451
|
Wu M, Li H, Sun X, Zhong R, Cai L, Chen R, Madeniyet M, Ren K, Peng Z, Yang Y, Chen W, Tu Y, Lai M, Deng J, Wu Y, Zhao S, Ruan Q, Rao M, Xie S, Ye Y, Wan J. Aerobic exercise prevents renal osteodystrophy via irisin-activated osteoblasts. JCI Insight 2025; 10:e184468. [PMID: 39883525 PMCID: PMC11949034 DOI: 10.1172/jci.insight.184468] [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: 07/03/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Renal osteodystrophy is commonly seen in patients with chronic kidney disease (CKD) due to disrupted mineral homeostasis. Given the impaired renal function in these patients, common antiresorptive agents, including bisphosphonates, must be used with caution or even contraindicated. Therefore, an alternative therapy without renal burden to combat renal osteodystrophy is urgently needed. Here, we report that clinically relevant aerobic exercise significantly prevents high-turnover renal osteodystrophy in CKD mice and patients with CKD without compromising renal function. Mechanistically, 4-week aerobic exercise in CKD mice increased expression of skeletal muscle PPARγ coactivator-1α (PGC-1α) and circulating irisin. Both exercise and irisin administration significantly activated osteoblasts, but not osteoclasts, via integrin αvβ5, thereby conferring bone quality benefits. Removal of irisin-influenced thermogenic adipose tissues or genetic ablation of uncoupling protein 1 did not alter the irisin-conferred antiosteodystrophy effect. Importantly, in a pilot clinical study, 12-week aerobic exercise in patients with high-grade CKD significantly increased circulating irisin and prevented osteodystrophy progression, without detectable renal burden. The combination of irisin and current antiresorptive agents effectively rescued renal osteodystrophy in mice. Our work provides mechanistic insights into the role of exercise and irisin in renal osteodystrophy, and it highlights a clinically relevant, low-cost, kidney-friendly therapy for patients with this devastating disease.
Collapse
Affiliation(s)
- Meng Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Huilan Li
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoting Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Rongrong Zhong
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Linli Cai
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ruibo Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Madiya Madeniyet
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kana Ren
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yujie Yang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Weiqin Chen
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yanling Tu
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Miaoxin Lai
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Jinxiu Deng
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuting Wu
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shumin Zhao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Qingyan Ruan
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Mei Rao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Sisi Xie
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, and
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
452
|
Zhu C, Zhang Z, Zhu Y, Du Y, Han C, Zhao Q, Li Q, Hou J, Zhang J, He W, Qin Y. Study on the role of Dihuang Yinzi in regulating the AMPK/SIRT1/PGC-1α pathway to promote mitochondrial biogenesis and improve Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118859. [PMID: 39341266 DOI: 10.1016/j.jep.2024.118859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dihuang Yinzi (DHYZ) is a classic prescription in traditional Chinese medicine. Its therapeutic effect on Alzheimer's disease (AD) has been widely validated. However, the underlying molecular mechanisms of DHYZ in AD treatment remain unclear and require further research. AIM OF THE STUDY Elucidating DHYZ's promotion of mitochondrial biogenesis through the AMPK/SIRT1/PGC-1α pathway improves neuronal loss, mitochondrial damage, and memory deficits in AD. MATERIALS AND METHODS Administering DHYZ by gavage to SAMP8 mice, after completing behavioral tests, the effects of DHYZ on hippocampal neuron loss and mitochondrial structural damage in AD model mice were assessed using Nissl staining and transmission electron microscopy. Western blot was used to detect the expression of mitochondrial biogenesis-related proteins PGC-1α, CREB, mitochondrial fusion protein MFN2, and mitochondrial fission proteins DRP1 and FIS1. At the same time, immunofluorescence (IF) was employed to measure the relative fluorescence intensity of mitochondrial fusion protein MFN1. After determining the optimal dose of DYHZ for treating AD, we conducted mechanistic studies. By intraperitoneally injecting SAMP8 mice with the AMPK inhibitor (Compound C) to inhibit AMPK protein expression and subsequently treating them with DHYZ, the impact of DHYZ on hippocampal neurons in AD model mice was evaluated using Nissl and hematoxylin-eosin staining. Western blot was used to detect the protein expression of AMPK, p-AMPK, SIRT1, PGC-1α, NRF1, and TFAM. In contrast, IF was used to measure the relative fluorescence intensity of PGC-1α, NRF1, and TFAM proteins in the hippocampal CA1 region. RESULTS DHYZ significantly improved AD model mice's cognitive impairment and memory deficits and mitigated hippocampal neuron loss and degeneration. Additionally, it ameliorated mitochondrial morphological structures. DHYZ upregulated the protein expression of mitochondrial biogenesis-related proteins PGC-1α, CREB, and mitochondrial fusion proteins MFN1 and MFN2 while inhibiting the expression of mitochondrial fission proteins DRP1 and FIS1. Further studies revealed that DHYZ could upregulate the expression of the AMPK/SIRT1/PGC-1α pathway proteins and their downstream proteins NRF1 and TFAM. CONCLUSION DHYZ promotes mitochondrial biogenesis by activating the AMPK/SIRT1/PGC-1α signaling pathway, thereby improving memory deficits, neuronal loss, and mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Chao Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Yuzhong Du
- School of Pharmaceutical Sciences, Shanxi Medical University, Jinzhong, Shanxi, 030607, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Jiangqi Hou
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China.
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China.
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, Shanxi, 030619, China; National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, Shanxi, 030619, China; Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi, 030619, China.
| |
Collapse
|
453
|
Karbanová J, Thamm K, Fargeas CA, Deniz IA, Lorico A, Corbeil D. Prominosomes - a particular class of extracellular vesicles containing prominin-1/CD133? J Nanobiotechnology 2025; 23:61. [PMID: 39881297 PMCID: PMC11776279 DOI: 10.1186/s12951-025-03102-w] [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: 09/17/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Extracellular membrane vesicles (EVs) offer promising values in various medical fields, e.g., as biomarkers in liquid biopsies or as native (or bioengineered) biological nanocarriers in tissue engineering, regenerative medicine and cancer therapy. Based on their cellular origin EVs can vary considerably in composition and diameter. Cell biological studies on mammalian prominin-1, a cholesterol-binding membrane glycoprotein, have helped to reveal new donor membranes as sources of EVs. For instance, small EVs can originate from microvilli and primary cilia, while large EVs might be produced by transient structures such as retracting cellular extremities of cancer cells during the mitotic rounding process, and the midbody at the end of cytokinesis. Here, we will highlight the various subcellular origins of prominin-1+ EVs, also called prominosomes, and the potential mechanism(s) regulating their formation. We will further discuss the molecular and cellular characteristics of prominin-1, notably those that have a direct effect on the release of prominin-1+ EVs, a process that might be directly implicated in donor cell reprogramming of stem and cancer stem cells. Prominin-1+ EVs also mediate intercellular communication during embryonic development and adult homeostasis in healthy individuals, while disseminating biological information during diseases.
Collapse
Affiliation(s)
- Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| | - Kristina Thamm
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- denovoMATRIX GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Christine A Fargeas
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ilker A Deniz
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV, 89014, USA
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| |
Collapse
|
454
|
Xia M, Li Y, Liu Y, Dong Z, Liu H. Single-Cell RNA-Sequencing Analysis Provides Insights into IgA Nephropathy. Biomolecules 2025; 15:191. [PMID: 40001494 PMCID: PMC11853383 DOI: 10.3390/biom15020191] [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: 11/19/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
IgA nephropathy (IgAN) is currently the most common primary glomerular disease worldwide, and early diagnosis and intervention contribute significantly to improving outcomes and reducing the incidence of renal failure. The pathogenesis of IgAN remains incompletely understood. In recent years, the rapid development of single-cell RNA-sequencing (scRNA-seq) technology has provided the high-resolution and rich data necessary to elucidate disease characteristics and enabled the analysis of complex interactions between individual cells and cell types. The application of scRNA-seq in IgAN successfully revealed the landscape of immunological features, including peripheral blood B-cell and Th-cell activation, cytotoxic T-cell depletion, and renal infiltrating cell subtypes, as well as the crucial role of mesangial cells and endothelial cells in the early stage of kidney injury, and also revealed the extensive intercellular interactions between renal cells. Here, we discuss the research progress of scRNA-seq technology in IgAN. These scRNA-seq profiles help us to understand the complex molecular mechanisms of IgAN and develop biomarkers and specific therapeutic strategies.
Collapse
Affiliation(s)
- Ming Xia
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; (M.X.); (Y.L.); (Y.L.); (Z.D.)
| | - Yifu Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; (M.X.); (Y.L.); (Y.L.); (Z.D.)
- Center for Medical Research, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; (M.X.); (Y.L.); (Y.L.); (Z.D.)
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; (M.X.); (Y.L.); (Y.L.); (Z.D.)
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
| | - Hong Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha 410011, China; (M.X.); (Y.L.); (Y.L.); (Z.D.)
| |
Collapse
|
455
|
Farella I, Chiarito M, Vitale R, D’Amato G, Faienza MF. The "Burden" of Childhood Obesity on Bone Health: A Look at Prevention and Treatment. Nutrients 2025; 17:491. [PMID: 39940349 PMCID: PMC11821239 DOI: 10.3390/nu17030491] [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: 01/12/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Childhood obesity represents a multifaceted challenge to bone health, influenced by a combination of endocrine, metabolic, and mechanical factors. Excess body fat correlates with an increase in bone mineral density (BMD) yet paradoxically elevates fracture risk due to compromised bone quality and increased mechanical loading on atypical sites. Additionally, subjects with syndromic obesity, as well as individuals with atypical nutritional patterns, including those with eating disorders, show bone fragility through unique genetic and hormonal dysregulations. Emerging evidence underscores the adverse effects of new pharmacological treatments for severe obesity on bone health. Novel drugs, such as glucagon-like peptide-1 (GLP-1) receptor agonists, and bariatric surgery demonstrate potential in achieving weight loss, though limited evidence is available regarding their short- and long-term impacts on skeletal health. This review provides a comprehensive analysis of the mechanisms underlying the impact of childhood obesity on bone health. It critically appraises evidence from in vitro studies, animal models, and clinical research in children with exogenous obesity, syndromic obesity, and eating disorders. It also explores the effects of emerging pharmacological and surgical treatments for severe obesity on skeletal integrity, highlights prevention strategies, and identifies research gaps.
Collapse
Affiliation(s)
- Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy;
| | - Mariangela Chiarito
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Rossella Vitale
- Giovanni XXIII Pediatric Hospital, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Gabriele D’Amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70012 Bari, Italy;
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
456
|
Tiwari RK, Rawat SG, Rai S, Kumar A. Stress regulatory hormones and cancer: the contribution of epinephrine and cancer therapeutic value of beta blockers. Endocrine 2025:10.1007/s12020-025-04161-7. [PMID: 39869294 DOI: 10.1007/s12020-025-04161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
The word "cancer" evokes myriad emotions, ranging from fear and despair to hope and determination. Cancer is aptly defined as a complex and multifaceted group of diseases that has unapologetically led to the loss of countless lives and affected innumerable families across the globe. The battle with cancer is not only a physical battle, but also an emotional, as well as a psychological skirmish for patients and for their loved ones. Cancer has been a part of our history, stories, and lives for centuries and has challenged the ingenuity of health and medical science, and the resilience of the human spirit. From the early days of surgery and radiation therapy to cutting-edge developments in chemotherapeutic agents, immunotherapy, and targeted treatments, the medical field continues to make significant headway in the fight against cancer. However, even after all these advancements, cancer is still among the leading cause of death globally. This urges us to understand the central hallmarks of neoplastic cells to identify novel molecular targets for the development of promising therapeutic approaches. Growing research suggests that stress mediators, including epinephrine, play a critical role in the development and progression of cancer by inducing neoplastic features through activating adrenergic receptors, particularly β-adrenoreceptors. Further, our experimental data has also shown that epinephrine mediates the growth of T-cell lymphoma by inducing proliferation, glycolysis, and apoptosis evasion via altering the expression levels of key regulators of these vital cellular processes. The beauty of receptor-based therapy lies in its precision and higher therapeutic value. Interestingly, the enhanced expression of β-adrenergic receptors (ADRBs), namely ADRB2 (β2-adrenoreceptor) and ADRB3 (β3-adrenoreceptor) has been noted in many cancers, such as breast, colon, gastric, pancreatic, and prostate and has been reported to play a pivotal role in facilitating cancer growth mainly by promoting proliferation, evasion of apoptosis, angiogenesis, invasion and metastasis, and chemoresistance. The present review article is an attempt to summarize the available findings which indicate a distinct relationship between stress hormones and cancer, with a special emphasis on epinephrine, considered as a key stress regulatory molecule. This article also discusses the possibility of using beta-blockers for cancer therapy.
Collapse
Affiliation(s)
- Rajan Kumar Tiwari
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- School of Medicine and Health Sciences, The George Washington University, Washington DC, USA
| | - Shiv Govind Rawat
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- MD Anderson Cancer Center, The University of Texas, Texas, USA
| | - Siddharth Rai
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
457
|
Slattery JD, Rambousek JR, Tsui E, Honeycutt MK, Goldberg M, Graham JL, Wietecha TA, Wolden-Hanson T, Williams AL, O'Brien KD, Havel PJ, Blevins JE. Effects of systemic oxytocin and beta-3 receptor agonist (CL 316243) treatment on body weight and adiposity in male diet-induced obese rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.27.615550. [PMID: 39502365 PMCID: PMC11537314 DOI: 10.1101/2024.09.27.615550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Previous studies have implicated hindbrain oxytocin (OT) receptors in the control of food intake and brown adipose tissue (BAT) thermogenesis. We recently demonstrated that hindbrain [fourth ventricle (4V)] administration of oxytocin (OT) could be used as an adjunct to drugs that directly target beta-3 adrenergic receptors (β3-AR) to elicit weight loss in diet-induced obese (DIO) rodents. What remains unclear is whether systemic OT can be used as an adjunct with the β3-AR agonist, CL 316243, to increase BAT thermogenesis and elicit weight loss in DIO rats. We hypothesized that systemic OT and β3-AR agonist (CL 316243) treatment would produce an additive effect to reduce body weight and adiposity in DIO rats by decreasing food intake and stimulating BAT thermogenesis. To test this hypothesis, we determined the effects of systemic (subcutaneous) infusions of OT (50 nmol/day) or vehicle (VEH) when combined with daily systemic (intraperitoneal) injections of CL 316243 (0.5 mg/kg) or VEH on body weight, adiposity, food intake and brown adipose tissue temperature (TIBAT). OT and CL 316243 monotherapy decreased body weight by 8.0±0.9% (P<0.05) and 8.6±0.6% (P<0.05), respectively, but OT in combination with CL 316243 produced more substantial weight loss (14.9±1.0%; P<0.05) compared to either treatment alone. These effects were associated with decreased adiposity, energy intake and elevated TIBAT during the treatment period. The findings from the current study suggest that the effects of systemic OT and CL 316243 to elicit weight loss are additive and appear to be driven primarily by OT-elicited changes in food intake and CL 316243-elicited increases in BAT thermogenesis.
Collapse
Affiliation(s)
- Jared D Slattery
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - June R Rambousek
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Edison Tsui
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Mackenzie K Honeycutt
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Matvey Goldberg
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - James L Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Tomasz A Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Amber L Williams
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Kevin D O'Brien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
458
|
Xu Y, Zhang Y, Sun W, Tang Q, Feng W, Xiao H, Wang J, Yuan X, Xiang M, Gao Y, Zhang H, Lu J. Characteristics of different lipid droplet-mitochondrial contacts patterns during lipid droplet metabolism in T2DM-induced MASLD. Sci Rep 2025; 15:3399. [PMID: 39870911 PMCID: PMC11772659 DOI: 10.1038/s41598-025-87871-2] [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: 06/07/2024] [Accepted: 01/22/2025] [Indexed: 01/29/2025] Open
Abstract
Mitochondrial function is crucial for hepatic lipid metabolism. Current research identifies two types of mitochondria based on their contact with lipid droplets: peridroplet mitochondria (PDM) and cytoplasmic mitochondria (CM). This work aimed to investigate the alterations of CM and PDM in metabolic dysfunction-associated steatotic liver disease (MASLD) induced by spontaneous type-2 diabetes mellitus (T2DM) in db/db mice. It was found that insulin resistance increased both the number and size of lipid droplets in the liver by enhancing the accumulation of free fatty acids, which is accompanied by an increase in contacts with mitochondria. We described the different patterns of tight contacts between small lipid droplets and mitochondria in purified CM and PDM by examining their oxidation states and morphological characteristics. In CM, enhanced fatty acid oxidation resulted in elongated mitochondria that surrounded single small lipid droplets and were responsible for lipid droplet consumption, while in PDM, increased substrates for lipid synthesis promoted lipid droplet expansion with the assistance of the endoplasmic reticulum. These data show the different ways in which mitochondrial contact with lipid droplets could provide new insights for future research on liver lipid metabolism.
Collapse
Affiliation(s)
- Ye Xu
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Yuan Zhang
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China.
- Jiangsu Collaborative Innovation Center for Sport and Health Project, Nanjing, China.
| | - Wen Sun
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Qiang Tang
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
- Jiangsu Collaborative Innovation Center for Sport and Health Project, Nanjing, China
| | - Wanyu Feng
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
- Sport Science Research Institute, Nanjing Sport Institute, Nanjing, China
| | - Hongjian Xiao
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Jingjie Wang
- Department of General Surgery, Nanjing Maternity and Child Health Care Hospital, Womens Hospital of Nanjing Medical University, Nanjing, China
| | - Xinmeng Yuan
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Mengqi Xiang
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Yaran Gao
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Hanyu Zhang
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China
| | - Jiao Lu
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China.
- Jiangsu Collaborative Innovation Center for Sport and Health Project, Nanjing, China.
| |
Collapse
|
459
|
Liu X, Sun X, Mu W, Li Y, Bu W, Yang T, Zhang J, Liu R, Ren J, Zhou J, Li P, Shi Y, Shao C. Autophagic flux-lipid droplet biogenesis cascade sustains mitochondrial fitness in colorectal cancer cells adapted to acidosis. Cell Death Discov 2025; 11:21. [PMID: 39856069 PMCID: PMC11761495 DOI: 10.1038/s41420-025-02301-6] [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: 07/25/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer development is associated with adaptation to various stressful conditions, such as extracellular acidosis. The adverse tumor microenvironment also selects for increased malignancy. Mitochondria are integral in stress sensing to allow for tumor cells to adapt to stressful conditions. Here, we show that colorectal cancer cells adapted to acidic microenvironment (CRC-AA) are more reliant on oxidative phosphorylation than their parental cells, and the acetyl-CoA in CRC-AA cells are generated from fatty acids and glutamine, but not from glucose. Consistently, CRC-AA cells exhibit increased mitochondrial mass and fitness that depends on an upregulated autophagic flux-lipid droplet axis. Lipid droplets (LDs) function as a buffering system to store the fatty acids derived from autophagy and to protect mitochondria from lipotoxicity in CRC-AA cells. Blockade of LD biogenesis causes mitochondrial dysfunction that can be rescued by inhibiting carnitine palmitoyltransferase 1 α (CPT1α). High level of mitochondrial superoxide is essential for the AMPK activation, resistance to apoptosis, high autophagic flux and mitochondrial function in CRC-AA cells. Thus, our results demonstrate that the cascade of autophagic flux and LD formation plays an essential role in sustaining mitochondrial fitness to promote cancer cell survival under chronic acidosis. Our findings provide insight into the pro-survival metabolic plasticity in cancer cells under microenvironmental or therapeutic stress and imply that this pro-survival cascade may potentially be targeted in cancer therapy.
Collapse
Affiliation(s)
- Xiaojie Liu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
- Biochip Laboratory, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, China
| | - Xue Sun
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Wenqing Mu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Yanan Li
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Wenqing Bu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Tingting Yang
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Jia Zhang
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Rui Liu
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Jiayu Ren
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Peishan Li
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Yufang Shi
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Changshun Shao
- The Third Affiliated of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, China.
| |
Collapse
|
460
|
Kim BR, Rauckhorst AJ, Chimenti MS, Rehman T, Keen HL, Karp PH, Taylor EB, Welsh MJ. The oxygen level in air directs airway epithelial cell differentiation by controlling mitochondrial citrate export. SCIENCE ADVANCES 2025; 11:eadr2282. [PMID: 39854459 PMCID: PMC11759043 DOI: 10.1126/sciadv.adr2282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025]
Abstract
Oxygen controls most metazoan metabolism, yet in mammals, tissue O2 levels vary widely. While extensive research has explored cellular responses to hypoxia, understanding how cells respond to physiologically high O2 levels remains uncertain. To address this problem, we investigated respiratory epithelia as their contact with air exposes them to some of the highest O2 levels in the body. We asked how the O2 level in air controls differentiation of airway basal stem cells into the ciliated epithelial cells essential for clearing airborne pathogens from the lung. Through a metabolomics screen and 13C tracing on primary cultures of human airway basal cells, we found that the O2 level in air directs ciliated cell differentiation by increasing mitochondrial citrate export. Unexpectedly, disrupting mitochondrial citrate export elicited hypoxia transcriptional responses independently of HIF1α stabilization and at O2 levels that would be hyperoxic for most tissues. These findings identify mitochondrial citrate export as a cellular mechanism for responding to physiologically high O2 levels.
Collapse
Affiliation(s)
- Bo Ram Kim
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA, USA
| | - Adam J. Rauckhorst
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Michael S. Chimenti
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Tayyab Rehman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Henry L. Keen
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Philip H. Karp
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA, USA
| | - Eric B. Taylor
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Michael J. Welsh
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
461
|
Guerrieri RA, Fischer GM, Kircher DA, Joon AY, Cortez JR, Grossman AH, Hudgens CW, Ledesma DA, Lazcano R, Onana CY, Knighton BG, Kumar S, Hu Q, Gopal YNV, McQuade JL, Deng W, Haydu LE, Gershenwald JE, Lazar AJ, Tetzlaff MT, Holmen SL, Davies MA. Oxidative Phosphorylation (OXPHOS) Promotes the Formation and Growth of Melanoma Lung and Brain Metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.633049. [PMID: 39896644 PMCID: PMC11785201 DOI: 10.1101/2025.01.23.633049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Melanoma mortality is driven by the formation and growth of distant metastases. Here, we interrogated the role of tumor oxidative phosphorylation (OXPHOS) in the formation of distant metastases in melanoma. OXPHOS was the most upregulated metabolic pathway in primary tumors that formed distant metastases in the RCAS-TVA mouse model of spontaneous lung and brain metastases, and in melanoma patients that developed brain or other distant metastases. Knockout of PGC1α in melanocytes in the RCAS-TVA melanoma mouse model had no impact on primary tumor formation, but markedly reduced the incidence of lung and brain metastases. Genetic knockout of a component of electron transport chain complex I, NDUFS4, in B16-F10 and D4M-UV2 murine melanoma cell lines did not impact tumor incidence following subcutaneous, intravenous, or intracranial injection, but decreased tumor burden specifically in the lungs and brain. Together, these data demonstrate that OXPHOS is critical for the formation of metastases in melanoma. STRUCTURED ABSTRACT Purpose: Melanoma mortality is driven by the formation and growth of distant metastases. However, the process and pathogenesis of melanoma metastasis remain poorly understood. Here, we interrogate the role of tumor oxidative phosphorylation (OXPHOS) in the formation of distant metastases in melanoma.Experimental Design: This study includes (1) new RNA-seq analysis of primary melanomas from patients characterized for distant metastasis events; (2) RNA-seq analysis and functional testing of genetic OXPHOS inhibition (PGC1α KO) the RCAS-TVA model, which is the only existing immunocompetent murine model of autochthonous lung and brain metastasis formation from primary melanoma tumors; and (3) functional experiments of genetic OXPHOS inhibition (NDUFS4 KO) in the B16-F10 and D4M-UV2 murine melanoma cell lines, including evaluation of subcutaneous, lung, and brain metastatic site dependencies.Results: OXPHOS was the most upregulated metabolic pathway in primary tumors that formed distant metastases in the RCAS-TVA mouse model of spontaneous lung and brain metastases, and in melanoma patients that developed brain or other distant metastases. Knockout of PGC1a in melanocytes in the RCAS-TVA melanoma mouse model had no impact on primary tumor formation, but markedly reduced the incidence of lung and brain metastases. Genetic knockout of a component of electron transport chain complex I, NDUFS4, in B16-F10 and D4M-UV2 murine melanoma cell lines did not impact tumor incidence following subcutaneous, intravenous, or intracranial injection, but decreased tumor burden specifically in the lungs and brain.Conclusions: Together, these data demonstrate that OXPHOS is critical for the formation of metastases in melanoma. TRANSLATIONAL RELEVANCE Melanoma is the most aggressive form of skin cancer. One hallmark of this disease is a high risk of distant metastasis formation. The process and pathogenesis of metastasis in this disease remain poorly understood and there is controversy regarding the role of oxidative phosphorylation (OXPHOS) in melanoma metastasis. This study incorporates RNAseq analysis of primary melanoma tumors from patients characterized for distant metastasis events, RNAseq analysis of the only existing immunocompetent murine model of autochthonous lung and brain metastasis formation from primary melanoma tumors, and functional testing in multiple syngeneic models of melanoma at different tissue sites. This integrated analysis consistently demonstrates that melanoma OXPHOS promotes distant metastasis to the lungs and brain, two of the most common and clinically relevant sites of melanoma metastasis. This improved understanding of tumor OXPHOS may represent novel vulnerabilities for therapeutics development and surveillance/preventative strategies for melanoma metastasis.
Collapse
|
462
|
Nolte MJ, Payseur BA. Phenotypic and Developmental Dissection of an Instance of the Island Rule. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634334. [PMID: 40034646 PMCID: PMC11875247 DOI: 10.1101/2025.01.22.634334] [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: 03/05/2025]
Abstract
Organismal body weight correlates with morphology, life history, physiology, and behavior, making it perhaps the most telling single indicator of an organism's evolutionary and ecological profile. Island populations provide an exceptional opportunity to study body weight evolution. In accord with the "island rule," insular small-bodied vertebrates often evolve larger sizes, whereas insular large-bodied vertebrates evolve smaller sizes. To understand how island populations evolve extreme sizes, we adopted a developmental perspective and compared a suite of traits with established connections to body size in the world's largest wild house mice from Gough Island and mice from a smaller-bodied mainland strain. We pinpoint 24-hour periods during the third and fifth week of age in which Gough mice gain exceptionally more weight than mainland mice. We show that Gough mice accumulate more visceral fat beginning early in postnatal development. During a burst of weight gain, Gough mice shift toward carbohydrates and away from fat as fuel, despite being more active than and consuming equivalent amounts of food as mainland mice. Our findings showcase the value of developmental phenotypic characterization for discovering how body weight evolves in the context of broader patterns of trait evolution.
Collapse
Affiliation(s)
- Mark J. Nolte
- Laboratory of Genetics, University of Wisconsin – Madison, Madison, WI 53706
| | - Bret A. Payseur
- Laboratory of Genetics, University of Wisconsin – Madison, Madison, WI 53706
| |
Collapse
|
463
|
Zhang M, Traspov A, Yang J, Zheng M, Kharzinova VR, Ai H, Zinovieva NA, Huang L. Genomic and transcriptomic insights into vitamin A-induced thermogenesis and gene reuse as a cold adaptation strategy in wild boars. Commun Biol 2025; 8:116. [PMID: 39856249 PMCID: PMC11759952 DOI: 10.1038/s42003-025-07536-8] [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: 09/08/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Wild boars inhabit diverse climates, including frigid regions like Siberia, but their migration history and cold adaptation mechanisms into high latitudes remain poorly understood. We constructed the most comprehensive wild boar whole-genome variant dataset to date, comprising 124 samples from tropical to frigid zones, among which 47 Russian, 8 South Chinese and 3 Vietnamese wild boars were newly supplemented. We also gathered 75 high-quality RNA-seq datasets from 10 tissues of 6 wild boars from Russia and 6 from southern China. Demographic analysis revealed the appearance of Russian wild boars in Far East of Asia (RUA) and Europe (RUE) after the last glacial maximum till ~ 10 thousand years ago. Recent gene flow (<100 years) from RUA to RUE reflects human-mediated introductions. Cold-region wild boars exhibit strong selection signatures indicative of genetic adaptation to cold climates. Further pathway and transcriptomic analyses reveal a novel cold resistance mechanism centered on enhanced vitamin A metabolism and catalysis, involving the reuse of UGT2B31 and rhythm regulation by ANGPTL8, RLN3 and ZBTB20. This may compensate for the pig's lack of brown fat/UCP1 thermogenesis. These findings provide new insights into the molecular basis of cold adaptation and improve our understanding of Eurasian wild boar migration history.
Collapse
Affiliation(s)
- Mingpeng Zhang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, P.R. China
| | - Aleksei Traspov
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Jiawen Yang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Min Zheng
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Veronika R Kharzinova
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Huashui Ai
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| | - Natalia A Zinovieva
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia.
| | - Lusheng Huang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| |
Collapse
|
464
|
Fu L, Ding H, Mo L, Pan X, Feng L, Wen S, Lan Q, Long L. The association between body composition and overall survival in patients with advanced non-small cell lung cancer. Sci Rep 2025; 15:3109. [PMID: 39856268 PMCID: PMC11761065 DOI: 10.1038/s41598-025-87073-w] [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: 11/20/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Nutritional status is associated with prognosis in a variety of cancers. Studies analyzing the association between the measurements of skeletal muscle and adipose tissue obtained from Computerized Tomography (CT) images at the time of diagnosis of advanced non-small cell lung cancer (NSCLC) and overall survival (OS) are relatively few. Data from 425 patients diagnosed with advanced NSCLC between January 2016 and December 2017 were retrospectively analyzed, with an average follow-up of 15.3 months. To outline the patient's chest CT plain image at the time of diagnosis,skeletal muscle and subcutaneous fat at the level of both thoracic vertebrae were quantified in terms of mass and quantity by the pectoral muscle index (PMI), pectoral muscle density (PMD), subcutaneous fat index (SFI), subcutaneous fat density (SFD), paravertebral muscle index (PVMI), and paravertebral muscle density (PVMD). The SFI value in the female survival group is significantly lower than that in the death group (P = 0.049), and the PVMI value in the overall survival group is significantly lower than that in the death group (P < 0.001). After adjusting for clinical variables such as gender, smoking status, clinical staging, degree of differentiation, and radiotherapy history, the multivariable Cox regression analysis showed that an increase in SFI significantly improves the overall survival rate of patients (Hazard Ratio [HR] = 1.410, 95% Confidence Interval [CI]: 1.042-1.908, P = 0.026). Conversely, a decrease in PVMD is significantly associated with improved overall survival and prognosis (HR = 0.762, 95% CI: 0.579-0.982, P = 0.048). No association was found between body mass index (BMI) and chest muscle status indicators and overall survival (P > 0.05). CT-measured body composition parameters provide precise prognostic information and are superior to BMI; an increased OS rate in advanced NSCLC is associated with a greater SFI and a lower PVMD.
Collapse
Affiliation(s)
- Liang Fu
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Haiming Ding
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Liupei Mo
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xiaoyu Pan
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Lijuan Feng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shenglian Wen
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Qiaoqing Lan
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Liling Long
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor of Gaungxi Medical University, Ministry of Education, Nanning, China.
| |
Collapse
|
465
|
Cornali K, Di Lauro M, Marrone G, Masci C, Montalto G, Giovannelli A, Schievano C, Tesauro M, Pieri M, Bernardini S, Noce A. The Effects of a Food Supplement, Based on Co-Micronized Palmitoylethanolamide (PEA)-Rutin and Hydroxytyrosol, in Metabolic Syndrome Patients: Preliminary Results. Nutrients 2025; 17:413. [PMID: 39940271 PMCID: PMC11820307 DOI: 10.3390/nu17030413] [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: 12/29/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) patients have impaired hypothalamic regulatory functions involved in food intake and energy expenditure and suffer from a state of meta-inflammation. Pre-clinical studies demonstrated that ultramicronized palmitoylethanolamide (PEA) acts both on the adipose tissue and the central nervous system, while hydroxytyrosol (HTyr) counteracts several types of dysmetabolism. OBJECTIVES The aim of our randomized crossover double-blind placebo-controlled pilot study was to evaluate the potential effects of a food supplement (FS) containing a co-micronized formulation of PEA and rutin along with HTyr, combined with a tailored calorie-controlled Mediterranean diet, in patients with MetS. METHODS Nineteen patients were enrolled and block-randomized to an eight-week MD together with the FS or placebo. After a two-week washout period, the treatments were reversed. Data on laboratory parameters and those detected by capillary sampling, anthropometry, body composition analysis, ultrasound examination, blood pressure monitoring, the 36-Item Short-Form Health Survey questionnaire, handgrip strength test, and physical performance tests were collected at each time point (protocol code R.S. 262.22, registered on 20 December 2022). RESULTS At the end of the study, patients supplemented with the FS showed a significant reduction in body weight, body mass index, fat mass, and inflammation biomarkers (CRP and ESR), compared to placebo-supplemented patients. In contrast, the fat-free mass, phase angle, and body cell mass were increased in FS compared to placebo patients. CONCLUSIONS Although preliminary, the results of our clinical study suggest that co-micronized PEA-rutin and HTyr may be of help against adiposopathy in patients with MetS.
Collapse
Affiliation(s)
- Kevin Cornali
- Department of Experimental Medicine, PhD School in Biochemistry and Molecular Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Manuela Di Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.L.); (G.M.); (C.M.); (M.T.)
| | - Giulia Marrone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.L.); (G.M.); (C.M.); (M.T.)
| | - Claudia Masci
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.L.); (G.M.); (C.M.); (M.T.)
| | - Giulia Montalto
- School of Specialization in Nephrology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Alfredo Giovannelli
- Unit of Laboratory Medicine, University Hospital Tor Vergata, 00133 Rome, Italy; (A.G.); (M.P.)
| | | | - Manfredi Tesauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.L.); (G.M.); (C.M.); (M.T.)
| | - Massimo Pieri
- Unit of Laboratory Medicine, University Hospital Tor Vergata, 00133 Rome, Italy; (A.G.); (M.P.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sergio Bernardini
- Unit of Laboratory Medicine, University Hospital Tor Vergata, 00133 Rome, Italy; (A.G.); (M.P.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Annalisa Noce
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.L.); (G.M.); (C.M.); (M.T.)
- UOSD Nephrology and Dialysis, Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
466
|
Arenas G, Barrera MJ, Contreras-Duarte S. The Impact of Maternal Chronic Inflammatory Conditions on Breast Milk Composition: Possible Influence on Offspring Metabolic Programming. Nutrients 2025; 17:387. [PMID: 39940245 PMCID: PMC11820913 DOI: 10.3390/nu17030387] [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: 12/26/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Breastfeeding is the best way to provide newborns with crucial nutrients and produce a unique bond between mother and child. Breast milk is rich in nutritious and non-nutritive bioactive components, such as immune cells, cytokines, chemokines, immunoglobulins, hormones, fatty acids, and other constituents. Maternal effects during gestation and lactation can alter these components, influencing offspring outcomes. Chronic inflammatory maternal conditions, such as obesity, diabetes, and hypertension, impact breast milk composition. Breast milk from obese mothers exhibits changes in fat content, cytokine levels, and hormonal concentrations, potentially affecting infant growth and health. Similarly, diabetes alters the composition of breast milk, impacting immune factors and metabolic markers. Other pro-inflammatory conditions, such as dyslipidemia and metabolic syndrome, have been barely studied. Thus, maternal obesity, diabetes, and altered tension parameters have been described as modifying the composition of breast milk in its macronutrients and other important biomolecules, likely affecting the offspring's weight. This review emphasizes the impact of chronic inflammatory conditions on breast milk composition and its potential implications for offspring development through the revision of full-access original articles.
Collapse
Affiliation(s)
- Gabriela Arenas
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile;
| | - María José Barrera
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago 7510157, Chile;
| | - Susana Contreras-Duarte
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago 8420524, Chile
| |
Collapse
|
467
|
Zhang Q, Yang Z, Ou X, Zhang M, Qin X, Wu G. The role of immunity in insulin resistance in patients with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2025; 15:1464561. [PMID: 39911236 PMCID: PMC11797073 DOI: 10.3389/fendo.2024.1464561] [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: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent disorder of the endocrine system with significant clinical implications, often leading to health complications related to adipose tissue accumulation, including obesity, insulin resistance (IR), metabolic syndrome, and type 2 diabetes mellitus. While the precise pathogenesis of PCOS remains unclear, it is now recognized that genetic, endocrine, and metabolic dysregulations all contribute significantly to its onset. The immunopathogenesis of PCOS has not been extensively explored, but there is growing speculation that immune system abnormalities may play a pivotal role. This chronic inflammatory state is exacerbated by factors such as obesity and hyperinsulinemia. Therefore, this review aims to elucidate the interplay between IR in PCOS patients, the controlled immune response orchestrated by immune cells and immunomodulatory molecules, and their interactions with adipocytes, hyperandrogenemia, chronic inflammation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Qixuan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhe Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Ou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengying Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyu Qin
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gengxiang Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
468
|
Ma Y, Yan M, Xie Z, Zhang H, Li Z, Li Y, Yang S, Zhang M, Li W, Li J. Generation of live mice from haploid ESCs with germline-DMR deletions or switch. Cell Discov 2025; 11:5. [PMID: 39833184 PMCID: PMC11747502 DOI: 10.1038/s41421-024-00757-x] [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: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 01/22/2025] Open
Abstract
Genomic imprinting is required for sexual reproduction and embryonic development of mammals, in which, differentially methylated regions (DMRs) regulate the parent-specific monoallelic expression of imprinted genes. Numerous studies on imprinted genes have highlighted their critical roles in development. However, what imprinting network is essential for development is still unclear. Here, we establish a stepwise system to reconstruct a development-related imprinting network, in which diploid embryonic stem cells (ESCs) are derived by fusing between parthenogenetic (PG)- and androgenetic (AG)-haploid embryonic stem cells (haESCs) with different DMR deletions (termed Ha-Ha-fusion system), followed by tetraploid complementation to produce all-haESC fetuses. Diploid ESCs fused between PG-haESCs carrying 8 maternally-derived DMR deletions and AG-haESCs with 2 paternally-derived DMR deletions give rise to live pups efficiently, among which, one lives to weaning. Strikingly, diploid ESCs derived from the fusion of PG-haESCs with 7 maternal DMR deletions and AG-haESCs with 2 paternal DMR deletions and maternal Snrpn-DMR deletion also support full-term embryonic development. Moreover, embryos reconstructed by injection of AG-haESCs with hypomethylated H19-DMR into oocytes with H19-DMR deletion develop into live mice sustaining inverted allelic gene expression. Together, our findings indicate that restoration of monoallelic expression of 10 imprinted regions is adequate for the full-term development of all-haESC pups, and it works irrespective of their parental origins. Meanwhile, Ha-Ha-fusion system provides a useful tool for deciphering imprinting regulation networks during embryonic development.
Collapse
Affiliation(s)
- Yongjian Ma
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Yan
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenfei Xie
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongling Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhoujie Li
- Animal Core Facility, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Suming Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meiling Zhang
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Li
- Center for Reproductive Medicine & Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jinsong Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
469
|
Wang N, Zhu S, Chen S, Zou J, Zeng P, Tan S. Neurological mechanism-based analysis of the role and characteristics of physical activity in the improvement of depressive symptoms. Rev Neurosci 2025:revneuro-2024-0147. [PMID: 39829004 DOI: 10.1515/revneuro-2024-0147] [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: 10/11/2024] [Accepted: 12/22/2024] [Indexed: 01/22/2025]
Abstract
Depression is a common mental disorder characterized by a high prevalence and significant adverse effects, making the searching for effective interventions an urgent priority. In recent years, physical activity (PA) has increasingly been recognized as a standard adjunctive treatment for mental disorders owing to its low cost, easy application, and high efficiency. Epidemiological data shows positive preventive and therapeutic effects of PA on mental illnesses such as depression. This article systematically describes the prophylactic and therapeutic effects of PA on depression and its biological basis. A comprehensive literature analysis reveals that PA significantly improves depressive symptoms by upregulating the expression of "exerkines" such as irisin, adiponectin, and BDNF to positively impacting neuropsychiatric conditions. In particular, lactate could also play a critical role in the ameliorating effects of PA on depression due to the findings about protein lactylation as a novel protein post-transcriptional modification. The literature also suggests that in terms of brain structure, PA may improve hippocampal volume, basal ganglia (neostriatum, caudate-crustal nucleus) and PFC density in patients with MDD. In summary, this study elucidates the multifaceted positive effects of PA on depression and its potential biological mechanisms with a particular emphasis on the roles of various exerkines. Future research may further investigate the effects of different types, intensities, and durations of PA on depression, as well as how to better integrate PA interventions into existing treatment strategies to achieve optimal outcomes in mental health interventions.
Collapse
Affiliation(s)
- Nan Wang
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Shanshan Zhu
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Shuyang Chen
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Ju Zou
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Peng Zeng
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
| | - Sijie Tan
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical School, 34706 University of South China , Hengyang 421001, China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China
| |
Collapse
|
470
|
Ferara N, Balta V, Đikić D, Odeh D, Mojsović-Ćuić A, Feher Turković L, Dilber D, Beletić A, Landeka Jurčević I, Šola I. The Effect of the Glucosinolate Sinigrin on Alterations in Molecular Biomarkers of the Myocardium in Swiss Mice. Foods 2025; 14:327. [PMID: 39856994 PMCID: PMC11765020 DOI: 10.3390/foods14020327] [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: 11/21/2024] [Revised: 12/27/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Glucosinolates are chemically stable compounds that exhibit biological activity in the body following hydrolysis catalyzed by the enzyme myrosinase. While existing in vitro and in vivo studies suggest that the hydrolysis products of glucosinolates predominantly exert beneficial effects in both human and animal organisms, some studies have found that the excessive consumption of glucosinolates may lead to toxic and anti-nutritional effects. Given that glucosinolates are primarily ingested in the human diet through dietary supplements and commercially available cruciferous vegetables, we investigated the in vivo effects of the glucosinolate sinigrin on molecular markers in the myocardia of healthy Swiss mice. This study aims to elucidate whether sinigrin induces positive or negative physiological effects in mammals following consumption. The alterations in myocardial parameters were assessed by measuring metabolic, inflammatory, structural, and antioxidant markers. Our findings revealed that subchronic exposure to sinigrin in the myocardia of female mice resulted in a significant increase (p ≤ 0.05) in the levels of the myokine irisin, matrix metalloproteinases (MMP-2, MMP-9), catalase (CAT), and total glutathione (tGSH), alongside a marked decrease (p ≤ 0.05) in the levels of atrial natriuretic peptide (ANP), compared to the control group consisting of both female and male mice. These results suggest that the hydrolysis products of sinigrin may exert a potentially toxic effect on the myocardial tissue of female mice and possess the capability to modulate transcription factors in vivo in a sex-dependent manner. This observation calls for further investigation into the mechanisms regulating the actions of glucosinolate hydrolysis products, their interactions with sex hormones, and the determination of permissible intake levels associated with both beneficial and adverse outcomes.
Collapse
Affiliation(s)
- Nikola Ferara
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Centre, Vinogradska cesta 29, 10000 Zagreb, Croatia;
| | - Vedran Balta
- Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Domagoj Đikić
- Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Dyana Odeh
- Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Ana Mojsović-Ćuić
- School of Applied Health Sciences, University of Zagreb, Mlinarska cesta 38, 10000 Zagreb, Croatia
| | - Lana Feher Turković
- School of Applied Health Sciences, University of Zagreb, Mlinarska cesta 38, 10000 Zagreb, Croatia
| | - Dario Dilber
- Department of Cardiology, Thalassotherapia Opatija, Maršala Tita 188, 51410 Opatija, Croatia;
| | - Anđelo Beletić
- Laboratory of Proteomics, Internal Diseases Clinic, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
| | - Irena Landeka Jurčević
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia
| | - Ivana Šola
- Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| |
Collapse
|
471
|
Zhu Y, Shrestha A. Metabolic syndrome and its effect on immune cells in apical periodontitis- a narrative review. Clin Oral Investig 2025; 29:67. [PMID: 39825203 DOI: 10.1007/s00784-025-06161-3] [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: 11/07/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES Apical periodontitis (AP) is an inflammatory immune response in periapical tissues caused by microbial infections. Failure of root canal treatment or delayed healing is often due to intracanal or extra-radicular bacteria. However, beyond microbial factors, the patient's systemic health can significantly influence the progression and healing of AP. Metabolic syndrome is a risk factor and it is characterized by a cluster of interconnected metabolic risk factors, including abdominal obesity, hyperlipidemia, hypertension, and hyperglycemia. MATERIALS AND METHODS A comprehensive literature review was conducted on apical periodontitis and metabolic syndrome, and their impact on the roles of different immune cell populations. RESULTS Both AP and metabolic syndrome are inflammatory diseases that involve complex and interwoven immune responses. The affected immune cells are categorized into the innate (neutrophils, macrophages, and dendritic cells) and adaptive immune systems (T cells and B cells). CONCLUSIONS Metabolic diseases and AP are closely correlated, possibly intertwined in a two-way relationship driven by a shared dysregulated immune response. CLINICAL RELEVANCE Understanding the pathophysiology and immune mechanisms underlying the two-way relationship between metabolic syndrome and AP can help improve treatment outcomes and enhance the overall well-being of patients with endodontic disease complicated by metabolic syndrome.
Collapse
Affiliation(s)
- Yi Zhu
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada
| | - Annie Shrestha
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Department of Dentistry, Mt. Sinai Hospital, Toronto 412-600 University Avenue, Toronto, ON, M5G 1X5, Canada.
| |
Collapse
|
472
|
Liu J, Yang J, Wu Q, Fang Z, Wang T, Wang Z, Xu D. Review of osteokines in spinal cord injury: potential biomarkers during rehabilitation. J Orthop Surg Res 2025; 20:64. [PMID: 39827357 PMCID: PMC11742232 DOI: 10.1186/s13018-024-05415-2] [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: 09/10/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
After spinal cord injury (SCI), mechanical unloading, denervation, as well as negative changes in blood supply, inflammation state, and hormone levels produce significant negative effects on bone density, leading to a high prevalence of osteoporosis after SCI. It has been recently discovered that skeletal bone also has endocrine functions. Osteokines, secreted from bone tissue, could play multiple roles in regulating bone density, muscle mass, glucose metabolism, and functions of the central nervous system-changes in the osteokine levels after SCI have been detected. Therefore, bone density and osteokine levels should be stressed in clinical settings. Clinical treatment measures for bone loss after SCI include exercise training, physical agent therapy, acupuncture, and so on. According to previous studies, these treatments could affect the expression levels of osteokines. In conclusion, bone loss and changes in osteokines after SCI are worthy of great attention during the rehabilitation of SCI. Osteokines could become biomarkers during SCI rehabilitation, reflecting both bone density and systemic functions. This review summarized recent findings regarding bone loss after SCI, changes in osteokines, and the effect of rehabilitation therapies, with a particular emphasis on the local and systemic regulatory roles of osteokines, as well as their potential as biomarkers during SCI rehabilitation.
Collapse
Affiliation(s)
- Jing Liu
- Department of Acupuncture and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Qinhuai District, Hanzhong Road 155th, Nanjing, 210029, China
| | - Jingyi Yang
- Rehabilitation therapy department, School of Acupuncture-Moxibustion and Tuina of Nanjing, University of Chinese Medicine·School of Health Preservation and Rehabilitation of Nanjing University of Chinese Medicine, Qixia District, Xianlin Road 138th, Nanjing, 210023, China
| | - Qi Wu
- Rehabilitation Medicine School, Nanjing Medical University, Nanjing, 210029, China
| | - Zixuan Fang
- Rehabilitation therapy department, School of Acupuncture-Moxibustion and Tuina of Nanjing, University of Chinese Medicine·School of Health Preservation and Rehabilitation of Nanjing University of Chinese Medicine, Qixia District, Xianlin Road 138th, Nanjing, 210023, China
| | - Tong Wang
- Rehabilitation Medicine School, Nanjing Medical University, Nanjing, 210029, China.
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Gulou District, Guangzhou Road 300th, Nanjing, 210029, China.
| | - Zun Wang
- Rehabilitation therapy department, School of Acupuncture-Moxibustion and Tuina of Nanjing, University of Chinese Medicine·School of Health Preservation and Rehabilitation of Nanjing University of Chinese Medicine, Qixia District, Xianlin Road 138th, Nanjing, 210023, China.
- Rehabilitation Medicine School, Nanjing Medical University, Nanjing, 210029, China.
| | - Daoming Xu
- Department of Acupuncture and Rehabilitation, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Qinhuai District, Hanzhong Road 155th, Nanjing, 210029, China.
| |
Collapse
|
473
|
Kim HJ, Kim YJ, Seong JK. Mouse models for metabolic health research: molecular mechanism of exercise effects on health improvement through adipose tissue remodelling. J Physiol 2025. [PMID: 39823247 DOI: 10.1113/jp285975] [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] [Accepted: 01/02/2025] [Indexed: 01/19/2025] Open
Abstract
Exercise provides health benefits to multiple metabolic tissues through complex biological pathways and interactions between organs. However, investigating these complex mechanisms in humans is still limited, making mouse models extremely useful for exploring exercise-induced changes in whole-body metabolism and health. In this review, we focus on gaining a broader understanding of the metabolic phenotypes and molecular mechanisms induced by exercise in mouse models. We first discuss the differences in adaptations induced by aerobic and resistance exercise, and compare voluntary wheel running and forced treadmill exercise, the two main methods of aerobic exercise research in mice, to show the similarities and differences between the same aerobic exercise but different methods, and their impact on experimental outcomes. The effects of exercise on metabolic phenotypes, including alleviation of obesity and metabolic disorders, and the mechanisms involved in adipose tissue remodelling and browning are explored, as well as the role of the gut microbiota in mediating the physiological responses and metabolic effects of exercise. Understanding these molecular mechanisms and methodological aspects of exercise experiments in mouse models can serve as a valuable template for the design of future basic research in exercise physiology and will provide a strong scientific evidence base for optimizing the design of exercise intervention programmes for metabolic health.
Collapse
Affiliation(s)
- Hye Jin Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Model Animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Youn Ju Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Model Animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Model Animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
- BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
474
|
Zhang Z, Schaefer C, Jiang W, Lu Z, Lee J, Sziraki A, Abdulraouf A, Wick B, Haeussler M, Li Z, Molla G, Satija R, Zhou W, Cao J. A panoramic view of cell population dynamics in mammalian aging. Science 2025; 387:eadn3949. [PMID: 39607904 PMCID: PMC11910726 DOI: 10.1126/science.adn3949] [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: 12/07/2023] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
To elucidate aging-associated cellular population dynamics, we present PanSci, a single-cell transcriptome atlas profiling >20 million cells from 623 mouse tissues across different life stages, sexes, and genotypes. This comprehensive dataset reveals >3000 different cellular states and >200 aging-associated cell populations. Our panoramic analysis uncovered organ-, lineage-, and sex-specific shifts in cellular dynamics during life-span progression. Moreover, we identify both systematic and organ-specific alterations in immune cell populations associated with aging. We further explored the regulatory roles of the immune system on aging and pinpointed specific age-related cell population expansions that are lymphocyte dependent. Our "cell-omics" strategy enhances comprehension of cellular aging and lays the groundwork for exploring the complex cellular regulatory networks in aging and aging-associated diseases.
Collapse
Affiliation(s)
- Zehao Zhang
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY, USA
| | - Chloe Schaefer
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
| | - Weirong Jiang
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
| | - Ziyu Lu
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY, USA
| | - Jasper Lee
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
| | - Andras Sziraki
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
- The David Rockefeller Graduate Program in Bioscience, The Rockefeller University, New York, NY, USA
| | - Abdulraouf Abdulraouf
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
- The Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Brittney Wick
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, CA, USA
| | | | - Zhuoyan Li
- New York Genome Center, New York, NY, USA
| | | | - Rahul Satija
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Wei Zhou
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
| | - Junyue Cao
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
475
|
Colonnello E, Libotte F, Masi D, Curreli M, Massetti C, Gandini O, Gangitano E, Watanabe M, Mariani S, Gnessi L, Lubrano C. Eating behavior patterns, metabolic parameters and circulating oxytocin levels in patients with obesity: an exploratory study. Eat Weight Disord 2025; 30:6. [PMID: 39820758 PMCID: PMC11742293 DOI: 10.1007/s40519-024-01698-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/20/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Obesity is a complex heterogeneous disease often associated with dysfunctional eating behavior patterns. Oxytocin (OT) is a neurohormone involved in the regulation of energy metabolism and eating behavior. The aim of the present study was to evaluate in a population of patients with obesity circulating levels of OT and dysfunctional eating behaviors in relation to anthropometric, hormonal and metabolic parameters. METHODS A prospective, observational, single-center study was conducted at the Center of High Specialization for the Care of Obesity of Sapienza University of Rome. Adult subjects with body mass index (BMI) ≥ 30 kg/m2 were recruited. Body impedance assessment (BIA), biochemical and hormonal parameters, plasma OT concentration analysis and the Eating Behaviors Assessment for Obesity (EBA-O) questionnaire were evaluated. RESULTS A total of 21 patients, 16 females and 5 males, with a mean age of 45.7 ± 15.1 years, mean BMI of 40.89 ± 8.02 kg/m2 and plasma OT concentration of 1365.61 ± 438.03 pg/mL were recruited. The dysfunctional eating behavior traits investigated by the EBA-O appear significantly associated with metabolic derangements. In particular, night eating is associated with alterations in lipid metabolism (p < 0.01). Circulating OT correlates positively with BMI (r = 0,43; p < 0.05), and Hepatic Steatosis Index (HIS) (r = 0.46; p < 0.05), while its role in subjects with obesity and alterations in glucose metabolism is less clear. Interestingly, circulating OT levels < 1312.55 pg/mL may be predictive of food addiction (100% sensitivity; 62.5% specificity). CONCLUSIONS Despite the need for larger studies to confirm their validity, the clinical utility of the EBA-O and circulating OT in identifying dysfunctional eating behaviors appears promising.
Collapse
Affiliation(s)
- Elena Colonnello
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy.
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Flavia Libotte
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Davide Masi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Mariaignazia Curreli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Chandra Massetti
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Orietta Gandini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena Gangitano
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Mikiko Watanabe
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Stefania Mariani
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome "La Sapienza", Policlinico Umberto Viale del Policlinico 155 - 00161, Rome, Italy.
| |
Collapse
|
476
|
Radoszkiewicz K, Rybkowska P, Szymanska M, Krzesniak NE, Sarnowska A. The influence of biomimetic conditions on neurogenic and neuroprotective properties of dedifferentiated fat cells. Stem Cells 2025; 43:sxae066. [PMID: 39576128 PMCID: PMC11811640 DOI: 10.1093/stmcls/sxae066] [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: 05/06/2024] [Accepted: 09/30/2024] [Indexed: 02/12/2025]
Abstract
In the era of a constantly growing number of reports on the therapeutic properties of dedifferentiated, ontogenetically rejuvenated cells and their use in the treatment of neurological diseases, the optimization of their derivation and long-term culture methods seem to be crucial. One of the solutions is seen in the use of dedifferentiated fat cells (DFATs) that are characterized by a greater homogeneity. Moreover, these cells seem to possess a higher expression of transcriptional factors necessary to maintain pluripotency (stemness-related transcriptional factors) as well as a greater ability to differentiate in vitro into 3 embryonic germ layers, and a high proliferative potential in comparison to adipose stem/stromal cells. However, the neurogenic and neuroprotective potential of DFATs is still insufficiently understood; hence, our research goal was to contribute to our current knowledge of the subject. To recreate the brain's physiological (biomimetic) conditions, the cells were cultured at 5% oxygen concentration. The neural differentiation capacity of DFATs was assessed in the presence of the N21 supplement containing the factors that are typically found in the natural environment of the neural cell niche or in the presence of cerebrospinal fluid and under various spatial conditions (microprinting). The neuroprotective properties of DFATs were assessed using the coculture method with the ischemically damaged nerve tissue.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Magdalena Szymanska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| | - Natalia Ewa Krzesniak
- Department of Plastic and Reconstructive Surgery, Centre of Postgraduate Medical Education, Prof. W. Orlowski Memorial Hospital, 00‐416 Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02‐106 Warsaw, Poland
| |
Collapse
|
477
|
Tang N, Li W, Shang H, Yang Z, Chen Z, Shi G. Irisin-mediated KEAP1 degradation alleviates oxidative stress and ameliorates pancreatitis. Immunol Res 2025; 73:37. [PMID: 39821708 DOI: 10.1007/s12026-024-09588-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Oxidative stress (OS) injury is pivotal in acute pancreatitis (AP) pathogenesis, contributing to inflammatory cascades. Irisin, a ubiquitous cytokine, exhibits antioxidant properties. However, the role of irisin in AP remains inconclusive. Our study aims to elucidate irisin expression in AP patients and investigate its mechanism of action to propose a novel treatment strategy for AP. Serum irisin levels in 65 AP patients were quantified using an enzyme-linked immunosorbent assay and correlated with disease severity scores. Core genes implicated in AP-related oxidative stress were identified and screened via bioinformatics analysis. The therapeutic efficacy of irisin in AP was confirmed using a murine cerulein-induced AP model. The intrinsic mechanism of irisin's antioxidative stress action was investigated and verified in pancreatic AR42J cells (Supplementary Fig. 1). Common targets shared by irisin and AP were further validated using a molecular docking model which was constructed for virtual docking analysis. This study investigated alterations in redox status in AP and found a significant reduction in serum irisin levels, correlating inversely with AP severity. In a murine AP model, we showed that irisin triggers an antioxidative stress program via the KEAP1 gene; this process helps reestablish redox balance by decreasing the buildup of reactive oxygen species (ROS) and suppressing the secretion of inflammatory mediators within pancreatic tissues Notably, increased KEAP1 expression counteracted the antioxidative effects of irisin. Our findings unveil a novel therapeutic mechanism for AP, wherein irisin inhibits KEAP1 to alleviate OS. Increasing irisin levels in vivo presents a promising strategy for AP treatment.
Collapse
Affiliation(s)
- Nan Tang
- Dalian Medical University, Dalian, Liaoning, China
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Wendi Li
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hezhen Shang
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zengyin Chen
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China.
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
478
|
Papp RS, Könczöl K, Sípos K, Tóth ZE. Nesfatin-1 Neurons in the Ventral Premammillary Nucleus Integrate Metabolic and Reproductive Signals in Male Rats. Int J Mol Sci 2025; 26:739. [PMID: 39859453 PMCID: PMC11765514 DOI: 10.3390/ijms26020739] [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: 11/18/2024] [Revised: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The ability to reproduce depends on metabolic status. In rodents, the ventral premammillary nucleus (PMv) integrates metabolic and reproductive signals. While leptin (adiposity-related) signaling in the PMv is critical for female fertility, male reproductive functions are strongly influenced by glucose homeostasis. The anorexigenic peptide nesfatin-1 is a leptin-independent central regulator of blood glucose. Therefore, its integrative role in male rats can be assumed. To investigate this, we mapped the distribution of nesfatin-1 mRNA- and protein-producing cells in the PMv during postnatal development via in situ hybridization and immunohistochemistry, respectively. Fos-nesfatin-1, double immunostaining was used to determine the combined effect of heterosexual pheromone challenge and insulin-induced hypoglycemia on neuronal activation in adults. We found that ~75% of the pheromone-activated neurons were nesfatin-1 cells. Hypoglycemia reduced pheromone-induced cell activation, particularly in nesfatin-1 neurons. Immuno-electron microscopy revealed innervation of PMv nesfatin-1 neurons by urocortin3-immunoreactive terminals, reportedly originating from the medial amygdala. Nesfatin-1 immunopositive neurons expressed GPR10 mRNA, a receptor associated with metabolic signaling, but did not respond with accumulation of phosphorylated STAT3 immunopositivity, a marker of leptin receptor signaling, in response to intracerebroventricular leptin treatment. Our results suggest that PMv nesfatin-1 neurons are primarily responsible for integrating reproductive and metabolic signaling in male rats.
Collapse
Affiliation(s)
- Rege Sugárka Papp
- Human Brain Tissue Bank and Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary;
| | - Katalin Könczöl
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary; (K.K.); (K.S.)
| | - Klaudia Sípos
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary; (K.K.); (K.S.)
| | - Zsuzsanna E. Tóth
- Laboratory of Neuroendocrinology and In Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary; (K.K.); (K.S.)
| |
Collapse
|
479
|
Russo L, Babboni S, Andreassi MG, Daher J, Canale P, Del Turco S, Basta G. Treating Metabolic Dysregulation and Senescence by Caloric Restriction: Killing Two Birds with One Stone? Antioxidants (Basel) 2025; 14:99. [PMID: 39857433 PMCID: PMC11763027 DOI: 10.3390/antiox14010099] [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: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest accompanied by metabolic activity and characteristic phenotypic changes. This process is crucial for developing age-related diseases, where excessive calorie intake accelerates metabolic dysfunction and aging. Overnutrition disturbs key metabolic pathways, including insulin/insulin-like growth factor signaling (IIS), the mammalian target of rapamycin (mTOR), and AMP-activated protein kinase. The dysregulation of these pathways contributes to insulin resistance, impaired autophagy, exacerbated oxidative stress, and mitochondrial dysfunction, further enhancing cellular senescence and systemic metabolic derangements. On the other hand, dysfunctional endothelial cells and adipocytes contribute to systemic inflammation, reduced nitric oxide production, and altered lipid metabolism. Numerous factors, including extracellular vesicles, mediate pathological communication between the vascular system and adipose tissue, amplifying metabolic imbalances. Meanwhile, caloric restriction (CR) emerges as a potent intervention to counteract overnutrition effects, improve mitochondrial function, reduce oxidative stress, and restore metabolic balance. CR modulates pathways such as IIS, mTOR, and sirtuins, enhancing glucose and lipid metabolism, reducing inflammation, and promoting autophagy. CR can extend the health span and mitigate age-related diseases by delaying cellular senescence and improving healthy endothelial-adipocyte interactions. This review highlights the crosstalk between endothelial cells and adipocytes, emphasizing CR potential in counteracting overnutrition-induced senescence and restoring vascular homeostasis.
Collapse
Affiliation(s)
- Lara Russo
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Babboni
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura 100, Lebanon;
| | - Paola Canale
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Giuseppina Basta
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| |
Collapse
|
480
|
Liu H, Wang G, Sui C, Guo Y, He X. Woxuanzhongzhou formula improves DHEAS and high-fat diet-induced IR and anovulatory mice via AMPK/PGC1- α/Irisin pathway. J Ovarian Res 2025; 18:7. [PMID: 39819643 PMCID: PMC11737048 DOI: 10.1186/s13048-025-01587-5] [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: 08/11/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in women of reproductive age. Anovulation is one of the most important clinical features of PCOS, and insulin resistance (IR) is one of the critical pathogenic factors. Woxuanzhongzhou (WXZZ) is a traditional herbal formulation that has shown efficacy in treating PCOS combined with IR, but the underlying mechanism is not clear. The aim of this study was to investigate the molecular mechanism of WXZZ on dehydroepiandrosterone sulfate and high fat diet induced PCOS with IR mice. METHODS 40 female C57BL/6 mice were randomized to 4 groups: control group, model group, metformin group, and WXZZ group. Some mice is induced by dehydroepiandrosterone sulfate (DHEA) and high-fat diet (HFD) for 3 weeks. Following model induction, metformin and WXZZ were administered by gavage. Body weight, fasting blood glucose (FBG), fasting insulin (FINS) levels, the homeostatic model assessment of insulin resistance (HOMA-IR), and gonadal hormones were measured. Estrous cycles were monitored. The structure of the gastrocnemius muscle and subcutaneous fatty tissue were also evaluated. Additionally, serum irisin and non-esterified fatty acids (NEAF) levels and the protein and gene expression levels of AMPK, PGC1-α, FNDC5, irisin in the gastrocnemius muscle and CaMKK, AMPK, PGC1-α, UCP1 in fat were analyzed. RESULTS The DHEA + HFD + WXZZ group exhibited significant improvements in several key parameters compared to the DHEA + HFD group. WXZZ ameliorated endocrine and metabolic disorders, resumed estrous cycle in DHEAS and high-fat diet-induced IR and anovulatory mice. Significant reductions were observed in body weight, serum testosterone, luteinizing hormone, luteinizing hormone/ follicle-stimulating hormone ratio, FINS, and HOMA-IR. Additionally, WXZZ promoted irisin expression and secretion by up-regulating the protein and gene AMPK/PGC1-α/FNDC5 expression in gastrocnemius muscle and up-regulated the protein and gene CaMKK/AMPK/PGC1-α/UCP1 expression in fat. WXZZ inhibited the overproduction of serum NEFA, and reduced lipid accumulation. Structural analysis of the gastrocnemius muscle and adipose tissue revealed partial restoration. CONCLUSION WXZZ exhibits therapeutic effects in DHEAS and high-fat diet-induced IR and anovulatory mice. These effects may be mediated through the activation of AMPK/PGC1-α pathway in muscle to promote the secretion of irisin.
Collapse
Affiliation(s)
- Haijuan Liu
- Department of Gynecology, Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Traditional Chinese Medicine, Northwest Women's and Children's Hospital, Xi'an, 710061, China
| | - Guohua Wang
- Department of Gynecology, Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Conglu Sui
- Department of Gynecology, Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yanan Guo
- Department of Gynecology, Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiangyu He
- Shunyi Hospital, Beijing Traditional Chinese Medicine Hospital, Beijing, 101300, China
| |
Collapse
|
481
|
Ma X, Zhang D, Yang Z, Sun M, Gao N, Mei C, Zan L. bta-miR-484 Inhibits Bovine Intramuscular Adipogenesis by Regulating Mitotic Clonal Expansion via the MAP3K9/JNK/CCND1 Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1062-1074. [PMID: 39719059 DOI: 10.1021/acs.jafc.4c07956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Intramuscular fat (IMF) content is a critical indicator of the beef nutritional value and flavor. In this study, we focused on bta-miR-484, a microRNA that is differentially expressed during the adipogenic differentiation of bovine intramuscular adipocytes and is negatively correlated with the IMF content across different cattle breeds. Our findings demonstrate that bta-miR-484 inhibits adipogenic differentiation without altering the fatty acid composition of bovine intramuscular adipocytes. miRNA pull-down and dual-luciferase reporter assays confirmed that MAP3K9 is a target gene of bta-miR-484. Furthermore, bta-miR-484 suppresses the JNK signaling pathway by targeting MAP3K9, leading to decreased CCND1 expression, which impedes the mitotic clonal expansion (MCE) process and inhibits intramuscular adipocyte differentiation. In summary, this study uncovers a novel mechanism by which bta-miR-484 regulates bovine IMF content and provides the first exploration of MCE during intramuscular adipocyte adipogenic differentiation. These findings offer valuable theoretical insights into beef cattle breeding and molecular improvements.
Collapse
Affiliation(s)
- Xinhao Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Dianqi Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhimei Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Meijun Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Ni Gao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Chugang Mei
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| |
Collapse
|
482
|
Stecco C, Pratt R, Nemetz LD, Schleip R, Stecco A, Theise ND. Towards a comprehensive definition of the human fascial system. J Anat 2025. [PMID: 39814456 DOI: 10.1111/joa.14212] [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: 09/03/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
The absence of a clear consensus on the definition and significance of fascia and the indiscriminate use of the term throughout the clinical and scientific literature has led to skepticism regarding its importance in the human body. To address this challenge, we propose that: (1) fasciae, and the fascial interstitia within them, constitute an anatomical system, defined as a layered body-wide multiscale network of connective tissue that allows tensional loading and shearing mobility along its interfaces; (2) the fascial system comprises four anatomical organs: the superficial fascia, musculoskeletal (deep) fascia, visceral fascia, and neural fascia; (3) these organs are further composed of anatomical structures, some of which are eponymous; (4) all these fascial organs and their structural components contain variable combinations and arrangements of the four classically defined tissues: epithelial, connective, muscle, and neural; (5) the overarching functions of the fascial system arise from the contrasting biomechanical properties of the two basic types of layers distributed throughout the system: one predominantly collagenous and relatively stiff, the other rich in hyaluronic acid and viscous, allowing for the free flow of fluid; (6) the topographical organization of these layers in different locations is related to local variations in function (e.g. unidirectional arrangements favor tensional loading, interwoven structures favor shear mobility) thereby accounting for both the system's universal functional aspects and the site-specific variations between them. A universal language related to fascia will break down linguistic barriers and facilitate cross-disciplinary cooperation, enabling scientists and practitioners from diverse backgrounds to contribute their expertise seamlessly.
Collapse
Affiliation(s)
- Carla Stecco
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Rebecca Pratt
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
| | - Laurice D Nemetz
- College of Health Professions, Pace University, Pleasantville, New York, USA
| | - Robert Schleip
- Department of Sport and Health Sciences, Technical University of Munich, Munich, Germany
- Department for Medical Professions, Diploma University of Applied Sciences, Bad Sooden-Allendorf, Germany
- Fascia Research Group, Experimental Anesthesiology, Ulm University, Ulm, Germany
| | - Antonio Stecco
- Department of Rehabilitation Medicine, Rusk Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Neil D Theise
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
483
|
van Eenige R, Hoekx CA, Sardjoe Mishre ASD, Straat ME, Boon MR, Martinez‐Tellez B, Rensen PCN, Kan HE. Cold exposure and thermoneutrality similarly reduce supraclavicular brown adipose tissue fat fraction in fasted young lean adults. FASEB J 2025; 39:e70307. [PMID: 39797666 PMCID: PMC11724390 DOI: 10.1096/fj.202402415r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025]
Abstract
Brown adipose tissue (BAT) is a metabolically highly active tissue that dissipates energy stored within its intracellular triglyceride droplets as heat. Others have previously utilized MRI to show that the fat fraction of human supraclavicular BAT (scBAT) decreases upon cold exposure, compared with baseline (i.e., pre-cooling). However, comparisons to a control group that was not exposed to cold are largely lacking. We recently developed a non-invasive dynamic MRI protocol that allows for quantifying scBAT fat fraction changes over time. Here, we aimed to study the effect of cold exposure versus thermoneutrality on fat fraction changes in human scBAT. Ten young (mean age: 21.5 ± 0.7 years), lean (mean BMI: 21.7 ± 0.5 kg/m2), 12 h-fasted volunteers (9 females; 1 male) underwent up to 70 consecutive MRI scans each on two separate study visits in a cross-over design. Participants were exposed to a temperature of 32°C for 10 scans (i.e., ±16 min), which was then either lowered to 18°C (i.e., cold exposure) or was maintained at 32°C (i.e., thermoneutrality). Dynamic fat fraction changes were quantified, and self-reported thermal perception scores were monitored. The fat fraction in scBAT decreased over time upon cold exposure (r = -.222, p < .001). Interestingly however, we also observed a decrease in scBAT fat fraction over time upon thermoneutrality (r = -.212, p < .001). No difference was observed between the two temperature conditions (p = .55), while self-reported thermal perception scores were consistently higher (i.e., colder) upon cold exposure. In the trapezius muscle and the humerus bone as control tissues, the fat fraction was unaffected in both temperature conditions. The fat fraction in subcutaneous white adipose tissue (sWAT) however, also decreased over time upon cold exposure (r = -.270, p < .001) and during thermoneutrality (r = -.190, p < .001), again with no difference (p = .92) between the two temperature conditions. In conclusion, our results show that in 12 h-fasted, healthy individuals cold exposure and thermoneutrality similarly reduce the fat fraction within scBAT and sWAT. While we interpret that the cold exposure was sufficient to induce thermogenesis, we suggest that an increased lipolytic activity within adipocytes, as a consequence of fasting, may be the primary cause of the decreased fat fraction in both sWAT and scBAT in our study. The current study highlights the potential influence of fasting on the fat fraction in scBAT and stresses the importance of inclusion of a thermoneutral control group in studies investigating the BAT-modulating effect of cold exposure.
Collapse
Affiliation(s)
- Robin van Eenige
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
| | - Carlijn A. Hoekx
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | | | - Maaike E. Straat
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Borja Martinez‐Tellez
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Nursing Physiotherapy and Medicine, SPORT Research Group (CTS‐1024), CIBIS Research CenterUniversity of AlmeríaAlmeríaSpain
- Biomedical Research UnitTorrecárdenas University HospitalAlmeríaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
| | - Patrick C. N. Rensen
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Hermien E. Kan
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
484
|
Chen W, Liu Y, Liu J, Chen Y, Wang X. Acute exercise promotes WAT browning by remodeling mRNA m 6A methylation. Life Sci 2025; 361:123269. [PMID: 39581460 DOI: 10.1016/j.lfs.2024.123269] [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: 09/27/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
AIMS Regular exercise promotes the beiging and metabolic adaptations of white adipose tissue (WAT) through the cumulative transcriptional responses that occur after each exercise session. However, the effects of a single bout of acute exercise and the role of N6-methyladenosine (m6A) in these adaptations remain unclear. We aim to investigate this further. MATERIALS AND METHODS We constructed mouse models for chronic (8 weeks of running) and acute (single 1-hour run) exercise to study the effects on white adipose tissue (WAT) metabolism and beiging through metabolic phenotyping and transcriptome sequencing. Additionally, we explored the impact of acute exercise on WAT m6A modification and target genes, combining m6A regulators with cell models to elucidate the role of m6A in WAT exercise adaptation. KEY FINDINGS Here, we reveal that upregulated m6A modification after acute exercise induces the formation of glycolytic beige fat (g-beige fat) in WAT. Mechanistically, the metabolite β-hydroxybutyrate (BHBA) secreted after acute exercise upregulates m6A modification in WAT. This enhances m6A-dependent translation of the histone acetyltransferase CREBBP, promoting the transcription of key beiging genes by increasing chromatin accessibility. Pharmacologically elevating circulating BHBA mimics the metabolic response induced by acute exercise, upregulating m6A modification and its downstream signals. Additionally, BHBA exhibits long-term effects, improving metabolic homeostasis in obesity by promoting thermogenesis in WAT. SIGNIFICANCE Our results reveal the role of metabolites in WAT metabolic adaptation through m6A-mediated chromatin accessibility after acute exercise, providing a novel therapeutic target for regulating WAT metabolism from a nutritional epigenetics perspective.
Collapse
Affiliation(s)
- Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
485
|
Serano M, Perni S, Pierantozzi E, Laurino A, Sorrentino V, Rossi D. Intracellular Membrane Contact Sites in Skeletal Muscle Cells. MEMBRANES 2025; 15:29. [PMID: 39852269 PMCID: PMC11767089 DOI: 10.3390/membranes15010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Intracellular organelles are common to eukaryotic cells and provide physical support for the assembly of specialized compartments. In skeletal muscle fibers, the largest intracellular organelle is the sarcoplasmic reticulum, a specialized form of the endoplasmic reticulum primarily devoted to Ca2+ storage and release for muscle contraction. Occupying about 10% of the total cell volume, the sarcoplasmic reticulum forms multiple membrane contact sites, some of which are unique to skeletal muscle. These contact sites primarily involve the plasma membrane; among these, specialized membrane contact sites between the transverse tubules and the terminal cisternae of the sarcoplasmic reticulum form triads. Triads are skeletal muscle-specific contact sites where Ca2+ channels and regulatory proteins assemble to form the so-called calcium release complex. Additionally, the sarcoplasmic reticulum contacts mitochondria to enable a more precise regulation of Ca2+ homeostasis and energy metabolism. The sarcoplasmic reticulum and the plasma membrane also undergo dynamic remodeling to allow Ca2+ entry from the extracellular space and replenish the stores. This process involves the formation of dynamic membrane contact sites called Ca2+ Entry Units. This review explores the key processes in biogenesis and assembly of intracellular membrane contact sites as well as the membrane remodeling that occurs in response to muscle fatigue.
Collapse
Affiliation(s)
- Matteo Serano
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Stefano Perni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Annunziatina Laurino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
486
|
Wu Y, Deng S, Wei S, Wei W, He Y, Guo J. Adipocyte-Targeted Nanotechnology and Cell-Based Therapy for Obesity Treatment. ChemMedChem 2025; 20:e202400611. [PMID: 39390653 DOI: 10.1002/cmdc.202400611] [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: 08/06/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Obesity is a critical risk factor for the development of metabolic diseases and is often associated with dysfunctional adipocytes. Prevalent treatments such as lifestyle intervention, pharmacotherapy, and bariatric surgery are often accompanied by adverse side effects and poor patient compliance. Nanotechnology and cell-based therapy offer innovative approaches for targeted obesity treatments, as they can directly target adipocytes, regulate lipid metabolism, and minimize off-target effects. Here, we provide an overview of the intricate relationship between adipocytes and obesity, highlighting the potential of nanotechnology and cell-based therapy in obesity treatment. Additionally, we discuss the advancements of adipose-derived mesenchymal stem cells (ADMSCs) in obesity progression, including the latest challenges and considerations for developing adipose-targeted treatments for obesity. The objective is to provide a perspective on the design and development of nanotechnology and cell-based therapy for treating obesity and related comorbidities.
Collapse
Affiliation(s)
- Yue Wu
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Siqi Deng
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Siyu Wei
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wenqi Wei
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Bioproducts Institute, Department of Chemical and Biological Engineering, The, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- State Key Laboratory of Polymer Materials Engineering, Department of Chemical and Biological Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
487
|
Kentistou KA, Lim BEM, Kaisinger LR, Steinthorsdottir V, Sharp LN, Patel KA, Tragante V, Hawkes G, Gardner EJ, Olafsdottir T, Wood AR, Zhao Y, Thorleifsson G, Day FR, Ozanne SE, Hattersley AT, O'Rahilly S, Stefansson K, Ong KK, Beaumont RN, Perry JRB, Freathy RM. Rare variant associations with birth weight identify genes involved in adipose tissue regulation, placental function and insulin-like growth factor signalling. Nat Commun 2025; 16:648. [PMID: 39809772 PMCID: PMC11733218 DOI: 10.1038/s41467-024-55761-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Investigating the genetic factors influencing human birth weight may lead to biological insights into fetal growth and long-term health. We report analyses of rare variants that impact birth weight when carried by either fetus or mother, using whole exome sequencing data in up to 234,675 participants. Rare protein-truncating and deleterious missense variants are collapsed to perform gene burden tests. We identify 9 genes; 5 with fetal-only effects on birth weight, 1 with maternal-only effects, 3 with both, and observe directionally concordant associations in an independent sample. Four of the genes were previously implicated by GWAS of birth weight. IGF1R and PAPPA2 (fetal and maternal-acting) have known roles in insulin-like growth factor bioavailability and signalling. PPARG, INHBE and ACVR1C (fetal-acting) are involved in adipose tissue regulation, and the latter two also show associations with favourable adiposity patterns in adults. We highlight the dual role of PPARG (fetal-acting) in adipocyte differentiation and placental angiogenesis. NOS3 (fetal and maternal-acting), NRK (fetal), and ADAMTS8 (maternal-acting) have been implicated in placental function and hypertension. To conclude, our analysis of rare coding variants identifies regulators of fetal adipose tissue and fetoplacental angiogenesis as determinants of birth weight, and further evidence for the role of insulin-like growth factors.
Collapse
Affiliation(s)
- Katherine A Kentistou
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Brandon E M Lim
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Andrew R Wood
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Yajie Zhao
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Susan E Ozanne
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., 102 Reykjavik, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ken K Ong
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robin N Beaumont
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - John R B Perry
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Rachel M Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
488
|
Serratì S, Zerlotin R, Manganelli M, Di Fonte R, Dicarlo M, Oranger A, Colaianni G, Porcelli L, Azzariti A, Guida S, Grano M, Colucci SC, Guida G. Irisin and Metastatic Melanoma: Selective Anti-Invasiveness Activity in BRAF Wild-Type Cells. Int J Mol Sci 2025; 26:652. [PMID: 39859367 PMCID: PMC11765811 DOI: 10.3390/ijms26020652] [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: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Irisin is a newly discovered 12 kDa messenger protein involved in energy metabolism. Irisin affects signaling pathways in several types of cancer; however, the role of irisin in metastatic melanoma (MM) has not been described yet. We explored the biological effects of irisin in in vitro models of MM cells (HBLwt/wt, LND1wt/wt, Hmel1V600K/wt and M3V600E/V600E) capable of the oncogenic activation of BRAF. We treated MM cells with different concentrations of r-irisin (10 nM, 25 nM, 50 nM, 100 nM) for 24 h-48 h. An MTT assay highlighted that r-irisin did not affect the proliferation of MM cells. We subsequently treated MM cells with 10 nM r-irisin, corresponding to the dose exhibiting biological activity in vitro. Irisin reduced the invasive ability of only LND1wt/wt (p < 0.05), which highly expressed αv gene levels, but did not affect the invasion of BRAFmut cells. Gelatin zymography analysis showed a reduction in the enzymatic activity of MMP-2 and MMP-9 in BRAFwt/wt cells treated with 10 nM r-irisin. Moreover, gene expression analysis (qPCR) of MMP-2 and MMP-9 and of the fibrinolytic system (uPAR, uPA and PAI-1) highlighted a crucial role of 10 nM r-irisin treatment in the inhibition of pro-invasive systems in BRAFwt/wt. In conclusion, our results may suggest a possible differential role of irisin in melanoma cells.
Collapse
Affiliation(s)
- Simona Serratì
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Michele Manganelli
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| | - Roberta Di Fonte
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Manuela Dicarlo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Letizia Porcelli
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Amalia Azzariti
- IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy; (S.S.); (R.D.F.); (L.P.); (A.A.)
| | - Stefania Guida
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- Dermatology Clinic, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.D.); (A.O.); (G.C.); (M.G.)
| | - Silvia Concetta Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| | - Gabriella Guida
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy; (M.M.); (S.C.C.)
| |
Collapse
|
489
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [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] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
490
|
Cicuéndez B, Mora A, López JA, Curtabbi A, Pérez-García J, Porteiro B, Jimenez-Blasco D, Latorre-Muro P, Vo P, Jerome M, Gómez-Santos B, Romero-Becerra R, Leiva M, Rodríguez E, León M, Leiva-Vega L, Gómez-Lado N, Torres JL, Hernández-Cosido L, Aguiar P, Marcos M, Jastroch M, Daiber A, Aspichueta P, Bolaños JP, Spinelli JB, Puigserver P, Enriquez JA, Vázquez J, Folgueira C, Sabio G. Absence of MCJ/DnaJC15 promotes brown adipose tissue thermogenesis. Nat Commun 2025; 16:229. [PMID: 39805849 PMCID: PMC11730624 DOI: 10.1038/s41467-024-54353-4] [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: 03/10/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity poses a global health challenge, demanding a deeper understanding of adipose tissue (AT) and its mitochondria. This study describes the role of the mitochondrial protein Methylation-controlled J protein (MCJ/DnaJC15) in orchestrating brown adipose tissue (BAT) thermogenesis. Here we show how MCJ expression decreases during obesity, as evident in human and mouse adipose tissue samples. MCJKO mice, even without UCP1, a fundamental thermogenic protein, exhibit elevated BAT thermogenesis. Electron microscopy unveils changes in mitochondrial morphology resembling BAT activation. Proteomic analysis confirms these findings and suggests involvement of the eIF2α mediated stress response. The pivotal role of eIF2α is scrutinized by in vivo CRISPR deletion of eIF2α in MCJKO mice, abrogating thermogenesis. These findings uncover the importance of MCJ as a regulator of BAT thermogenesis, presenting it as a promising target for obesity therapy.
Collapse
Grants
- K99 DK133502 NIDDK NIH HHS
- R01 DK136640 NIDDK NIH HHS
- This work has been supported by the following projects: PMP21/00057 funded by the Instituto de Salud Carlos III (ISCIII) - European Union (FEDER/FSE) "Una manera de hacer Europa"/ "El FSE invierte en tu futuro"/ Next Generation EU and cofunded by the European Union / Plan de Recuperación, Transformación y Resiliencia (PRTR); PID2022-138525OB-I00 de la Agencia Estatal de Investigación 10.13039/501100011033, financiado por MICIU/AEI/10.13039/501100011033 fondos FEDER and EU, PDC2021-121147-I00 and PID2019-104399RB-I00 funded by MCIN/AEI/10.13039/501100011033 and the European Union “NextGenerationEU”/Plan de Recuperación Transformación y Resiliencia -PRTR; Grant RED2022-134397-T funded by MCIN/AEI/10.13039/501100011033 and, as appropriate, by “ERDF A way of making Europe”, by the “European Union” or by the “European Union NextGenerationEU/PRTR”; Fundación Jesús Serra; EFSD/Lilly Dr Sabio; 2017 Leonardo Grant BBVA Foundation (Investigadores-BBVA-2017); Comunidad de Madrid IMMUNOTHERCAN-CM S2010/BMD-2326 and B2017/BMD-373; Fundación AECC PROYE19047SABI, PGC2018-097019-B-I00 and PT17/0019/0003- ISCIII-SGEFI /ERDF, ProteoRed. PreMed-Exp: PMP21/00057, PMP21/00113 Infraestructura de Medicina de Precisión asociada a la Ciencia y Tecnología IMPACT-2021 Instituto de Salud Carlos III (GS, JLT).. G.S is a Miembro Numerario of the RACVE. The CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation) and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033).
- A.C was supported by the European Union's Horizon 2020 research and 328 innovation program under the Marie Skłodowska-Curie grant agreement n. 713,673.
- J.P-G was supported by the fellowship from” la Caixa” Foundation (ID 100010434), the fellowship code is LCF/BQ/DR24/12080018.
- M.M is supported by Instituto de Salud Carlos III (ISCIII) and the European Union project PI20/00743.
- P.A is supported by MCIU/AEI/FEDER, UE (PID2021-124425OB-I00) and Basque Government, Department of Education (IT1476-22).
- J.P.B is funded by AEI grants PID2019-105699RB-I00, PID2022-138813OB-I00 and PDC2021-121013-I00; HORIZON-MSCA-2021-DN-01grant 101072759; and La Caixa Research Health grant HR23-00793.
- C.F was funded with Sara Borrell (CD19/ 00078), NNF23SA0083952-EASO/Novo Nordisk New Investigator Award in Basic Sciences 2023, EFSD/Lilly Young Investigator Award 2022, Society for Endocrinology/Early Career Grant 2022, FSEEN/ Jóvenes endocrinólogos 2022, EFSD/Novo Nordisk Rising Star 2024, IBSA Foundation Fellowship Endocrinology 2023.
Collapse
Affiliation(s)
- Beatriz Cicuéndez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Juan Antonio López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Curtabbi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Pérez-García
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Begoña Porteiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Jimenez-Blasco
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU. Leioa, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Magdalena Leiva
- Department of Immunology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Marta León
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Noemi Gómez-Lado
- Molecular Imaging Biomarkers and Theragnosis Lab, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Nuclear Medicine Service, University Clinical Hospital of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | | | - Lourdes Hernández-Cosido
- Bariatric Surgery Unit. Department of General Surgery, University Hospital of Salamanca. Department of Surgery. University of Salamanca, Salamanca, Spain
| | - Pablo Aguiar
- Molecular Imaging Biomarkers and Theragnosis Lab, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Nuclear Medicine Service, University Clinical Hospital of Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Salamanca, Spain
- Department of Medicine. University of Salamanca, Salamanca, Spain
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Andreas Daiber
- Department of Cardiology 1, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU. Leioa, Biobizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red sobre enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Pedro Bolaños
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
- UMass Chan Medical School Cancer Center, Worcester, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - José Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Organ Crosstalk in Metabolic Diseases Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain.
| |
Collapse
|
491
|
So J, Strobel O, Wann J, Kim K, Paul A, Acri DJ, Dabin LC, Kim J, Peng G, Roh HC. Robust single-nucleus RNA sequencing reveals depot-specific cell population dynamics in adipose tissue remodeling during obesity. eLife 2025; 13:RP97981. [PMID: 39804687 PMCID: PMC11729396 DOI: 10.7554/elife.97981] [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: 01/16/2025] Open
Abstract
Single-nucleus RNA sequencing (snRNA-seq), an alternative to single-cell RNA sequencing (scRNA-seq), encounters technical challenges in obtaining high-quality nuclei and RNA, persistently hindering its applications. Here, we present a robust technique for isolating nuclei across various tissue types, remarkably enhancing snRNA-seq data quality. Employing this approach, we comprehensively characterize the depot-dependent cellular dynamics of various cell types underlying mouse adipose tissue remodeling during obesity. By integrating bulk nuclear RNA-seq from adipocyte nuclei of different sizes, we identify distinct adipocyte subpopulations categorized by size and functionality. These subpopulations follow two divergent trajectories, adaptive and pathological, with their prevalence varying by depot. Specifically, we identify a key molecular feature of dysfunctional hypertrophic adipocytes, a global shutdown in gene expression, along with elevated stress and inflammatory responses. Furthermore, our differential gene expression analysis reveals distinct contributions of adipocyte subpopulations to the overall pathophysiology of adipose tissue. Our study establishes a robust snRNA-seq method, providing novel insights into the biological processes involved in adipose tissue remodeling during obesity, with broader applicability across diverse biological systems.
Collapse
Affiliation(s)
- Jisun So
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| | - Olivia Strobel
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| | - Jamie Wann
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| | - Kyungchan Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| | - Avishek Paul
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| | - Dominic J Acri
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Luke C Dabin
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisUnited States
| | - Gang Peng
- Department of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolisUnited States
| | - Hyun Cheol Roh
- Department of Biochemistry and Molecular Biology, Indiana University School of MedicineIndianapolisUnited States
| |
Collapse
|
492
|
Zhang L, Zhou Y, Yang Z, Jiang L, Yan X, Zhu W, Shen Y, Wang B, Li J, Song J. Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J Neuroinflammation 2025; 22:7. [PMID: 39806503 PMCID: PMC11730833 DOI: 10.1186/s12974-025-03334-5] [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: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Lipid droplets (LDs), serving as the convergence point of energy metabolism and multiple signaling pathways, have garnered increasing attention in recent years. Different cell types within the central nervous system (CNS) can regulate energy metabolism to generate or degrade LDs in response to diverse pathological stimuli. This article provides a comprehensive review on the composition of LDs in CNS, their generation and degradation processes, their interaction mechanisms with mitochondria, the distribution among different cell types, and the roles played by these cells-particularly microglia and astrocytes-in various prevalent neurological disorders. Additionally, we also emphasize the paradoxical role of LDs in post-cerebral ischemia inflammation and explore potential underlying mechanisms, aiming to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yunfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhongbo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liangchao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyang Yan
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wenkai Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bolong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiaxi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
493
|
Ribichini E, Pallotta N, Badiali D, Carlucci M, Ceccanti M, Cambieri C, Libonati L, Corazziari ES, Ruoppolo G, Inghilleri M. Assessment of upper GI motor activity and GI symptoms in patients with amyotrophic lateral sclerosis: an observational study. Front Neurol 2025; 15:1509917. [PMID: 39871987 PMCID: PMC11769799 DOI: 10.3389/fneur.2024.1509917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Background/aims Oro-pharyngeal dysfunction has been reported in Amyotrophic Lateral Sclerosis (ALS). We aimed to assess ALS patients upper gastrointestinal (GI) motor activity and GI symptoms according to bulbar and spinal onset and severity of ALS. Methods ALS bulbar (B) and spinal (S) patients with ALS Functional Rating Scale (ALSFRS-r) ≥35, bulbar sub-score ≥10, and Forced Vital Capacity (FVC) >50%, underwent to: Fiberoptic Endoscopic Evaluation of Swallowing (FEES); esophageal manometry; gastric emptying; Rome symptom questionnaire. Medical Research Council Scale for Muscle Strength (MRC) was performed for the upper and lower limbs. Mann-Whitney's U, Fisher's ranks test, Pearson's test was used. Results Thirteen ALS patients were included (6 F; mean age 61.2 ± 13.7 years, range: 37-87), 5 with B and 8 with S onset (ALSFRS-R score 39.5 ± 4.9, MRC score 128.6 ± 23.3, disease duration 22.8 ± 17.9 months). FEES detected a high dysphagia score in 5 patients with no difference between S and B phenotype. Lower esophageal sphincter pressure was normal in all patients. Esophageal dysmotility was observed in three S and two B onset patients. Upper esophageal sphincter (UES) pressure was higher in all ALS patients. UES spasms and delayed gastric emptying were detected in two B and one S and in two B and four S patients, respectively. There was no correlation between esophagogastric motor abnormalities and clinical characteristics of ALS, nor GI symptoms. Conclusions The presence of UES spasm and the delayed gastric emptying in a subgroup of ALS patients may suggest the role of ANS dysfunction in ALS.
Collapse
Affiliation(s)
- Emanuela Ribichini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Nadia Pallotta
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Danilo Badiali
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Maria Carlucci
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Marco Ceccanti
- Neuromuscular Disorders Unit, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - Chiara Cambieri
- Neuromuscular Disorders Unit, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - Laura Libonati
- Neuromuscular Disorders Unit, Department of Human Neurosciences, Sapienza University, Rome, Italy
| | | | | | - Maurizio Inghilleri
- Department of Human Neurosciences, Sapienza University, Rome, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| |
Collapse
|
494
|
Yao X, Mai X, Tian Y, Liu Y, Jin G, Li Z, Chen S, Dai X, Huang L, Fan Z, Pan G, Pan X, Li X, Yu MC, Sun J, Ou J, Chen H, Xie L. Skeletal muscle-specific Bambi deletion induces hypertrophy and oxidative switching coupling with adipocyte thermogenesis against metabolic disorders. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-023-2586-x. [PMID: 39821828 DOI: 10.1007/s11427-023-2586-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/01/2024] [Indexed: 01/19/2025]
Abstract
Skeletal muscle plays a significant role in both local and systemic energy metabolism. The current investigation aims to explore the role of the Bambi gene in skeletal muscle, focusing on its implications for muscle hypertrophy and systemic metabolism. We hypothesize that skeletal muscle-specific deletion of Bambi induces muscle hypertrophy, improves metabolic performance, and activates thermogenic adipocytes via the reprogramming of progenitor of iWAT, offering potential therapeutic strategies for metabolic syndromes. Leveraging the Chromatin immunoprecipitation (ChIP)-seq and bioinformatics analysis, Bambi gene is shown to be a direct target of HIF2α, which is further confirmed by ChIP-qPCR and promoter luciferase assay. Skeletal muscle-specific Bambi deletion led to significant muscle hypertrophy and improved metabolic parameters, even under high-fat diet conditions. This deletion induced metabolic reprogramming of stromal vascular fractions (SVFs) into thermogenic adipocytes, contributing to systemic metabolic improvements, potentially through the secretory factor. Notably, mice with skeletal muscle-specific Bambi deletion demonstrate resistance to high-fat diet-induced metabolic disorders, highlighting a potential therapeutic pathway for metabolic syndrome management. Thus, skeletal muscle-specific deletion of Bambi triggers muscle growth, enhances metabolic performance, and activates thermogenic adipocytes. These findings suggest Bambi as a novel therapeutic target for metabolic syndromes, providing new insights into the interaction between muscle hypertrophy and systemic metabolic improvement. The study underscores the potential of manipulating muscle physiology to regulate whole-body metabolism, offering a novel perspective on treating metabolic disorders.
Collapse
Affiliation(s)
- Xiangping Yao
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ye Tian
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China
| | - Guanghui Jin
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ze Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shujie Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Liujing Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Zijing Fan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Guihua Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xiaohan Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Xiangmin Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Michael C Yu
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, 14260, USA
| | - Jia Sun
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jingxing Ou
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, 528300, China.
- College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, 510300, China.
| |
Collapse
|
495
|
Xu W, Dong L, Dai J, Zhong L, Ouyang X, Li J, Feng G, Wang H, Liu X, Zhou L, Xia Q. The interconnective role of the UPS and autophagy in the quality control of cancer mitochondria. Cell Mol Life Sci 2025; 82:42. [PMID: 39800773 PMCID: PMC11725563 DOI: 10.1007/s00018-024-05556-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Uncontrollable cancer cell growth is characterized by the maintenance of cellular homeostasis through the continuous accumulation of misfolded proteins and damaged organelles. This review delineates the roles of two complementary and synergistic degradation systems, the ubiquitin-proteasome system (UPS) and the autophagy-lysosome system, in the degradation of misfolded proteins and damaged organelles for intracellular recycling. We emphasize the interconnected decision-making processes of degradation systems in maintaining cellular homeostasis, such as the biophysical state of substrates, receptor oligomerization potentials (e.g., p62), and compartmentalization capacities (e.g., membrane structures). Mitochondria, the cellular hubs for respiration and metabolism, are implicated in tumorigenesis. In the subsequent sections, we thoroughly examine the mechanisms of mitochondrial quality control (MQC) in preserving mitochondrial homeostasis in human cells. Notably, we explored the relationships between mitochondrial dynamics (fusion and fission) and various MQC processes-including the UPS, mitochondrial proteases, and mitophagy-in the context of mitochondrial repair and degradation pathways. Finally, we assessed the potential of targeting MQC (including UPS, mitochondrial molecular chaperones, mitochondrial proteases, mitochondrial dynamics, mitophagy and mitochondrial biogenesis) as cancer therapeutic strategies. Understanding the mechanisms underlying mitochondrial homeostasis may offer novel insights for future cancer therapies.
Collapse
Affiliation(s)
- Wanting Xu
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Ji Dai
- Institute of International Technology and Economy, Development Research Center of the State Council, Beijing, 102208, China
| | - Lu Zhong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Xiao Ouyang
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Jiaqian Li
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Gaoqing Feng
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Huahua Wang
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Xuan Liu
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liying Zhou
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
496
|
Cano-Barquilla P, Jiménez-Ortega V, Fernández-Mateos P, Virto L, Maldonado Bautista E, Perez-Miguelsanz J, Esquifino AI. Daily Lipolysis Gene Expression in Male Rat Mesenteric Adipose Tissue: Obesity and Melatonin Effects. Int J Mol Sci 2025; 26:577. [PMID: 39859293 PMCID: PMC11765279 DOI: 10.3390/ijms26020577] [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: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Melatonin is involved in various functions such as the timing of circadian rhythms, energy metabolism, and body mass gain in experimental animals. However, its effects on adipose tissue lipid metabolism are still unclear. This study analyzes the effects of melatonin on the relative gene expression of lipolytic proteins in rat mesenteric adipose tissue and free fatty acid (FFA) and glycerol plasma levels of male Wistar rats fed a high-fat (HFD) or maintenance diet. Four experimental groups were established: control, obese, and control or obese plus 2.3 mg/kg/day of melatonin in tap water. After 11 weeks, animals were sacrificed at different times throughout a 24 h cycle, and mesenteric adipose tissue and plasma samples were collected and analyzed. Cgi58, Perilipin, and Dgat1 gene expression, as well as FFA and glycerol concentrations, showed rhythm patterns in the control group. HFD disrupted those rhythm patterns and increased FFA and glycerol concentrations during the dark photoperiod. In both melatonin-treated groups, almost all analyzed genes showed circadian patterns. Notably, melatonin significantly prevented the increase in FFA levels during the dark photoperiod in obese rats (obese group: ~1100 mM vs. obese + melatonin group: ~600 μM, similar to control levels). However, the rhythmic pattern observed in control animals was not sustained. According to our results, melatonin could regulate circadian gene transcription of mesenteric adipose tissue lipolysis proteins. The effect of melatonin on preventing elevated FFA plasma levels associated with high-fat diet intake warrants further investigation.
Collapse
Affiliation(s)
- Pilar Cano-Barquilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Vanesa Jiménez-Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Pilar Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Leire Virto
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Estela Maldonado Bautista
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Juliana Perez-Miguelsanz
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Ana I. Esquifino
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| |
Collapse
|
497
|
Lee Y, Tukei KL, Fang Y, Kuila S, Liu X, Imoukhuede PI. Integrative analysis of angiogenic signaling in obesity: capillary features and VEGF binding kinetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630107. [PMID: 39763822 PMCID: PMC11703262 DOI: 10.1101/2024.12.23.630107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Obesity is a global health crisis, with its prevalence particularly severe in the United States, where over 42% of adults are classified as obese. Obesity is driven by complex molecular and tissue-level mechanisms that remain poorly understood. Among these, angiogenesis-primarily mediated by vascular endothelial growth factor (VEGF-A)-is critical for adipose tissue expansion but presents unique challenges for therapeutic targeting due to its intricate regulation. Systems biology approaches have advanced our understanding of VEGF-A signaling in vascular diseases, but their application to obesity is limited by scattered and sometimes contradictory data. To address this gap, we performed a comprehensive analysis of the existing literature to synthesize key findings, standardize data, and provide a holistic perspective on the adipose vascular microenvironment. The data mining revealed five key findings: (1) obesity increases adipocyte size by 78%; (2) vessel density in adipose tissue decreases by 51% in obese mice, with vessels being 47-58% smaller and 4-9 times denser in comparison with tumor vessels; (3) capillary basement membrane thickness remains similar regardless of obesity; (4) VEGF-A shows the strongest binding affinity for VEGFR1, with four times stronger affinity for VEGFR2 than for NRP1; and (5) binding affinities measured by radioligand binding assay and surface plasmon resonance (SPR) are significantly different. These consolidated findings provide essential parameters for systems biology modeling, new insights into obesity-induced changes in adipose tissue, and a foundation for developing angiogenesis-targeting therapies for obesity.
Collapse
|
498
|
Cariati I, Bonanni R, Romagnoli C, Caprioli L, D’Arcangelo G, Tancredi V, Annino G. Bone Adaptations to a Whole Body Vibration Protocol in Murine Models of Different Ages: A Preliminary Study on Structural Changes and Biomarker Evaluation. J Funct Morphol Kinesiol 2025; 10:26. [PMID: 39846667 PMCID: PMC11755639 DOI: 10.3390/jfmk10010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
Background/Objectives: Whole body vibration (WBV) is a valuable tool to mitigate physiological adaptations related to age and inactivity. Although significant benefits have been found at the musculoskeletal level, including increased bone mass and reduced muscle atrophy, the underlying biological mechanisms remain largely unknown. Therefore, our study aimed to evaluate the effects of vibratory training on bone tissue in murine models of different age groups by investigating the structural and distribution changes in some crucial biomarkers involved in musculoskeletal homeostasis. Methods: Specifically, 4-, 12-, and 24-month-old mice were trained with a WBV protocol characterized by three series of 2 min and 30 s, interspersed with a recovery period of the same duration, on a 3-weekly frequency for 3 months. At the end of the training, histological and morphometric analyses were conducted, in association with immunohistochemical analysis to investigate changes in the distribution of fibronectin type III domain-containing protein 5 (FNDC5), NADPH oxidase 4 (NOX4), and sirtuin 1 (SIRT1). Results: Our preliminary results showed that WBV improves musculoskeletal health by preserving bone architecture and promoting up-regulation of FNDC5 and SIRT1 and down-regulation of NOX4. Conclusions: Our study confirms vibratory training as a viable alternative to counter musculoskeletal decline in elderly and/or sedentary subjects. Further investigations should be conducted to deepen knowledge in this field and explore the role of other molecular mediators in physiological adaptations to vibration.
Collapse
Affiliation(s)
- Ida Cariati
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (I.C.); (G.D.); (V.T.); (G.A.)
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, “Tor Vergata” University of Rome, 00133 Rome, Italy
| | - Cristian Romagnoli
- Department of Human Science and Promotion of Quality of Life, San Raffaele Open University, 00166 Rome, Italy;
| | - Lucio Caprioli
- Sports Engineering Laboratory, Department of Industrial Engineering, “Tor Vergata” University of Rome, 00133 Rome, Italy;
| | - Giovanna D’Arcangelo
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (I.C.); (G.D.); (V.T.); (G.A.)
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (I.C.); (G.D.); (V.T.); (G.A.)
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| | - Giuseppe Annino
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (I.C.); (G.D.); (V.T.); (G.A.)
- Sports Engineering Laboratory, Department of Industrial Engineering, “Tor Vergata” University of Rome, 00133 Rome, Italy;
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| |
Collapse
|
499
|
Gianazza E, Papaianni GG, Brocca L, Banfi C, Mallia A. Omics Approaches to Study Perilipins and Their Significant Biological Role in Cardiometabolic Disorders. Int J Mol Sci 2025; 26:557. [PMID: 39859272 PMCID: PMC11765208 DOI: 10.3390/ijms26020557] [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: 12/02/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Lipid droplets (LDs), highly dynamic cellular organelles specialized in lipid storage and maintenance of lipid homeostasis, contain several proteins on their surface, among which the perilipin (Plin) family stands out as the most abundant group of LD-binding proteins. They play a pivotal role in influencing the behavior and functionality of LDs, regulating lipase activity, and preserving a balance between lipid synthesis and degradation, which is crucial in the development of obesity and abnormal accumulation of fat in non-adipose tissues, causing negative adverse biological effects, such as insulin resistance, mitochondrial dysfunction, and inflammation. The expression levels of Plins are often associated with various diseases, such as hepatic steatosis and atherosclerotic plaque formation. Thus, it becomes of interest to investigate the Plin roles by using appropriate "omics" approaches that may provide additional insight into the mechanisms through which these proteins contribute to cellular and tissue homeostasis. This review is intended to give an overview of the most significant omics studies focused on the characterization of Plin proteins and the identification of their potential targets involved in the development and progression of cardiovascular and cardiometabolic complications, as well as their interactors that could be useful for more efficient therapeutic and preventive approaches for patients.
Collapse
Affiliation(s)
| | | | | | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (E.G.); (G.G.P.); (L.B.); (A.M.)
| | | |
Collapse
|
500
|
Homan EA, Gilani A, Rubio-Navarro A, Johnson MA, Schaepkens OM, Cortada E, Pereira de Lima R, Stoll L, Lo JC. Complement 3a receptor 1 on macrophages and Kupffer cells is not required for the pathogenesis of metabolic dysfunction-associated steatotic liver disease. eLife 2025; 13:RP100708. [PMID: 39773465 PMCID: PMC11709426 DOI: 10.7554/elife.100708] [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] [Indexed: 01/30/2025] Open
Abstract
Together with obesity and type 2 diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global epidemic. Activation of the complement system and infiltration of macrophages has been linked to progression of metabolic liver disease. The role of complement receptors in macrophage activation and recruitment in MASLD remains poorly understood. In human and mouse, C3AR1 in the liver is expressed primarily in Kupffer cells, but is downregulated in humans with MASLD compared to obese controls. To test the role of complement 3a receptor (C3aR1) on macrophages and liver resident macrophages in MASLD, we generated mice deficient in C3aR1 on all macrophages (C3aR1-MφKO) or specifically in liver Kupffer cells (C3aR1-KpKO) and subjected them to a model of metabolic steatotic liver disease. We show that macrophages account for the vast majority of C3ar1 expression in the liver. Overall, C3aR1-MφKO and C3aR1-KpKO mice have similar body weight gain without significant alterations in glucose homeostasis, hepatic steatosis and fibrosis, compared to controls on a MASLD-inducing diet. This study demonstrates that C3aR1 deletion in macrophages or Kupffer cells, the predominant liver cell type expressing C3ar1, has no significant effect on liver steatosis, inflammation or fibrosis in a dietary MASLD model.
Collapse
Affiliation(s)
- Edwin A Homan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Maya A Johnson
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Odin M Schaepkens
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Eric Cortada
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Renan Pereira de Lima
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| | - James C Lo
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, Weill Center for Metabolic Health, Weill Cornell MedicineNew YorkUnited States
| |
Collapse
|