered daily to C57BL/6J mice at doses of 1 mg/kg (SFN1) and 3 mg/kg (SFN3) for 8 weeks. Both doses of SFN accelerated body weight increment. The cross-sectional area and diameter of Longissimus dorsi (LD) muscle fibers were enlarged in SFN3 group. Triglyceride (TG) and total cholesterol (TC) levels in LD muscle were decreased in SFN groups. RNA sequencing results revealed that 2455 and 2318 differentially expressed genes (DEGs) were found in SFN1 and SFN3 groups, respectively. Based on GO enrichment analysis, 754 and 911 enriched GO terms in the SFN1 and SFN3 groups, respectively. KEGG enrichment analysis shown that one KEGG pathway was enriched in the SFN1 group, while six KEGG pathways were enriched in the SFN3 group. The expressions of nine selected DEGs validated with qRT-PCR were in line with the RNA sequencing data. Furthermore, SFN treatment influenced lipid and protein metabolism related pathways including AMPK signaling, fatty acid metabolism signaling, cholesterol metabolism signalling, PPAR signaling, peroxisome signaling, TGFβ signaling, and mTOR signaling. In summary, SFN elevated muscle fibers size and reduced TG and TC content of in LD muscle by modulating protein and lipid metabolism-related signaling pathways.
Collapse
Affiliation(s)
- Rui Zhang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Suqin Chen
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Feng Zhao
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Wang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Dayu Liu
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Lin Chen
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Ting Bai
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Zhoulin Wu
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Lili Ji
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Jiamin Zhang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| |
Collapse
|
34
|
Li Y, Lin Y, Tang Y, Jiang M, Chen X, Chen H, Nie Q, Wu J, Tong X, Li J, Yu L, Hou J, Guo W, Chen L, Chen M, Zhang J, Lin S, Fu F, Wang C. MAZ-mediated up-regulation of BCKDK reprograms glucose metabolism and promotes growth by regulating glucose-6-phosphate dehydrogenase stability in triple-negative breast cancer. Cell Death Dis 2024; 15:516. [PMID: 39025830 PMCID: PMC11258276 DOI: 10.1038/s41419-024-06835-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: 01/11/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Tumour metabolic reprogramming is pivotal for tumour survival and proliferation. Investigating potential molecular mechanisms within the heterogeneous and clinically aggressive triple-negative breast cancer (TNBC) subtype is essential to identifying novel therapeutic targets. Accordingly, we investigated the role of branched-chain α-keto acid dehydrogenase kinase (BCKDK) in promoting tumorigenesis in TNBC. We analysed The Cancer Genome Atlas dataset and immunohistochemically stained surgical specimens to investigate BCKDK expression and its prognostic implications in TNBC. The effects of BCKDK on tumorigenesis were assessed using cell viability, colony formation, apoptosis, and cell cycle assays, and subsequently validated in vivo. Metabolomic screening was performed via isotope tracer studies. The downstream target was confirmed using mass spectrometry and a co-immunoprecipitation experiment coupled with immunofluorescence analysis. Upstream transcription factors were also examined using chromatin immunoprecipitation and luciferase assays. BCKDK was upregulated in TNBC tumour tissues and associated with poor prognosis. BCKDK depletion led to reduced cell proliferation both in vitro and vivo. MYC-associated zinc finger protein (MAZ) was confirmed as the major transcription factor directly regulating BCKDK expression in TNBC. Mechanistically, BCKDK interacted with glucose-6-phosphate dehydrogenase (G6PD), leading to increased flux in the pentose phosphate pathway for macromolecule synthesis and detoxification of reactive oxygen species. Forced expression of G6PD rescued the growth defect in BCKDK-deficient cells. Notably, the small-molecule inhibitor of BCKDK, 3,6-dichlorobenzo(b)thiophene-2-carboxylic acid, exhibited anti-tumour effects in a patient-derived tumour xenograft model. Our findings hold significant promise for developing targeted therapies aimed at disrupting the MAZ/BCKDK/G6PD signalling pathway, offering potential advancements in treating TNBC through metabolic reprogramming.
Collapse
Affiliation(s)
- Yan Li
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yali Tang
- School of Life Sciences, Xiamen University, Xiamen, Fujian Province, China
| | - Meichen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Xiaobin Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hanxi Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qian Nie
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jinqiao Wu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xin Tong
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jing Li
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Liuwen Yu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jialin Hou
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wenhui Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lili Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Minyan Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Shuhai Lin
- School of Life Sciences, Xiamen University, Xiamen, Fujian Province, China.
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China.
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China.
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
35
|
Bruschi M, Granata S, Candiano G, Petretto A, Bartolucci M, Kajana X, Spinelli S, Verlato A, Provenzano M, Zaza G. Proteomic Changes Induced by the Immunosuppressant Everolimus in Human Podocytes. Int J Mol Sci 2024; 25:7336. [PMID: 39000447 PMCID: PMC11242170 DOI: 10.3390/ijms25137336] [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: 06/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
mTOR inhibitors (mTOR-Is) may induce proteinuria in kidney transplant recipients through podocyte damage. However, the mechanism has only been partially defined. Total cell lysates and supernatants of immortalized human podocytes treated with different doses of everolimus (EVE) (10, 100, 200, and 500 nM) for 24 h were subjected to mass spectrometry-based proteomics. Support vector machine and partial least squares discriminant analysis were used for data analysis. The results were validated in urine samples from 28 kidney transplant recipients receiving EVE as part of their immunosuppressive therapy. We identified more than 7000 differentially expressed proteins involved in several pathways, including kinases, cell cycle regulation, epithelial-mesenchymal transition, and protein synthesis, according to gene ontology. Among these, after statistical analysis, 65 showed an expression level significantly and directly correlated with EVE dosage. Polo-Like Kinase 1 (PLK1) content was increased, whereas osteopontin (SPP1) content was reduced in podocytes and supernatants in a dose-dependent manner and significantly correlated with EVE dose (p < 0.0001, FDR < 5%). Similar results were obtained in the urine of kidney transplant patients. This study analyzed the impact of different doses of mTOR-Is on podocytes, helping to understand not only the biological basis of their therapeutic effects but also the possible mechanisms underlying proteinuria.
Collapse
Affiliation(s)
- Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Simona Granata
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Andrea Petretto
- Proteomics and Clinical Metabolomics Unit at the Core Facilities, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (A.P.); (M.B.)
| | - Martina Bartolucci
- Proteomics and Clinical Metabolomics Unit at the Core Facilities, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (A.P.); (M.B.)
| | - Xhuliana Kajana
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.B.); (G.C.); (X.K.); (S.S.)
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University Hospital of Verona, 37124 Verona, Italy;
| | - Michele Provenzano
- Nephrology, Dialysis and Transplantation Unit, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| |
Collapse
|
36
|
Sonsalla MM, Babygirija R, Johnson M, Cai S, Cole M, Yeh CY, Grunow I, Liu Y, Vertein D, Calubag MF, Trautman ME, Green CL, Rigby MJ, Puglielli L, Lamming DW. Acarbose ameliorates Western diet-induced metabolic and cognitive impairments in the 3xTg mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600472. [PMID: 39005334 PMCID: PMC11244897 DOI: 10.1101/2024.06.27.600472] [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
Age is the greatest risk factor for Alzheimer's disease (AD) as well as for other disorders that increase the risk of AD such as diabetes and obesity. There is growing interest in determining if interventions that promote metabolic health can prevent or delay AD. Acarbose is an anti-diabetic drug that not only improves glucose homeostasis, but also extends the lifespan of wild-type mice. Here, we test the hypothesis that acarbose will not only preserve metabolic health, but also slow or prevent AD pathology and cognitive deficits in 3xTg mice, a model of AD, fed either a Control diet or a high-fat, high-sucrose Western diet (WD). We find that acarbose decreases the body weight and adiposity of WD-fed 3xTg mice, increasing energy expenditure while also stimulating food consumption, and improves glycemic control. Both male and female WD-fed 3xTg mice have worsened cognitive deficits than Control-fed mice, and these deficits are ameliorated by acarbose treatment. Molecular and histological analysis of tau and amyloid pathology identified sex-specific effects of acarbose which are uncoupled from the dramatic improvements in cognition, suggesting that the benefits of acarbose on AD are largely driven by improved metabolic health. In conclusion, our results suggest that acarbose may be a promising intervention to prevent, delay, or even treat AD, especially in individuals consuming a Western diet.
Collapse
|
37
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-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: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
38
|
Xu D, Yin S, Shu Y. NF2: An underestimated player in cancer metabolic reprogramming and tumor immunity. NPJ Precis Oncol 2024; 8:133. [PMID: 38879686 PMCID: PMC11180135 DOI: 10.1038/s41698-024-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024] Open
Abstract
Neurofibromatosis type 2 (NF2) is a tumor suppressor gene implicated in various tumors, including mesothelioma, schwannomas, and meningioma. As a member of the ezrin, radixin, and moesin (ERM) family of proteins, merlin, which is encoded by NF2, regulates diverse cellular events and signalling pathways, such as the Hippo, mTOR, RAS, and cGAS-STING pathways. However, the biological role of NF2 in tumorigenesis has not been fully elucidated. Furthermore, cross-cancer mutations may exert distinct biological effects on tumorigenesis and treatment response. In addition to the functional inactivation of NF2, the codeficiency of other genes, such as cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B), BRCA1-associated protein-1 (BAP1), and large tumor suppressor 2 (LATS2), results in unique tumor characteristics that should be considered in clinical treatment decisions. Notably, several recent studies have explored the metabolic and immunological features associated with NF2, offering potential insights into tumor biology and the development of innovative therapeutic strategies. In this review, we consolidate the current knowledge on NF2 and examine the potential connection between cancer metabolism and tumor immunity in merlin-deficient malignancies. This review may provide a deeper understanding of the biological roles of NF2 and guide possible therapeutic avenues.
Collapse
Affiliation(s)
- Duo Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyuan Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
39
|
Lin Y, Huang H, Cao J, Zhang K, Chen R, Jiang J, Yi X, Feng S, Liu J, Zheng S, Ling Q. An integrated proteomics and metabolomics approach to assess graft quality and predict early allograft dysfunction after liver transplantation: a retrospective cohort study. Int J Surg 2024; 110:3480-3494. [PMID: 38502860 PMCID: PMC11175820 DOI: 10.1097/js9.0000000000001292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Early allograft dysfunction (EAD) is a common complication after liver transplantation (LT) and is associated with poor prognosis. Graft itself plays a major role in the development of EAD. We aimed to reveal the EAD-specific molecular profiles to assess graft quality and establish EAD predictive models. METHODS A total of 223 patients who underwent LT were enrolled and divided into training ( n =73) and validation ( n =150) sets. In the training set, proteomics was performed on graft biopsies, together with metabolomics on paired perfusates. Differential expression, enrichment analysis, and protein-protein interaction network were used to identify the key molecules and pathways involved. EAD predictive models were constructed using machine learning and verified in the validation set. RESULTS A total of 335 proteins were differentially expressed between the EAD and non-EAD groups. These proteins were significantly enriched in triglyceride and glycerophospholipid metabolism, neutrophil degranulation, and the MET-related signaling pathway. The top 12 graft proteins involved in the aforementioned processes were identified, including GPAT1, LPIN3, TGFB1, CD59, and SOS1. Moreover, downstream metabolic products, such as lactate dehydrogenase, interleukin-8, triglycerides, and the phosphatidylcholine/phosphorylethanolamine ratio in the paired perfusate displayed a close relationship with the graft proteins. To predict the occurrence of EAD, an integrated model using perfusate metabolic products and clinical parameters showed areas under the curve of 0.915 and 0.833 for the training and validation sets, respectively. It displayed superior predictive efficacy than that of currently existing models, including donor risk index and D-MELD scores. CONCLUSIONS We identified novel biomarkers in both grafts and perfusates that could be used to assess graft quality and provide new insights into the etiology of EAD. Herein, we also offer a valid tool for the early prediction of EAD.
Collapse
Affiliation(s)
- Yimou Lin
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Haitao Huang
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiaying Cao
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Ke Zhang
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Ruihan Chen
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Jingyu Jiang
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Xuewen Yi
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Shi Feng
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jimin Liu
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Shusen Zheng
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, Hangzhou, China
| | - Qi Ling
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, Hangzhou, China
| |
Collapse
|
40
|
Zhang W, Sha Z, Tang Y, Jin C, Gao W, Chen C, Yu L, Lv N, Liu S, Xu F, Wang D, Shi L. Defective Lamtor5 Leads to Autoimmunity by Deregulating v-ATPase and Lysosomal Acidification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400446. [PMID: 38639386 PMCID: PMC11165510 DOI: 10.1002/advs.202400446] [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: 01/12/2024] [Revised: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Despite accumulating evidence linking defective lysosome function with autoimmune diseases, how the catabolic machinery is regulated to maintain immune homeostasis remains unknown. Late endosomal/lysosomal adaptor, MAPK and mTOR activator 5 (Lamtor5) is a subunit of the Ragulator mediating mechanistic target of rapamycin complex 1 (mTORC1) activation in response to amino acids, but its action mode and physiological role are still unclear. Here it is demonstrated that Lamtor5 level is markedly decreased in peripheral blood mononuclear cells (PBMCs) of patients with systemic lupus erythematosus (SLE). In parallel, the mice with myeloid Lamtor5 ablation developed SLE-like manifestation. Impaired lysosomal function and aberrant activation of mTORC1 are evidenced in Lamtor5 deficient macrophages and PBMCs of SLE patients, accompanied by blunted autolysosomal pathway and undesirable inflammatory responses. Mechanistically, it is shown that Lamtor5 is physically associated with ATP6V1A, an essential subunit of vacuolar H+-ATPase (v-ATPase), and promoted the V0/V1 holoenzyme assembly to facilitate lysosome acidification. The binding of Lamtor5 to v-ATPase affected the lysosomal tethering of Rag GTPase and weakened its interaction with mTORC1 for activation. Overall, Lamtor5 is identified as a critical factor for immune homeostasis by intergrading v-ATPase activity, lysosome function, and mTOR pathway. The findings provide a potential therapeutic target for SLE and/or other autoimmune diseases.
Collapse
Affiliation(s)
- Wei Zhang
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Zhou Sha
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Yunzhe Tang
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Cuiyuan Jin
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhou310022China
| | - Wenhua Gao
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Changmai Chen
- School of PharmacyFujian Medical UniversityFuzhou350122China
| | - Lang Yu
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Nianyin Lv
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Shijia Liu
- The Affiliated Hospital of Nanjing University of Chinese MedicineNanjing210029China
| | - Feng Xu
- Department of Infectious DiseasesThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Dandan Wang
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210093China
| | - Liyun Shi
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhou310022China
| |
Collapse
|
41
|
Sun L, Jin Y, Nishio M, Watanabe M, Kamakura T, Nagata S, Fukuda M, Maekawa H, Kawai S, Yamamoto T, Toguchida J. Oxidative phosphorylation is a pivotal therapeutic target of fibrodysplasia ossificans progressiva. Life Sci Alliance 2024; 7:e202302219. [PMID: 38365425 PMCID: PMC10875110 DOI: 10.26508/lsa.202302219] [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: 06/15/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024] Open
Abstract
Heterotopic ossification (HO) is a non-physiological bone formation where soft tissue progenitor cells differentiate into chondrogenic cells. In fibrodysplasia ossificans progressiva (FOP), a rare genetic disease characterized by progressive and systemic HO, the Activin A/mutated ACVR1/mTORC1 cascade induces HO in progenitors in muscle tissues. The relevant biological processes aberrantly regulated by activated mTORC1 remain unclear, however. RNA-sequencing analyses revealed the enrichment of genes involved in oxidative phosphorylation (OXPHOS) during Activin A-induced chondrogenesis of mesenchymal stem cells derived from FOP patient-specific induced pluripotent stem cells. Functional analyses showed a metabolic transition from glycolysis to OXPHOS during chondrogenesis, along with increased mitochondrial biogenesis. mTORC1 inhibition by rapamycin suppressed OXPHOS, whereas OXPHOS inhibitor IACS-010759 inhibited cartilage matrix formation in vitro, indicating that OXPHOS is principally involved in mTORC1-induced chondrogenesis. Furthermore, IACS-010759 inhibited the muscle injury-induced enrichment of fibro/adipogenic progenitor genes and HO in transgenic mice carrying the mutated human ACVR1. These data indicated that OXPHOS is a critical downstream mediator of mTORC1 signaling in chondrogenesis and therefore is a potential FOP therapeutic target.
Collapse
Affiliation(s)
- Liping Sun
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yonghui Jin
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Megumi Nishio
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Makoto Watanabe
- Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Takeshi Kamakura
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sanae Nagata
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Masayuki Fukuda
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hirotsugu Maekawa
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Shunsuke Kawai
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Kyoto, Japan
| | - Junya Toguchida
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| |
Collapse
|
42
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- 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
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - 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
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
43
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- 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
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - 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
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
44
|
Wu W, Jin Q, Östlund C, Tanji K, Shin JY, Han J, Leu CS, Kushner J, Worman HJ. mTOR Inhibition Prolongs Survival and Has Beneficial Effects on Heart Function After Onset of Lamin A/C Gene Mutation Cardiomyopathy in Mice. Circ Heart Fail 2024; 17:e011110. [PMID: 38567527 PMCID: PMC11008450 DOI: 10.1161/circheartfailure.123.011110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 02/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Mutations in LMNA encoding nuclear envelope proteins lamin A/C cause dilated cardiomyopathy. Activation of the AKT/mTOR (RAC-α serine/threonine-protein kinase/mammalian target of rapamycin) pathway is implicated as a potential pathophysiologic mechanism. The aim of this study was to assess whether pharmacological inhibition of mTOR signaling has beneficial effects on heart function and prolongs survival in a mouse model of the disease, after onset of heart failure. METHODS We treated male LmnaH222P/H222P mice, after the onset of heart failure, with placebo or either of 2 orally bioavailable mTOR inhibitors: everolimus or NV-20494, a rapamycin analog highly selective against mTORC1. We examined left ventricular remodeling, and the cell biological, biochemical, and histopathologic features of cardiomyopathy, potential drug toxicity, and survival. RESULTS Everolimus treatment (n=17) significantly reduced left ventricular dilatation and increased contractility on echocardiography, with a 7% (P=0.018) reduction in left ventricular end-diastolic diameter and a 39% (P=0.0159) increase fractional shortening compared with placebo (n=17) after 6 weeks of treatment. NV-20494 treatment (n=15) yielded similar but more modest and nonsignificant changes. Neither drug prevented the development of cardiac fibrosis. Drug treatment reactivated suppressed autophagy and inhibited mTORC1 signaling in the heart, although everolimus was more potent. With regards to drug toxicity, everolimus alone led to a modest degree of glucose intolerance during glucose challenge. Everolimus (n=20) and NV-20494 (n=20) significantly prolonged median survival in LmnaH222P/H222P mice, by 9% (P=0.0348) and 11% (P=0.0206), respectively, compared with placebo (n=20). CONCLUSIONS These results suggest that mTOR inhibitors may be beneficial in patients with cardiomyopathy caused by LMNA mutations and that further study is warranted.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons (W.W., Q.J., C.Ö., K.T., H.J.W.), Columbia University, New York, NY
| | - Qi Jin
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons (W.W., Q.J., C.Ö., K.T., H.J.W.), Columbia University, New York, NY
| | - Cecilia Östlund
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons (W.W., Q.J., C.Ö., K.T., H.J.W.), Columbia University, New York, NY
| | - Kurenai Tanji
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons (W.W., Q.J., C.Ö., K.T., H.J.W.), Columbia University, New York, NY
| | - Ji-Yeon Shin
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
| | - Jiying Han
- Department of Biostatistics, Mailman School of Public Health (J.H., C.-S.L.), Columbia University, New York, NY
| | - Cheng-Shiun Leu
- Department of Biostatistics, Mailman School of Public Health (J.H., C.-S.L.), Columbia University, New York, NY
| | - Jared Kushner
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
| | - Howard J. Worman
- Department of Medicine, Vagelos College of Physicians and Surgeons, (W.W., Q.J., C.Ö., J.-Y.S., J.K., H.J.W.), Columbia University, New York, NY
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons (W.W., Q.J., C.Ö., K.T., H.J.W.), Columbia University, New York, NY
| |
Collapse
|
45
|
Szalai F, Sztankovics D, Krencz I, Moldvai D, Pápay J, Sebestyén A, Khoor A. Rictor-A Mediator of Progression and Metastasis in Lung Cancer. Cancers (Basel) 2024; 16:543. [PMID: 38339294 PMCID: PMC10854599 DOI: 10.3390/cancers16030543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lung carcinoma is one of the most common cancer types for both men and women. Despite recent breakthroughs in targeted therapy and immunotherapy, it is characterized by a high metastatic rate, which can significantly affect quality of life and prognosis. Rictor (encoded by the RICTOR gene) is known as a scaffold protein for the multiprotein complex mTORC2. Among its diverse roles in regulating essential cellular functions, mTORC2 also facilitates epithelial-mesenchymal transition and metastasis formation. Amplification of the RICTOR gene and subsequent overexpression of the Rictor protein can result in the activation of mTORC2, which promotes cell survival and migration. Based on recent studies, RICTOR amplification or Rictor overexpression can serve as a marker for mTORC2 activation, which in turn provides a promising druggable target. Although selective inhibitors of Rictor and the Rictor-mTOR association are only in a preclinical phase, they seem to be potent novel approaches to reduce tumor cell migration and metastasis formation. Here, we summarize recent advances that support an important role for Rictor and mTORC2 as potential therapeutic targets in the treatment of lung cancer. This is a traditional (narrative) review based on Pubmed and Google Scholar searches for the following keywords: Rictor, RICTOR amplification, mTORC2, Rictor complexes, lung cancer, metastasis, progression, mTOR inhibitors.
Collapse
Affiliation(s)
- Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Andras Khoor
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
46
|
Chen J, Cui L, Lu S, Xu S. Amino acid metabolism in tumor biology and therapy. Cell Death Dis 2024; 15:42. [PMID: 38218942 PMCID: PMC10787762 DOI: 10.1038/s41419-024-06435-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Amino acid metabolism plays important roles in tumor biology and tumor therapy. Accumulating evidence has shown that amino acids contribute to tumorigenesis and tumor immunity by acting as nutrients, signaling molecules, and could also regulate gene transcription and epigenetic modification. Therefore, targeting amino acid metabolism will provide new ideas for tumor treatment and become an important therapeutic approach after surgery, radiotherapy, and chemotherapy. In this review, we systematically summarize the recent progress of amino acid metabolism in malignancy and their interaction with signal pathways as well as their effect on tumor microenvironment and epigenetic modification. Collectively, we also highlight the potential therapeutic application and future expectation.
Collapse
Affiliation(s)
- Jie Chen
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Likun Cui
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Shaoteng Lu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology, Naval Medical University/Second Military Medical University, Shanghai, 200433, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
47
|
Wang K, Zhang R, Lehwald N, Tao GZ, Liu B, Liu B, Koh Y, Sylvester KG. Wnt/β-catenin signaling activation promotes lipogenesis in the steatotic liver via physical mTOR interaction. Front Endocrinol (Lausanne) 2023; 14:1289004. [PMID: 38152126 PMCID: PMC10751342 DOI: 10.3389/fendo.2023.1289004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Background and aims Wnt/β-catenin signaling plays an important role in regulating hepatic metabolism. This study is to explore the molecular mechanisms underlying the potential crosstalk between Wnt/β-catenin and mTOR signaling in hepatic steatosis. Methods Transgenic mice (overexpress Wnt1 in hepatocytes, Wnt+) mice and wild-type littermates were given high fat diet (HFD) for 12 weeks to induce hepatic steatosis. Mouse hepatocytes cells (AML12) and those transfected to cause constitutive β-catenin stabilization (S33Y) were treated with oleic acid for lipid accumulation. Results Wnt+ mice developed more hepatic steatosis in response to HFD. Immunoblot shows a significant increase in the expression of fatty acid synthesis-related genes (SREBP-1 and its downstream targets ACC, AceCS1, and FASN) and a decrease in fatty acid oxidation gene (MCAD) in Wnt+ mice livers under HFD. Wnt+ mice also revealed increased Akt signaling and its downstream target gene mTOR in response to HFD. In vitro, increased lipid accumulation was detected in S33Y cells in response to oleic acid compared to AML12 cells reinforcing the in vivo findings. mTOR inhibition by rapamycin led to a down-regulation of fatty acid synthesis in S33Y cells. In addition, β-catenin has a physical interaction with mTOR as verified by co-immunoprecipitation in hepatocytes. Conclusions Taken together, our results demonstrate that β-catenin stabilization through Wnt signaling serves a central role in lipid metabolism in the steatotic liver through up-regulation of fatty acid synthesis via Akt/mTOR signaling. These findings suggest hepatic Wnt signaling may represent a therapeutic strategy in hepatic steatosis.
Collapse
Affiliation(s)
- Kewei Wang
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Rong Zhang
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Nadja Lehwald
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of General, Visceral and Pediatric Surgery, School of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Guo-Zhong Tao
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Bowen Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Bo Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Yangseok Koh
- Department of Surgery, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Karl G. Sylvester
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Metabolic Health Center, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
48
|
Elliehausen CJ, Anderson RM, Diffee GM, Rhoads TW, Lamming DW, Hornberger TA, Konopka AR. Geroprotector drugs and exercise: friends or foes on healthy longevity? BMC Biol 2023; 21:287. [PMID: 38066609 PMCID: PMC10709984 DOI: 10.1186/s12915-023-01779-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Physical activity and several pharmacological approaches individually combat age-associated conditions and extend healthy longevity in model systems. It is tantalizing to extrapolate that combining geroprotector drugs with exercise could extend healthy longevity beyond any individual treatment. However, the current dogma suggests that taking leading geroprotector drugs on the same day as exercise may limit several health benefits. Here, we review leading candidate geroprotector drugs and their interactions with exercise and highlight salient gaps in knowledge that need to be addressed to identify if geroprotector drugs can have a harmonious relationship with exercise.
Collapse
Affiliation(s)
- Christian J Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rozalyn M Anderson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Gary M Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam R Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
49
|
Trautman ME, Braucher LN, Elliehausen C, Zhu WG, Zelenovskiy E, Green M, Sonsalla MM, Yeh CY, Hornberger TA, Konopka AR, Lamming DW. Resistance exercise protects mice from protein-induced fat accretion. eLife 2023; 12:RP91007. [PMID: 38019262 PMCID: PMC10686620 DOI: 10.7554/elife.91007] [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] [Indexed: 11/30/2023] Open
Abstract
Low-protein (LP) diets extend the lifespan of diverse species and are associated with improved metabolic health in both rodents and humans. Paradoxically, many athletes and bodybuilders consume high-protein (HP) diets and protein supplements, yet are both fit and metabolically healthy. Here, we examine this paradox using weight pulling, a validated progressive resistance exercise training regimen, in mice fed either an LP diet or an isocaloric HP diet. We find that despite having lower food consumption than the LP group, HP-fed mice gain significantly more fat mass than LP-fed mice when not exercising, while weight pulling protected HP-fed mice from this excess fat accretion. The HP diet augmented exercise-induced hypertrophy of the forearm flexor complex, and weight pulling ability increased more rapidly in the exercised HP-fed mice. Surprisingly, exercise did not protect from HP-induced changes in glycemic control. Our results confirm that HP diets can augment muscle hypertrophy and accelerate strength gain induced by resistance exercise without negative effects on fat mass, and also demonstrate that LP diets may be advantageous in the sedentary. Our results highlight the need to consider both dietary composition and activity, not simply calories, when taking a precision nutrition approach to health.
Collapse
Affiliation(s)
- Michaela E Trautman
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
| | - Leah N Braucher
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Christian Elliehausen
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Wenyuan G Zhu
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Esther Zelenovskiy
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Madelyn Green
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
| | - Troy A Hornberger
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- School of Veterinary Medicine, University of Wisconsin-MadisonMadisonUnited States
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-MadisonMadisonUnited States
- William S. Middleton Memorial Veterans HospitalMadisonUnited States
- Nutrition and Metabolism Graduate Program, University of Wisconsin- MadisonMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- University of Wisconsin Carbone Cancer CenterMadisonUnited States
| |
Collapse
|
50
|
Arp NL, Seim GL, Votava JA, Josephson J, Fan J. Reactive nitrogen species inhibit branched chain alpha-ketoacid dehydrogenase complex and impact muscle cell metabolism. J Biol Chem 2023; 299:105333. [PMID: 37827290 PMCID: PMC10656228 DOI: 10.1016/j.jbc.2023.105333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Branched chain α-ketoacid dehydrogenase complex (BCKDC) is the rate-limiting enzyme in branched chain amino acid (BCAA) catabolism, a metabolic pathway with great importance for human health. BCKDC belongs to the mitochondrial α-ketoacid dehydrogenase complex family, which also includes pyruvate dehydrogenase complex and oxoglutarate dehydrogenase complex. Here, we revealed that BCKDC can be substantially inhibited by reactive nitrogen species (RNS) via a mechanism similar to what we recently discovered with pyruvate dehydrogenase complex and oxoglutarate dehydrogenase complex-RNS can cause inactivating covalent modifications of the lipoic arm on its E2 subunit. In addition, we showed that such reaction between RNS and the lipoic arm of the E2 subunit can further promote inhibition of the E3 subunits of α-ketoacid dehydrogenase complexes. We examined the impacts of this RNS-mediated BCKDC inhibition in muscle cells, an important site of BCAA metabolism, and demonstrated that the nitric oxide production induced by cytokine stimulation leads to a strong inhibition of BCKDC activity and BCAA oxidation in myotubes and myoblasts. More broadly, nitric oxide production reduced the level of functional lipoic arms across the multiple α-ketoacid dehydrogenases and led to intracellular accumulation of their substrates (α-ketoacids), decrease of their products (acyl-CoAs), and a lower cellular energy charge. In sum, this work revealed a new mechanism for BCKDC regulation, demonstrated that RNS can generally inhibit all α-ketoacid dehydrogenases, which has broad physiological implications across multiple cell types, and elucidated the mechanistic connection between RNS-driven inhibitory modifications on the E2 and E3 subunits of α-ketoacid dehydrogenases.
Collapse
Affiliation(s)
- Nicholas L Arp
- Morgridge Institute for Research, Madison, Wisconsin, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Gretchen L Seim
- Morgridge Institute for Research, Madison, Wisconsin, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - James A Votava
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | | | - Jing Fan
- Morgridge Institute for Research, Madison, Wisconsin, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|