1
|
Jiang H, Ye J. The Warburg effect: The hacked mitochondrial-nuclear communication in cancer. Semin Cancer Biol 2025; 112:93-111. [PMID: 40147702 DOI: 10.1016/j.semcancer.2025.03.006] [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: 07/31/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Mitochondrial-nuclear communication is vital for maintaining cellular homeostasis. This communication begins with mitochondria sensing environmental cues and transmitting signals to the nucleus through the retrograde cascade, involving metabolic signals such as substrates for epigenetic modifications, ATP and AMP levels, calcium flux, etc. These signals inform the nucleus about the cell's metabolic state, remodel epigenome and regulate gene expression, and modulate mitochondrial function and dynamics through the anterograde feedback cascade to control cell fate and physiology. Disruption of this communication can lead to cellular dysfunction and disease progression, particularly in cancer. The Warburg effect is the metabolic hallmark of cancer, characterized by disruption of mitochondrial respiration and increased lactate generation from glycolysis. This metabolic reprogramming rewires retrograde signaling, leading to epigenetic changes and dedifferentiation, further reprogramming mitochondrial function and promoting carcinogenesis. Understanding these processes and their link to tumorigenesis is crucial for uncovering tumorigenesis mechanisms. Therapeutic strategies targeting these disrupted pathways, including metabolic and epigenetic components, provide promising avenues for cancer treatment.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Luo Z, Ji R, Ye R, Shi Y, Pang Q, Yin M. Reduced serum levels of mitochondria-derived peptide MOTS-c in patients with obstructive sleep apnea. Sleep Biol Rhythms 2025; 23:305-311. [PMID: 40538389 PMCID: PMC12174021 DOI: 10.1007/s41105-025-00578-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/30/2025] [Indexed: 06/22/2025]
Abstract
Recent research has identified the mitochondrial open reading frame of the 12S rRNA-c (MOTS-c) as a crucial mitochondrial peptide that significantly influences metabolic regulation, mimics the effects of exercise, and mitigates oxidative stress. This study aims to investigate the relationship between serum MOTS-c levels and obstructive sleep apnea (OSA) to enhance our understanding of the disease's pathophysiology. By elucidating this relationship, we hope to uncover new insights into the mechanisms underlying OSA and its associated metabolic complications. Seventy-seven participants were enrolled in this study, including 53 patients with OSA and 24 controls. We measured serum MOTS-c levels and collected participants' demographic characteristics, polysomnography (PSG) data, complete blood count (CBC) data, and sleep-related questionnaires. The study included 77 participants, consisting of 8 patients with mild OSA, 16 with moderate OSA, 29 with severe OSA, and 24 controls. The cohort comprised 26 women and 51 men. Analysis revealed that serum MOTS-c levels were significantly correlated with BMI, AHI (Apnea-Hypopnea Index), and ODI (Oxygen Desaturation Index), independent of age. Additionally, the severity of OSA was inversely related to serum MOTS-c levels, with lower levels observed in patients with more severe OSA. Variations in serum MOTS-c levels were also noted across different BMI classifications. Analysis of covariance (ANCOVA), with BMI as a covariate, demonstrated that the severity of OSA was an independent factor influencing serum MOTS-c levels. Serum MOTS-c levels correlate with both severity of OSA and BMI classification, suggesting that MOTS-c may have significant therapeutic potential for treating OSA.
Collapse
Affiliation(s)
- Zhuoding Luo
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029 China
| | - Rui Ji
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029 China
| | - Renjing Ye
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029 China
| | - Yawen Shi
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029 China
| | - Qingfeng Pang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Min Yin
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029 China
| |
Collapse
|
3
|
Wadding-Lee CA, Makarewich CA. Microproteins in Metabolism. Cells 2025; 14:859. [PMID: 40558487 PMCID: PMC12190854 DOI: 10.3390/cells14120859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/30/2025] [Accepted: 06/05/2025] [Indexed: 06/29/2025] Open
Abstract
Metabolism is a complex network of biochemical pathways that break down macromolecules to produce energy essential for cellular function. Disruptions in metabolic homeostasis are closely linked to noncommunicable diseases (NCDs) such as cardiovascular disease, type 2 diabetes, and cancer, which are leading causes of death worldwide. Many NCD-associated conditions, including obesity and insulin resistance, stem from metabolic dysfunction, and current therapies often fall short in preventing disease progression, highlighting the need for novel therapeutic targets. Microproteins, small proteins of ≤100-150 amino acids, have recently emerged as important regulators of metabolism. Encoded by short open reading frames (sORFs), many of these proteins were historically overlooked due to their small size and misclassification as noncoding RNAs. Advances in genomics and proteomics have revealed that these sORFs can encode functional proteins with critical roles in metabolic pathways. In this review, we highlight the microproteins involved in energy metabolism, mitochondrial function, and nutrient signaling. We discuss their emerging roles in the pathogenesis of NCDs and explore their potential as novel therapeutic targets. As microprotein biology continues to evolve, these small but powerful regulators may offer new strategies for treating metabolic dysfunction and reducing the global burden of NCDs.
Collapse
Affiliation(s)
- Caris A. Wadding-Lee
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Catherine A. Makarewich
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
4
|
Jaunbocus N, Ebenki V, Su H, Slavoff SA. Eukaryotic Microproteins. Annu Rev Biochem 2025; 94:1-28. [PMID: 40245354 DOI: 10.1146/annurev-biochem-080124-012840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Microproteins are polypeptides of 100-150 amino acids or fewer that have not been annotated by genome annotation consortia, given their small size and other noncanonical properties. Translated microproteins are now known to number in the thousands in the human genome, to function in critical cellular and physiological processes, and to be dysregulated or mutated in diseases including neurodegeneration and cancer. Knowledge about microproteins has rapidly accumulated since the advent of ribosome profiling enabled their global discovery 15 years ago. In this review, we summarize what is known about eukaryotic microprotein discovery, the sequences and expression mechanisms of small open reading frames, and microprotein functions from yeast to human.
Collapse
Affiliation(s)
- Nadiya Jaunbocus
- Department of Chemistry, Yale University, New Haven, Connecticut, USA;
- Institute for Biomolecular Design and Discovery, Yale University, West Haven, Connecticut, USA
| | - Valerie Ebenki
- Department of Chemistry, Yale University, New Haven, Connecticut, USA;
- Institute for Biomolecular Design and Discovery, Yale University, West Haven, Connecticut, USA
| | - Haomiao Su
- Department of Chemistry, Yale University, New Haven, Connecticut, USA;
- Institute for Biomolecular Design and Discovery, Yale University, West Haven, Connecticut, USA
| | - Sarah A Slavoff
- Department of Chemistry, Yale University, New Haven, Connecticut, USA;
- Institute for Biomolecular Design and Discovery, Yale University, West Haven, Connecticut, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Yerra VG, Connelly KA. MOTS-c: Magical Molecule for Diabetic Cardiomyopathy? Cardiovasc Drugs Ther 2025; 39:473-476. [PMID: 40172798 DOI: 10.1007/s10557-025-07689-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 bond St, Toronto, ON, M5B1W8, Canada
| | - Kim A Connelly
- Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 bond St, Toronto, ON, M5B1W8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Wu N, Shen C, Wang J, Chen X, Zhong P. MOTS-c Peptide Attenuated Diabetic Cardiomyopathy in STZ-Induced Type 1 Diabetic Mouse Model. Cardiovasc Drugs Ther 2025; 39:491-498. [PMID: 38141139 DOI: 10.1007/s10557-023-07540-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) pathogenesis is a common complication of diabetes, but effective treatments remain limited. Mitochondrial-derived peptide MOTS-c has shown therapeutic promise in animal models of various heart diseases, but its efficacy in DCM is unknown. This study investigates the effects of MOTS-c treatment in a mouse model of type 1 diabetes-induced DCM. METHODS Type 1 diabetes (T1DM) was induced in mice by streptozotocin (STZ) injection. After diabetes establishment, the mice were randomly dividend into two groups treated with or without MOTS-c peptide, which was administered subcutaneously by osmotic pump for 12 weeks. At the end of the experiment, cardiac function, histology, and molecular changes were determined. RESULTS The results showed that diabetic mice exhibited significant cardiac dysfunction, dilatation, and adverse cardiac remodeling. MOTS-c treatment markedly ameliorated these diabetes-associated myocardial function and structure abnormalities. Additionally, MOTS-c reversed AMPK signaling deactivation and inhibited inflammation in the diabetic heart. CONCLUSIONS Our data demonstrated a protective effect of MOTS-c against diabetic cardiomyopathy potentially by activating the AMPK pathway and inhibiting inflammation. These findings demonstrate the therapeutic efficacy of MOTS-c for diabetic cardiomyopathy and warrant further investigation into its clinical potential.
Collapse
Affiliation(s)
- Nan Wu
- Department of Cardiovascular Medicine, The First Affliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Caijie Shen
- Department of Cardiovascular Medicine, The First Affliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Jian Wang
- Department of Cardiovascular Medicine, The First Affliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Xiaomin Chen
- Department of Cardiovascular Medicine, The First Affliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| | - Peng Zhong
- Department of Cardiology Research Institute, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
7
|
Chen F, Li Z, Wang T, Fu Y, Lyu L, Xing C, Li S, Li. MOTS-c mimics exercise to combat diabetic liver fibrosis by targeting Keap1-Nrf2-Smad2/3. Sci Rep 2025; 15:18460. [PMID: 40425777 PMCID: PMC12116857 DOI: 10.1038/s41598-025-03526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 05/21/2025] [Indexed: 05/29/2025] Open
Abstract
Liver fibrosis is a common complication of T2DM(Type 2 diabetes mellitus). Appropriate intervention (exercise or drugs) in the early stage of liver fibrosis can slow down or even reverse liver fibrosis. MOTS-c (Mitochondrial open reading frame of the 12 S r RNA type-c ) has been described as an exercise-mimicking substance, and its effects are similar to those achieved by aerobic exercise; however, the exact mechanism remains to be elucidated. In this study, liver function was impaired in a T2DM rat model, leading to the aggravation of liver fibrosis. T2DM rats with liver fibrosis were subjected to MOTS-c, aerobic exercise therapy, or their combination. HE staining, Masson's trichrome staining and immunohistochemistry were used for histopathological examination. Transcriptome sequencing, q-PCR and WB were used to detect the expression of Keap1 (Kelch-like ECH-associated protein 1), Nrf2 (Nuclear factor erythroid 2-related factor 2 ), Smad2/3/4 and other genes. MOTS-c and aerobic exercise therapy improved T2DM-induced liver fibrosis. Additionally, cells were transfected with MOTS-c overexpression or interference plasmids or MOTS-c was added to the culture medium. MOTS-c overexpression or MOTS-c addition to the culture medium inhibited ROS levels, increased the mRNA and protein expression of Keap1-Nrf2 pathway genes and decreased the expression of TGF-β1(Transforming growth factor-beta1)/Smad pathway genes. Our findings demonstrate that MOTS-c modulates the progression of T2DM complicated by liver fibrosis through a Keap1-Nrf2-Smad2/3 signaling pathway-dependent mechanism.
Collapse
Affiliation(s)
- Feilong Chen
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Zhiyu Li
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Tutu Wang
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Yu Fu
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Lei Lyu
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Chengyuan Xing
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Shunchang Li
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China.
| | - Li
- Sports Medicine Key Laboratory of Sichuan Province, Expert Centre of Sichuan Province, Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| |
Collapse
|
8
|
Girişgen İ, Altıncık SA, Avcı E, Öcal M, Becerir T, Öztekin GM, Özhan B, Yüksel S. Could MOTS-c Levels in Children with Type 1 Diabetes Mellitus Be an Indicator for Early Diabetic Kidney Disease? J Clin Res Pediatr Endocrinol 2025; 17:168-175. [PMID: 39711006 PMCID: PMC12118320 DOI: 10.4274/jcrpe.galenos.2024.2024-5-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024] Open
Abstract
Objective To compare serum mitochondrial open reading frame of 12S rRNA-c (MOTS-c) levels, a new potential biomarker for oxidative stress, in children with type 1 diabetes mellitus (T1DM) and healthy children. A further aim was to investigate serum MOTS-c levels as a potential early indicator of diabetic kidney disease (DKD) by correlating levels with changes in glomerular filtration and microalbuminuria. Methods Patients with a diagnosis of T1DM and healthy controls were recruited. MOTS-c, urinary albumin excretion, estimated glomerular filtration rate (eGFR), and hemoglobin A1c (HbA1c) were evaluated and clinical features and anthropometric measurements were collected. Patients were stratified according to diabetes duration, presence of albuminuria, glomerular hyperfiltration, eGFR decline and metabolic control. Results The T1DM group included 82 [female:male (F:M) 1:1.64] patients while the controls numbered 61 (F:M 1:0.97), with respective mean ages of 14.3±3.3 and 10.6±4.2 years (p<0.01). MOTS-c levels were significantly lower in the T1DM group than controls (76.2±1.3 vs 105.2±7.0, p<0.001). No difference was found in MOTS-c levels between patient subgroups categorized by diabetes duration, obesity, metabolic control, hypertension, hyperlipidemia, glomerular hyperfiltration, decline in eGFR, and presence of microalbuminuria. Simple linear regression indicated that MOTS-c was not predictive for DKD. Conclusion MOTS-c levels were lower in children with T1DM than in healthy children. However, the lack of association of MOTS-c with renal biomarkers suggested that it is not an effective early marker for DKD. However, this finding suggests that the onset of oxidative damage and mitochondrial dysfunction in T1DM is independent of DKD. In addition, the results suggests that HbA1c and duration of diabetes are significant risk factors for development of microalbuminuria, while changes in eGFR and microalbuminuria continue to serve as indicators of DKD.
Collapse
Affiliation(s)
- İlknur Girişgen
- Pamukkale University Faculty of Medicine, Department of Pediatric Nephrology, Denizli, Türkiye
| | - Selda Ayça Altıncık
- Pamukkale University Faculty of Medicine, Department of Pediatric Endocrinology, Denizli, Türkiye
| | - Esin Avcı
- Pamukkale University Faculty of Medicine, Department of Biochemistry, Denizli, Türkiye
| | - Murat Öcal
- Batman State Hospital, Clinic of Pediatric Endocrinology, Batman, Türkiye
| | - Tülay Becerir
- Pamukkale University Faculty of Medicine, Department of Pediatric Nephrology, Denizli, Türkiye
| | - Gaye Malaş Öztekin
- Pamukkale University Faculty of Medicine, Department of Biochemistry, Denizli, Türkiye
| | - Bayram Özhan
- Pamukkale University Faculty of Medicine, Department of Pediatric Endocrinology, Denizli, Türkiye
| | - Selçuk Yüksel
- Çanakkale Onsekiz Mart University Faculty of Medicine, Department of Pediatric Nephrology and Rheumatology, Çanakkale, Türkiye
| |
Collapse
|
9
|
Shi B, Yang P, Qiao H, Yu D, Zhang S. Extrachromosomal circular DNA drives dynamic genome plasticity: emerging roles in disease progression and clinical potential. Theranostics 2025; 15:6387-6411. [PMID: 40521196 PMCID: PMC12160019 DOI: 10.7150/thno.111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/29/2025] [Indexed: 06/18/2025] Open
Abstract
Extrachromosomal circular DNA (eccDNA) has emerged as a dynamic and versatile genomic element with key roles in physiological regulation and disease pathology. This review synthesizes current knowledge on eccDNA, covering its classification, biogenesis, detection methods, biological functions, and clinical implications. Once considered rare anomalies, eccDNAs are now recognized as major drivers of oncogene amplification, genomic plasticity, and therapeutic resistance, particularly in cancer. EccDNA subtypes such as microDNA, double minutes, and ecDNA are defined by their structural, genomic, and pathological features. EccDNAs originate through diverse mechanisms including DNA repair, chromothripsis, breakage fusion bridge cycles, and apoptosis, occurring in both normal and stressed cells. Advances in long-read and single-cell sequencing, CRISPR-based synthesis, and computational tools have improved detection and functional analysis. Functionally, eccDNAs contribute to transcriptional amplification, activate immune responses through cGAS-STING signaling, and facilitate intercellular communication. They are found across a range of tissues and disease states-including cancer, cardiovascular, neurological, autoimmune, and metabolic disorders-and serve as both biomarkers and regulatory elements. We introduce the concept of the stress selection theory, which proposes eccDNA as an adaptive reservoir that enhances cellular fitness in response to environmental and therapeutic pressures. Despite growing insights, challenges remain in understanding tissue-specific roles, achieving clinical translation, and standardizing methodologies. Emerging tools in multi-omics, spatial biology, and artificial intelligence are expected to drive future breakthroughs in precision medicine.
Collapse
Affiliation(s)
- Bin Shi
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563006, China
| | - Ping Yang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Huaijin Qiao
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Daojiang Yu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
- Medical College of Tibet University, Lasa 850000, China
| |
Collapse
|
10
|
Pliouta L, Lampsas S, Kountouri A, Korakas E, Thymis J, Kassi E, Oikonomou E, Ikonomidis I, Lambadiari V. Mitochondrial Dysfunction in the Development and Progression of Cardiometabolic Diseases: A Narrative Review. J Clin Med 2025; 14:3706. [PMID: 40507468 PMCID: PMC12155522 DOI: 10.3390/jcm14113706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/19/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Mitochondria play a central role in energy metabolism and continuously adapt through dynamic processes such as fusion and fission. When the balance between these processes is disrupted, it can lead to mitochondrial dysfunction and increased oxidative stress, contributing to the development and progression of various cardiometabolic diseases (CMDs). Their role is crucial in diabetes mellitus (DM), since their dysfunction drives β-cell apoptosis, immune activation, and chronic inflammation through excessive ROS production, worsening endogenous insulin secretion. Moreover, sympathetic nervous system activation and altered dynamics, contribute to hypertension through oxidative stress, impaired mitophagy, endothelial dysfunction, and cardiomyocyte hypertrophy. Furthermore, the role of mitochondria is catalytic in endothelial dysfunction through excessive reactive oxygen species (ROS) production, disrupting the vascular tone, permeability, and apoptosis, while impairing antioxidant defense and promoting inflammatory processes. Mitochondrial oxidative stress, resulting from an imbalance between ROS/Reactive nitrogen species (RNS) imbalance, promotes atherosclerotic alterations and oxidative modification of oxidizing low-density lipoprotein (LDL). Mitochondrial DNA (mtDNA), situated in close proximity to the inner mitochondrial membrane where ROS are generated, is particularly susceptible to oxidative damage. ROS activate redox-sensitive inflammatory signaling pathways, notably the nuclear factor kappa B (NF-κB) pathway, leading to the transcriptional upregulation of proinflammatory cytokines, chemokines, and adhesion molecules. This proinflammatory milieu promotes endothelial activation and monocyte recruitment, thereby perpetuating local inflammation and enhancing atherogenesis. Additionally, mitochondrial disruptions in heart failure promote further ischemic injury and excessive oxidative stress release and impair ATP production and Ca2⁺ dysregulation, contributing to cell death, fibrosis, and decreased cardiac performance. This narrative review aims to investigate the intricate relationship between mitochondrial dysfunction and CMDs.
Collapse
Affiliation(s)
- Loukia Pliouta
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Stamatios Lampsas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Aikaterini Kountouri
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - Emmanouil Korakas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| | - John Thymis
- 2nd Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Eva Kassi
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Vaia Lambadiari
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (L.P.); (S.L.); (A.K.); (E.K.); (V.L.)
| |
Collapse
|
11
|
Shero JA, Lindholm ME, Sandri M, Stanford KI. Skeletal Muscle as a Mediator of Interorgan Crosstalk During Exercise: Implications for Aging and Obesity. Circ Res 2025; 136:1407-1432. [PMID: 40403102 PMCID: PMC12101524 DOI: 10.1161/circresaha.124.325614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 05/24/2025]
Abstract
Physical exercise is critical for preventing and managing chronic conditions, such as cardiovascular disease, type 2 diabetes, hypertension, and sarcopenia. Regular physical activity significantly reduces cardiovascular and all-cause mortality. Exercise also enhances metabolic health by promoting muscle growth, mitochondrial biogenesis, and improved nutrient storage while preventing age-related muscle dysfunction. Key metabolic benefits include increased glucose uptake, enhanced fat oxidation, and the release of exercise-induced molecules called myokines, which mediate interorgan communication and improve overall metabolic function. These myokines and other exercise-induced signaling molecules hold promise as therapeutic targets for aging and obesity-related conditions.
Collapse
Affiliation(s)
- Julia A. Shero
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Maléne E. Lindholm
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California, United States
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Kristin I. Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
12
|
Li X, Zhan F, Qiu G, Lu P, Shen Z, Qi Y, Wu M, Chu M, Feng J, Wen Z, Yao X, Wang A, Jin W, Zhang X, Liao J, Zhang J, Song M, Wang W, Wang X. MOTS-c attenuates lung ischemia-reperfusion injury via MYH9-Dependent nuclear translocation and transcriptional activation of antioxidant genes. Redox Biol 2025; 84:103681. [PMID: 40403491 DOI: 10.1016/j.redox.2025.103681] [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: 04/10/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) following cardiopulmonary bypass (CPB) is driven by oxidative stress during lung ischemia-reperfusion injury (LIRI). Mitochondrial-derived peptide MOTS-c has emerged as a regulator of mitochondrial-nuclear communication, yet its role in CPB-induced ARDS remains unclear. Here, we identify MOTS-c as a critical mediator of endothelial protection against LIRI through MYH9-dependent nuclear translocation and transcriptional activation of antioxidant genes. In rat LIRI models, endothelial cells exhibited the most significant MOTS-c upregulation, correlating with barrier preservation and reduced oxidative stress. Mechanistically, hypoxia-reoxygenation (HR) triggered reactive oxygen species (ROS)-dependent phosphorylation of MYH9 at Ser1943 via casein kinase II subunit alpha (CK2A), enabling MOTS-c binding to MYH9-γ-Actin complexes for nuclear transport. RNA sequencing (RNA-seq) combined with chromatin immunoprecipitation sequencing (ChIP-seq) revealed direct MOTS-c interaction with promoters of antioxidant genes (e.g., HMOX1, NQO1), which harbor antioxidant response elements (AREs). Clinically, serum MOTS-c increments within 24 h post-CPB (ΔMOTS-c) outperformed traditional biomarkers in predicting ARDS incidence, with multivariate models incorporating ΔMOTS-c achieving superior discriminative power (AUC = 0.885). Exogenous MOTS-c administration in rats attenuated lung injury by reducing oxidative damage, inflammation, and mortality, recapitulating endogenous protective mechanisms. Our findings establish MOTS-c as a dual-function molecule-acting via ROS-CK2A-MYH9 signaling to activate nuclear antioxidant defenses and serving as a prognostic biomarker for CPB-related complications. This study bridges mitochondrial dynamics, nuclear transcriptional regulation, and clinical outcomes, offering novel preventive avenues for IRI-associated pathologies.
Collapse
Affiliation(s)
- Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Faliang Zhan
- Department of Cardiothoracic Surgery, Yili Friendship Hospital, Yining, China
| | - Guangfeng Qiu
- Department of Respiratory and Critical Care Medicine, Yili Friendship Hospital, Yining, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Minchao Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Mingyu Chu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jia Feng
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xin Yao
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ao Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wanjun Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xiao Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Junjie Liao
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jialin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Wei Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Canada.
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China; Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| |
Collapse
|
13
|
Strømland PP, Bertelsen BE, Viste K, Chatziioannou AC, Bellerba F, Robinot N, Trolat A, Flågeng MH, Scalbert A, Keski-Rahkonen P, Sears DD, Bonanni B, Gandini S, Johansson H, Mellgren G. Effects of metformin on transcriptomic and metabolomic profiles in breast cancer survivors enrolled in the randomized placebo-controlled MetBreCS trial. Sci Rep 2025; 15:16897. [PMID: 40374694 DOI: 10.1038/s41598-025-01705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 05/07/2025] [Indexed: 05/17/2025] Open
Abstract
Metformin reduces the incidence of breast cancer in patients with obesity and type 2 diabetes. However, our knowledge of the effects of metformin on breast cancer recurrence is limited. Within the randomized double-blind placebo-controlled phase II trial MetBreCS, we examined changes in breast tissue from breast cancer survivors with BMI > 25 kg/m2 after treatment with metformin. To identify metformin-regulated signaling pathways, we integrated the transcriptomic, metabolomic and steroid hormone profiles using bivariate and functional analyses. We identified MS4A1, HBA2, MT-RNR1, MT-RNR2, EGFL6 and FDCSP expression to be differentially expressed in breast tissues from metformin-treated postmenopausal women. The integration of transcriptomic and metabolomic profiles revealed down-regulation of immune response genes associated with reduced levels of arginine and citrulline in the metformin-treated group. The integration of transcriptomic and steroid hormone profiles showed an enrichment of steroid hormone biosynthesis and metabolism pathways with highly negatively correlated CYP11A1 and CYP1B1 expression in breast tissue from postmenopausal metformin-treated women. Our results indicate that postmenopausal breast cancer survivors treated with metformin have specific changes in breast tissue gene expression that may prevent the development of new tumors.Trial registration: MetBreCs trial is registered at European Union Clinical Trials Register (EudraCT Protocol # 2015-001001-14) on 07/10/2015.
Collapse
Affiliation(s)
- Pouda Panahandeh Strømland
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn-Erik Bertelsen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Kristin Viste
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Federica Bellerba
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nivonirina Robinot
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Amarine Trolat
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Marianne Hauglid Flågeng
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Augustin Scalbert
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Dorothy D Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Moores Cancer Center, University of California San Diego, La Jolla, San Diego, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
14
|
Ran Y, Guo Z, Zhang L, Li H, Zhang X, Guan X, Cui X, Chen H, Cheng M. Mitochondria‑derived peptides: Promising microproteins in cardiovascular diseases (Review). Mol Med Rep 2025; 31:127. [PMID: 40084698 PMCID: PMC11924172 DOI: 10.3892/mmr.2025.13492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
Mitochondria‑derived peptides (MDPs) are a unique class of peptides encoded by short open reading frames in mitochondrial DNA, including the mitochondrial open reading frame of the 12S ribosomal RNA type‑c (MOTS‑c). Recent studies suggest that MDPs offer therapeutic benefits in various diseases, including neurodegenerative disorders and types of cancer, due to their ability to increase cellular resilience. Mitochondrial dysfunction is a key factor in the onset and progression of cardiovascular diseases (CVDs), such as atherosclerosis and heart failure, as it disrupts energy metabolism, increases oxidative stress and promotes inflammation. MDPs such as humanin and MOTS‑c have emerged as important regulators of mitochondrial health, as they show protective effects against these processes. Recent studies have shown that MDPs can restore mitochondrial function, reduce oxidative damage and alleviate inflammation, thus counteracting the pathological mechanisms that drive CVDs. Therefore, MDPs hold promise as therapeutic agents that are capable of slowing, stopping, or even reversing CVD progression and their use presents a promising strategy for future treatments. However, the clinical application of MDPs remains challenging due to their low bioavailability, poor stability and high synthesis costs. Thus, it is necessary to improve drug delivery systems to enhance the bioavailability of MDPs. Moreover, integrating basic research with clinical trials is essential to bridge the gap between experimental findings and clinical applications.
Collapse
Affiliation(s)
- Yutong Ran
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiliang Guo
- Department of Spinal Surgery, The 80th Group Army Hospital of Chinese PLA, Weifang, Shandong 261021, P.R. China
| | - Lijuan Zhang
- Stroke Centre, Second People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Hong Li
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiaoyun Zhang
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiumei Guan
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiaodong Cui
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Hao Chen
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Min Cheng
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
15
|
Sun H, Gu R, Tang T, Rai KR, Chen JL. Current Perspectives on Functional Involvement of Micropeptides in Virus-Host Interactions. Int J Mol Sci 2025; 26:3651. [PMID: 40332243 PMCID: PMC12026789 DOI: 10.3390/ijms26083651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Micropeptides (miPEPs), encoded by short open reading frames (sORFs) within various genomic regions, have recently emerged as critical regulators of multiple biological processes. In particular, these small molecules are now increasingly being recognized for their role in modulating viral replication, pathogenesis, and host immune responses. Both host miPEPs and virus-derived miPEPs have been noted for their ability to regulate virus-host interactions through diversified mechanisms such as altering protein stability and modulating protein-protein interactions. Although thousands of sORFs have been annotated as having the potential to encode miPEPs, only a small number have been experimentally validated so far, with some directly linked to virus-host interactions and a small subset associated with immune modulation, indicating that the investigation of miPEPs is still in its infancy. The systematic identification, translational status assessment, in-depth characterization, and functional analysis of a substantial fraction of sORFs encoding miPEPs remain largely underexplored. Further studies are anticipated to uncover the intricate mechanisms underlying virus-host interactions, host immune modulation, and the broader biological functions of miPEPs. This article will review the emerging roles of miPEPs in virus-host interactions and host immunity, and discuss the challenges and future perspectives of miPEP studies.
Collapse
Affiliation(s)
- Haowen Sun
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.S.); (R.G.); (T.T.)
| | - Rongrong Gu
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.S.); (R.G.); (T.T.)
| | - Tingting Tang
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.S.); (R.G.); (T.T.)
| | - Kul Raj Rai
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.S.); (R.G.); (T.T.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ji-Long Chen
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (H.S.); (R.G.); (T.T.)
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
16
|
Zhong A, Li S, Zhang J, Zhao J, Yao C. Endogenous micropeptides as potential diagnostic biomarkers and therapeutic drugs. Front Pharmacol 2025; 16:1545575. [PMID: 40264667 PMCID: PMC12011824 DOI: 10.3389/fphar.2025.1545575] [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: 12/15/2024] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
Micropeptides, these small proteins derived from non-coding RNA, typically consist of no more than 100 amino acids in length. Despite the challenges in analysis and identification, their various critical functions within organisms cannot be overlooked. They play a significant role in maintaining energy metabolism balance, regulating the immune system, and influencing the development of tumors, which also gives them a decisive impact on the occurrence and development of various diseases. This review aims to outline the role and potential value of micropeptides, introducing their tissue classification and distribution, biological functions, and mechanisms, with a focus on their potential as diagnostic markers and therapeutic drugs.
Collapse
Affiliation(s)
- Aixi Zhong
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University, Dalian, China
| | - Shuai Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingxuan Zhang
- Zhongshan College of Dalian Medical University, Dalian, China
| | - Jingyuan Zhao
- Central Hospital of Dalian University of Technology, Dalian, China
| | - Chenhui Yao
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Schlesinger D, Dirks C, Navarro C, Lafranchi L, Spinner A, Raja GL, Mun-Sum Tong G, Eirich J, Martinez TF, Elsässer SJ. A large-scale sORF screen identifies putative microproteins involved in cancer cell fitness. iScience 2025; 28:111884. [PMID: 40124493 PMCID: PMC11929002 DOI: 10.1016/j.isci.2025.111884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/11/2024] [Accepted: 01/21/2025] [Indexed: 03/25/2025] Open
Abstract
The human genome contains thousands of potentially coding short open reading frames (sORFs). While a growing set of microproteins translated from these sORFs have been demonstrated to mediate important cellular functions, the majority remains uncharacterized. In our study, we performed a high-throughput CRISPR-Cas9 knock-out screen targeting 11,776 sORFs to identify microproteins essential for cancer cell line growth. We show that the CENPBD2P gene encodes a translated sORF and promotes cell fitness. We selected five additional candidate sORFs encoding microproteins between 11 and 63 amino acids in length for further functional assessment. Green fluorescent protein fusion constructs of these microproteins localized to distinct subcellular compartments, and the majority showed reproducible biochemical interaction partners. Studying the fitness and transcriptome of sORF knock-outs and complementation with the corresponding microprotein, we identify rescuable phenotypes while also illustrating the limitations and caveats of our pipeline for sORF functional screening and characterization.
Collapse
Affiliation(s)
- Dörte Schlesinger
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Christopher Dirks
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Carmen Navarro
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Lorenzo Lafranchi
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anna Spinner
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
| | - Glancis Luzeena Raja
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
| | - Gregory Mun-Sum Tong
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92617, USA
| | - Jürgen Eirich
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- University of Münster, Institute of Plant Biology and Biotechnology (IBBP), 48143 Münster, Germany
| | - Thomas Farid Martinez
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92617, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92617, USA
| | - Simon Johannes Elsässer
- Science for Life Laboratory, Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Division of Genome Biology, 17165 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 17165 Stockholm, Sweden
| |
Collapse
|
18
|
Giacomello E, Nicoletti C, Canato M, Toniolo L. Exercise Mimetics in Aging: Suggestions from a Systematic Review. Nutrients 2025; 17:969. [PMID: 40289996 PMCID: PMC11944853 DOI: 10.3390/nu17060969] [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: 02/17/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Growth in the aging world population is accompanied by an increase in comorbidities, profoundly impacting the quality of life of older people. This development has motivated a large effort to investigate the mechanisms underlying aging and the search for countermeasures. The most investigated strategies envisage the control of diet and physical exercise, which exploit both common and distinct mechanisms to promote health. Since the application of nutritional and exercise protocols to aged persons introduces several issues due to their disabled state, some strategies have been developed. The nutritional approach exploits a wide range of compounds, including calorie restriction mimetics, supplements, antioxidants, and others. In the context of exercise, in recent years, molecules able to provide similar effects to exercise, the so-called exercise mimetics, have been developed. Methods: To have a better perspective on exercise mimetics and their connection with nutrition, we performed a systematic search of the PubMed and Scopus databases using the term "exercise mimetics". Results: In total, 97 research articles were selected and discussed. The present review provides evidence of the presence of multiple exercise-mimetic compounds and physical strategies that can target metabolic pathways, oxidative stress defense mechanisms, or myokine modulation. Conclusions: Interestingly, this review highlights that an important number of exercise mimetics are represented by products of natural origin and supplements assimilable with diet. This evidence provides a further link between exercise and nutrition and confers a central role on nutrition in the context of exercise mimetics.
Collapse
Affiliation(s)
- Emiliana Giacomello
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Claudio Nicoletti
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy;
| | - Marta Canato
- Laboratory of Muscle Biophysics, Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| | - Luana Toniolo
- Laboratory of Muscle Biophysics, Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| |
Collapse
|
19
|
Ikonomidis I, Pavlidis G, Pliouta L, Katogiannis K, Maratou E, Thymis J, Michalopoulou E, Prentza V, Katsanaki E, Vlachomitros D, Kountouri A, Korakas E, Andreadou I, Kouretas D, Parissis J, Lambadiari V. Effects of Glucagon-Like Peptide-1 Receptor Agonists, Sodium-Glucose Cotransporter-2 Inhibitors, and Their Combination on Neurohumoral and Mitochondrial Activation in Patients With Diabetes. J Am Heart Assoc 2025; 14:e039129. [PMID: 40008510 DOI: 10.1161/jaha.124.039129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND We investigated the effects of the combined treatment with glucagon like peptide-1 receptor agonists (GLP-1RA) and sodium-glucose cotransporter-2 inhibitors (SGLT-2i) on NT-proBNP (N-terminal pro-brain natriuretic peptide), GDF-15 (growth differentiation factor 15), and MOTS-c (mitochondrial-derived peptide-c) in patients with type 2 diabetes (T2D) and high or very high cardiovascular risk. METHODS We studied 163 consecutive patients with type 2 diabetes who were treated with insulin (n=40), liraglutide (n=41), empagliflozin (n=42), or their combination (GLP-1RA+SGLT-2i) (n=40) and were matched using propensity score analysis. We measured the following at baseline and 4 and 12 months of treatment: (1) NT-proBNP, GDF-15, and MOTS-c; (2) 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid), and (3) left ventricular global longitudinal strain, left atrial strain during atrial reservoir phase, and global work index using speckle-tracking imaging. RESULTS At 12 months, GLP-1RA, SGLT-2i, and their combination showed a greater reduction of NT-proBNP (-43.1% versus -54.2% versus -56.9% versus -14.7%) and GDF-15 than insulin. Only treatment with SGLT-2i and GLP-1RA+SGLT-2i improved MOTS-c. GLP-1RA, SGLT-2i, or GLP-1RA+SGLT-2i provided an increase of global longitudinal strain, left atrial strain, and global work index compared with insulin. In all patients, the reduction of NT-proBNP was associated with the improvement of global longitudinal strain, left atrial strain during atrial reservoir phase, and global work index; the decrease of GDF-15 with the increase of ABTS and MOTS-c; and the increase of MOTs-c with improved global longitudinal strain and constructive myocardial work at 12 months (P<0.05). CONCLUSIONS Twelve-month treatment with combination of GLP-1RA+SGLT-2i was associated with a greater reduction of neurohumoral markers and increase of antioxidant ability than each treatment alone and insulin. SGLT-2i appear more effective in the improvement of neurohumoral and mitochondrial activation. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03878706.
Collapse
Affiliation(s)
- Ignatios Ikonomidis
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - George Pavlidis
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Loukia Pliouta
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Athens Greece
| | - Konstantinos Katogiannis
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Eirini Maratou
- Laboratory of Clinical Biochemistry, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Athens Greece
| | - John Thymis
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Eleni Michalopoulou
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Vasiliki Prentza
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Eleni Katsanaki
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Dimitrios Vlachomitros
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Aikaterini Kountouri
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Athens Greece
| | - Emmanouil Korakas
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Athens Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy National and Kapodistrian University of Athens Athens Greece
| | - Dimitrios Kouretas
- Department of Biochemistry and Biotechnology University of Thessaly Larissa Greece
| | - John Parissis
- 2nd Department of Cardiology, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Greece
| | - Vaia Lambadiari
- 2nd Propaedeutic Department of Internal Medicine, Research Unit and Diabetes Center, School of Medicine University General Hospital "Attikon", National and Kapodistrian University of Athens Athens Greece
| |
Collapse
|
20
|
Shi C, Liu F, Su X, Yang Z, Wang Y, Xie S, Xie S, Sun Q, Chen Y, Sang L, Tan M, Zhu L, Lei K, Li J, Yang J, Gao Z, Yu M, Wang X, Wang J, Chen J, Zhuo W, Fang Z, Liu J, Yan Q, Neculai D, Sun Q, Shao J, Lin W, Liu W, Chen J, Wang L, Liu Y, Li X, Zhou T, Lin A. Comprehensive discovery and functional characterization of the noncanonical proteome. Cell Res 2025; 35:186-204. [PMID: 39794466 PMCID: PMC11909191 DOI: 10.1038/s41422-024-01059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 11/14/2024] [Indexed: 01/13/2025] Open
Abstract
The systematic identification and functional characterization of noncanonical translation products, such as novel peptides, will facilitate the understanding of the human genome and provide new insights into cell biology. Here, we constructed a high-coverage peptide sequencing reference library with 11,668,944 open reading frames and employed an ultrafiltration tandem mass spectrometry assay to identify novel peptides. Through these methods, we discovered 8945 previously unannotated peptides from normal gastric tissues, gastric cancer tissues and cell lines, nearly half of which were derived from noncoding RNAs. Moreover, our CRISPR screening revealed that 1161 peptides are involved in tumor cell proliferation. The presence and physiological function of a subset of these peptides, selected based on screening scores, amino acid length, and various indicators, were verified through Flag-knockin and multiple other methods. To further characterize the potential regulatory mechanisms involved, we constructed a framework based on artificial intelligence structure prediction and peptide‒protein interaction network analysis for the top 100 candidates and revealed that these cancer-related peptides have diverse subcellular locations and participate in organelle-specific processes. Further investigation verified the interacting partners of pep1-nc-OLMALINC, pep5-nc-TRHDE-AS1, pep-nc-ZNF436-AS1 and pep2-nc-AC027045.3, and the functions of these peptides in mitochondrial complex assembly, energy metabolism, and cholesterol metabolism, respectively. We showed that pep5-nc-TRHDE-AS1 and pep2-nc-AC027045.3 had substantial impacts on tumor growth in xenograft models. Furthermore, the dysregulation of these four peptides is closely correlated with clinical prognosis. Taken together, our study provides a comprehensive characterization of the noncanonical proteome, and highlights critical roles of these previously unannotated peptides in cancer biology.
Collapse
Affiliation(s)
- Chengyu Shi
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Fangzhou Liu
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Xinwan Su
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Zuozhen Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Ying Wang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Shanshan Xie
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine and Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaofang Xie
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, Zhejiang, China
| | - Qiang Sun
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yu Chen
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Lingjie Sang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Manman Tan
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Linyu Zhu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Kai Lei
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Junhong Li
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Jiecheng Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Zerui Gao
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Meng Yu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Xinyi Wang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Junfeng Wang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China
| | - Jing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Zhuo
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine and Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Fang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Liu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, China
- Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Qingfeng Yan
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dante Neculai
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Qiming Sun
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jianzhong Shao
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiqiang Lin
- Department of Nephrology, Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Wei Liu
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jian Chen
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liangjing Wang
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine and Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yang Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xu Li
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, Zhejiang, China
| | - Tianhua Zhou
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of Cell Biology and Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Aifu Lin
- The Center for RNA Medicine, International Institutes of Medicine, International School of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, Zhejiang, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan, Zhejiang, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Ajala I, Vanderperre B. Non-canonical ORFs-derived protein products in mitochondria: A multifaceted exploration of their functions in health and disease. Protein Sci 2025; 34:e70053. [PMID: 39969119 PMCID: PMC11837024 DOI: 10.1002/pro.70053] [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: 10/09/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/20/2025]
Abstract
Traditionally, eukaryotic mRNAs were perceived as inherently monocistronic. However, recent insights from ribosome profiling (Ribo-seq) and proteomics studies challenge this paradigm. These investigations reveal that, beyond the currently annotated reference proteins (RefProts), there exist additional proteins known as alternative proteins (AltProts) and small open reading frames derived microproteins encoded in regions of mRNAs previously considered untranslated or in non-coding transcripts. This experimental evidence broadens the spectrum of functional proteins within cells, tissues, and organs, potentially offering crucial insights into biological processes. Notably, a significant proportion of these newly identified AltProts and microproteins demonstrates localization in mitochondria, contributing to the functions of mitochondrial complexes. This review delves into the overlooked realm of the alternative proteome within mitochondria, exploring the role of nuclear or mitochondrial-genome-encoded AltProts and microproteins in physiological and pathological cellular processes.
Collapse
Affiliation(s)
- Ikram Ajala
- Department of Biological Sciences, Université du Québec à MontréalCERMO‐FC Research CenterMontrealQuebecCanada
- Network for Research on Protein FunctionEngineering and Applications (PROTEO)MontréalQuebecCanada
| | - Benoît Vanderperre
- Department of Biological Sciences, Université du Québec à MontréalCERMO‐FC Research CenterMontrealQuebecCanada
- Network for Research on Protein FunctionEngineering and Applications (PROTEO)MontréalQuebecCanada
| |
Collapse
|
22
|
Wang DD, Xu B, Sun JJ, Sui M, Li SP, Chen YJ, Zhang YL, Wu JB, Teng SY, Pang QF, Hu CX. MOTS-c mimics remote ischemic preconditioning in protecting against lung ischemia-reperfusion injury by alleviating endothelial barrier dysfunction. Free Radic Biol Med 2025; 229:127-138. [PMID: 39827923 DOI: 10.1016/j.freeradbiomed.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Remote ischemic preconditioning (RIPC) induces the expression of unidentified protective cytokines that mitigate lung ischemia-reperfusion injury (LIRI). This study hypothesizes that MOTS-c, a mitokine with potent protective effects against mitochondrial damage, contributes to RIPC-mediated protection by alleviating endothelial barrier dysfunction. In human lung transplantation patients, serum levels of MOTS-c significantly decreased following IR injury but were markedly increased when RIPC was performed prior to transplantation. Similarly, in a mouse model of LIRI, RIPC restored serum MOTS-c levels and improved lung injury outcomes. Intravenous administration of MOTS-c in mice replicated the protective effects observed with RIPC. Mechanistic studies demonstrated that repeated hypoxia in human primary skeletal muscle immortalized cells (HPSMIC) led to the secretion of conditioned media that protected HUVECs from OGD/R-induced injury; silencing MOTS-c abolished these protective effects. Further investigations using nuclear factor erythroid 2-related factor 2 (Nrf2) knockout mice and the Nrf2 inhibitor ML385 revealed that MOTS-c exerts its protective function by increasing Nrf2 protein levels, thereby maintaining endothelial barrier integrity. In conclusion, this study identifies MOTS-c as a novel mediator of RIPC's protective effects against LIRI and highlights its potential as a therapeutic alternative for preventing lung injury and preserving vascular endothelial function.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China; Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Bo Xu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Jiao-Jiao Sun
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China; Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Meng Sui
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Sheng-Peng Li
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China
| | - Yi-Jing Chen
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China
| | - Yan-Li Zhang
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China
| | - Jin-Bo Wu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Shi-Yong Teng
- Department of Anesthesiology, First Hospital of Jilin University, Changchun, Jilin, China
| | - Qing-Fang Pang
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu province, China.
| | - Chun-Xiao Hu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| |
Collapse
|
23
|
Liu YJ, Sulc J, Auwerx J. Mitochondrial genetics, signalling and stress responses. Nat Cell Biol 2025; 27:393-407. [PMID: 40065146 DOI: 10.1038/s41556-025-01625-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/22/2025] [Indexed: 03/15/2025]
Abstract
Mitochondria are multifaceted organelles with crucial roles in energy generation, cellular signalling and a range of synthesis pathways. The study of mitochondrial biology is complicated by its own small genome, which is matrilineally inherited and not subject to recombination, and present in multiple, possibly different, copies. Recent methodological developments have enabled the analysis of mitochondrial DNA (mtDNA) in large-scale cohorts and highlight the far-reaching impact of mitochondrial genetic variation. Genome-editing techniques have been adapted to target mtDNA, further propelling the functional analysis of mitochondrial genes. Mitochondria are finely tuned signalling hubs, a concept that has been expanded by advances in methodologies for studying the function of mitochondrial proteins and protein complexes. Mitochondrial respiratory complexes are of dual genetic origin, requiring close coordination between mitochondrial and nuclear gene-expression systems (transcription and translation) for proper assembly and function, and recent findings highlight the importance of the mitochondria in this bidirectional signalling.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
24
|
Di Pierro E, Di Stefano V, Migone De Amicis M, Graziadei G. Are Mitochondria a Potential Target for Treating β-Thalassemia? J Clin Med 2025; 14:1095. [PMID: 40004626 PMCID: PMC11856739 DOI: 10.3390/jcm14041095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/14/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The inherited genetic disorder β-thalassemia affects the hematopoietic system and is caused by the low production or absence of adult hemoglobin (HbA). Ineffective erythropoiesis is the hallmark of β-thalassemia pathophysiology and is characterized by an erythropoietin-driven substantial increase in erythroblast proliferation, coupled with an increase in late-stage precursor apoptosis, which results in low levels of circulating mature red blood cells (RBCs) and chronic anemia. Mitochondrial dysfunction commonly occurs in these cells because of the increased demand for energy production and the need to manage abnormal hemoglobin chain synthesis. Moreover, several studies have highlighted the importance of gradual mitochondrial clearance for mature erythroid cell production. This review offers an overview of the mitochondrial role in essential cellular processes, particularly those crucial for maintaining RBC health and function. Additionally, recent evidence regarding the contribution of mitochondrial dysfunction to the pathophysiology and severity of β-thalassemia is discussed, along with updated insights into indirect mitochondria-targeting treatments, which present potential pharmacological targets.
Collapse
Affiliation(s)
- Elena Di Pierro
- SC di Medicina ad Indirizzo Metabolico, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122 Milano, Italy; (V.D.S.); (M.M.D.A.); (G.G.)
| | | | | | | |
Collapse
|
25
|
Yen K, Miller B, Kumagai H, Silverstein A, Cohen P. Mitochondrial-derived microproteins: from discovery to function. Trends Genet 2025; 41:132-145. [PMID: 39690001 PMCID: PMC11794013 DOI: 10.1016/j.tig.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024]
Abstract
Given the uniqueness of the mitochondria, and the fact that they have their own genome, mitochondrial-derived microproteins (MDPs) are similar to, but different from, nuclear-encoded microproteins. The discovery of an increasing number of microproteins from this organelle and the importance of mitochondria to cellular and organismal health make it a priority to study this novel class of proteins in search of possible therapeutic targets and cures. In this review, we discuss the history of MDP discovery, describe the function of each MDP, and conclude with future goals and techniques to help discover more MDPs.
Collapse
Affiliation(s)
- Kelvin Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | - Brendan Miller
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hiroshi Kumagai
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Ana Silverstein
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
26
|
Feng Y, Rao Z, Tian X, Hu Y, Yue L, Meng Y, Zhong Q, Chen W, Xu W, Li H, Hu Y, Shi R. Endurance training enhances skeletal muscle mitochondrial respiration by promoting MOTS-c secretion. Free Radic Biol Med 2025; 227:619-628. [PMID: 39706498 DOI: 10.1016/j.freeradbiomed.2024.12.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
The mitochondrial open reading frame of 12S rRNA-c (MOTS-c) is a biologically active mitochondria-derived peptide. However, the relationship between MOTS-c, skeletal muscle mitochondrial function, and endurance exercise adaptations is unknown. Here, we tested indices such as maximal oxygen uptake and serum MOTS-c levels in marathon runners and sedentary subjects. In addition, we tested aerobic exercise capacity, skeletal muscle mitochondrial respiration rate, and serum MOTS-c levels in mice subjected to long-term endurance training groups and sedentary groups. Our results indicated a close association between serum MOTS-c levels and aerobic exercise capacity. Circulating MOTS-c levels are expected to be an important indicator for predicting aerobic exercise capacity and assessing body fat status, endurance training load, and physical function. More importantly, we found that endurance training may enhance the mitochondrial respiratory function of skeletal muscle by promoting the secretion of MOTS-c and activating the AMPK/PGC-1α pathway.
Collapse
Affiliation(s)
- Yiwei Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Zhijian Rao
- School of Physical Education, Shanghai Normal University, Shanghai, 200234, China
| | - Xu Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yi Hu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Liantian Yue
- School of Sport, Exercise and Health Sciences, Loughborough University, UK
| | - Yifan Meng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Qiuling Zhong
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Wei Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Wenlong Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Haoran Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yingjia Hu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Rengfei Shi
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
27
|
Gao Y, Zhang J, Cao M, Zhang Y, Cao M, Gu W, Wang M. MDPAO1 peptide from human milk enhances brown adipose tissue thermogenesis and mitigates obesity. Mol Cell Endocrinol 2025; 597:112443. [PMID: 39710295 DOI: 10.1016/j.mce.2024.112443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 11/19/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
The regulatory effect of breastfeeding on offspring metabolism has garnered significant attention as an effective strategy in combating childhood obesity. However, the underlying mechanism remains largely unknown. Through integrated analysis of multiple human milk peptide databases and functional screening, MDPAO1 (milk-derived peptide associated with obesity 1) was identified as having potential activity in promoting the expression of thermogenic genes. In lactating mice, intervention with MDPAO1 enhanced the thermogenic phenotype of brown adipose tissue (BAT) and overall metabolic activity. Moreover, MDPAO1 intervention led to reduced body weight gain, increased brown fat mass, and improved glucose tolerance and insulin sensitivity in a mouse model of high-fat diet (HFD)-induced obesity. RNA-seq analysis of BAT post-MDPAO1 intervention revealed close association with mitochondrial oxidative respiratory chain and mitophagy. Subsequent in vitro experiments conducted on primary brown adipocytes confirmed that MDPAO1 inhibited mitophagy, increased mitochondrial mass, and elevated levels of mitochondrial respiratory chain complexes. In conclusion, this study underscores the potential of MDPAO1, a peptide enriched in breast milk, in activating the thermogenic phenotype of brown adipose tissue and mitigating obesity, thus offering novel insights into the mechanisms underlying breastfeeding's role in preventing childhood obesity.
Collapse
Affiliation(s)
- Yao Gao
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Jiahui Zhang
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Mengda Cao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210044, China
| | - Yiting Zhang
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, China; Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, China
| | - Minkai Cao
- Department of Obstetrics and Gynecology, Affiliated Women's Hospital of Jiangnan University ,Wuxi Maternity and Child Health Care Hospital, Wuxi 214002, China.
| | - Wei Gu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| | - Mingxin Wang
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, China; Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, Wuxi, 214023, China.
| |
Collapse
|
28
|
Yang M, Chen W, He L, Wang X, Liu D, Xiao L, Sun L. The Role of Mitokines in Diabetic Nephropathy. Curr Med Chem 2025; 32:1276-1287. [PMID: 37921178 DOI: 10.2174/0109298673255403230919061828] [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: 03/28/2023] [Revised: 08/06/2023] [Accepted: 08/17/2023] [Indexed: 11/04/2023]
Abstract
Diabetic nephropathy (DN) has gradually become one of the main causes of end-stage renal disease (ESRD). However, there is still a lack of effective preventive measures to delay its progression. As the energy factory in the cell, mitochondria play an irreplaceable role in maintaining cell homeostasis. Interestingly, recent studies have shown that in addition to maintaining homeostasis in cells in which mitochondria reside, when mitochondrial perturbations occur in one tissue, distal tissues can also sense and act through mitochondrial stress response pathways through a group of proteins or peptides called "mitokines". Here, we reviewed the mitokines that have been found thus far and summarized their research progress in DN. Finally, we explored the possibility of mitokines as potential therapeutic targets for DN.
Collapse
Affiliation(s)
- Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xi Wang
- Department of Nutrition, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Di Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
29
|
Zhao X, Zhang J, Li C, Kuang W, Deng J, Tan X, Li C, Li S. Mitochondrial mechanisms in Treg cell regulation: Implications for immunotherapy and disease treatment. Mitochondrion 2025; 80:101975. [PMID: 39491776 DOI: 10.1016/j.mito.2024.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining immune homeostasis and preventing autoimmune diseases. Recent advances in immunometabolism have revealed the pivotal role of mitochondrial dynamics and metabolism in shaping Treg functionality. Tregs depend on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to support their suppressive functions and long-term survival. Mitochondrial processes such as fusion and fission significantly influence Treg activity, with mitochondrial fusion enhancing bioenergetic efficiency and reducing reactive oxygen species (ROS) production, thereby promoting Treg stability. In contrast, excessive mitochondrial fission disrupts ATP synthesis and elevates ROS levels, impairing Treg suppressive capacity. Furthermore, mitochondrial ROS act as critical signaling molecules in Treg regulation, where controlled levels stabilize FoxP3 expression, but excessive ROS leads to mitochondrial dysfunction and immune dysregulation. Mitophagy, as part of mitochondrial quality control, also plays an essential role in preserving Treg function. Understanding the intricate interplay between mitochondrial dynamics and Treg metabolism provides valuable insights for developing novel therapeutic strategies to treat autoimmune disorders and enhance immunotherapy in cancer.
Collapse
Affiliation(s)
- Xiaozhen Zhao
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Junmei Zhang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Caifeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Weiying Kuang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jianghong Deng
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xiaohua Tan
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chao Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shipeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Shen H, Nie J, Wang X, Li G, Zhao L, Jin Y, Jin L. MOTS-c relieves hepatocellular carcinoma resistance to TRAIL-induced apoptosis under hypoxic conditions by activating MEF2A. Exp Cell Res 2025; 444:114354. [PMID: 39581216 DOI: 10.1016/j.yexcr.2024.114354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Mitochondrial ORF of the 12S rRNA type-c (MOTS-c) as an AMPK agonist can regulate the expression of adaptive nuclear genes to promote cell homeostasis. However, the investigation of MOTS-c in hepatocellular carcinoma (HCC) is insufficient. This study aims to reveal the role of MOTS-c on HCC cell apoptosis. METHODS Huh7 and HCCLM3 cells were incubated with MOTS-c under a hypoxic condition. MOTS-c levels were quantified by enzyme-linked immunosorbent assay in the peripheral blood of HCC patients and healthy controls. Cell viability was detected by 3-(4,5-Dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis was investigated by flow cytometry and Tunel assay. Protein expression was detected by western blotting or immunohistochemistry assay. Dual-luciferase reporter assay and chromatin immunoprecipitation assay were performed to identify the association among myocyte enhancer factor 2A (MEF2A), death receptor 4 (DR4) and DR5. A tumor-bearing nude mouse model was conducted to assess the effect of MOTS-c on HCC tumor formation in vivo. RESULTS MOTS-c levels in the peripheral blood of HCC patients were significantly lower compared to healthy individuals. MOTS-c promoted HCC cell apoptosis under hypoxia conditions. Hypoxia stimulation decreased the protein expression of MEF2A, DR4, DR5, fas-associating via death domain (FADD) and caspase-8, while these effects were attenuated after MOTS-c treatment. MOTS-c induced TRAIL-induced apoptosis of HCC cells by activating MEF2A through the phosphorylation of AMPK under hypoxia treatment. In addition, MEF2A transcriptionally up-regulated DR4 and DR5. MOTS-c activated MEF2A to regulate DR4 and DR5 expression, further mediating TRAIL-induced apoptosis. Further, MOTS-c treatment relieved hypoxia-induced tumor growth in vivo. CONCLUSION MOTS-c relieved hypoxia-induced HCC cell resistance to TRAIL-caused apoptosis by activating MEF2A.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Apoptosis/drug effects
- Animals
- Mice
- TNF-Related Apoptosis-Inducing Ligand/metabolism
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- MEF2 Transcription Factors/metabolism
- MEF2 Transcription Factors/genetics
- Mice, Nude
- Cell Line, Tumor
- Male
- Female
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Mice, Inbred BALB C
- Xenograft Model Antitumor Assays
- Middle Aged
- Cell Hypoxia/drug effects
Collapse
Affiliation(s)
- Haiying Shen
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China.
| | - Junjie Nie
- Department of Nuclear Medicine, Jilin People's Hospital, Jilin 132001, Jilin Province, PR China
| | - Xiaojun Wang
- School of Public Health, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Guangqing Li
- Department of Computer Application, School of Biomedical Engineering, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Liwei Zhao
- Department of Pathology, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Yuji Jin
- Department of Medical Genetics, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Lianhai Jin
- Hypoxia and Health Medicine Research Center, Jilin Medical University, Jilin 132013, Jilin Province, PR China.
| |
Collapse
|
31
|
Fu Y, Tang M, Duan Y, Pan Y, Liang M, Yuan J, Wang M, Laher I, Li S. MOTS-c regulates the ROS/TXNIP/NLRP3 pathway to alleviate diabetic cardiomyopathy. Biochem Biophys Res Commun 2024; 741:151072. [PMID: 39616938 DOI: 10.1016/j.bbrc.2024.151072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
Chronic low-grade inflammation is a characteristic of diabetes, which often culminates in cardiovascular events including myocardial damage, thereby increasing the risk of debilitating cardiac complications. The mitochondria-derived peptide MOTS-c regulates glucose and lipid metabolism while improving insulin resistance, making it a potential candidate for the treatment of diabetes and cardiovascular diseases. We investigated the impact of MOTS-c on cardiac structure and inflammation in diabetic rats induced by a high-sugar-fat diet combined with low-dose streptozotocin (30 mg/kg, i.p.). Our results confirm that high glucose levels activate the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome and increase reactive oxygen species (ROS), ultimately leading to myocardial injury. Furthermore, treatment with MOTS-c (0.5 mg/kg/day, i.p.) for 8 weeks reduced the expression of ROS/TXNIP/NLRP3 pathway proteins to inhibit the diabetic myocardial inflammatory response. These findings suggested that MOTS-c alleviates myocardial damage by inhibiting the ROS/TXNIP/NLRP3 pathway.
Collapse
Affiliation(s)
- Yu Fu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Mi Tang
- School of Physical Education, Xihua University, Chengdu, China
| | - Yimei Duan
- School of Physical Education, Sichuan Normal University, Chengdu, China
| | - Yanrong Pan
- School of Physical Education, Sichuan Minzu College, Kangding, China
| | - Min Liang
- College of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Manda Wang
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Medicine, University of British Columbia, Vancouver, Canada
| | - Shunchang Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China.
| |
Collapse
|
32
|
Li M, Liu Z, Cao X, Xiao W, Wang S, Zhao C, Zhao Y, Xie Y. [Gly14]-Humanin ameliorates high glucose-induced endothelial senescence via SIRT6. Sci Rep 2024; 14:30924. [PMID: 39730568 DOI: 10.1038/s41598-024-81878-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: 06/02/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
High glucose (HG) induced endothelial senescence is related to endothelial dysfunction and cardiovascular complications in diabetic patients. Humanin, a member of mitochondrial derived peptides (MDPs), is thought to contribute to aging-related cardiovascular protection. The goal of the study is to explore the pathogenesis of HG-induced endothelial senescence and potential anti-senescent effects of Humanin. Human umbilical vein endothelial cells (HUVECs) were exposed to glucose to induce senescence, determined by β-galactosidase staining and the expressions of p21, p53, and p16. A clinically relevant dose of HG (15 mM, HG) induced endothelial senescence after 72 h incubation without elevated apoptosis. HG-induced senescence was attributed to the induction of reactive oxygen species (ROS) caused by SIRT6 downregulation, as ROS inhibitor N-acetyl cysteine blocked HG-induced senescence, while inactivation of SIRT6 increased ROS levels and promoted senescence. Strikingly. pretreatment with [Gly14]-Humanin (HNG) antagonized the downregulation of SIRT6 in response to HG and alleviated ROS production and cell senescence. HG-induced reduction of SIRT6 results in ROS overproduction and endothelial senescence. Humanin protects against HG-induced endothelial senescence via SIRT6. This study provides new directions for biological products related to Humanin to be a potential candidate for the prevention of vascular aging in diabetes.
Collapse
Affiliation(s)
- Muqin Li
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of Endocrinology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, JiangSu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215004, China
| | - Zhihua Liu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xueqin Cao
- Department of Endocrinology, The Fourth Affiliated Hospital of Soochow University, Chongwen Road No. 9, Suzhou, 215000, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Shurong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Chengyuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of endocrinology, Taizhou school of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Soochow Medical College of Soochow University, Suzhou, 215123, China.
| | - Ying Xie
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
33
|
Coskun E, Ekmekci OB, Gungor Z, Tuten A, Oncul M, Hamzaoğlu K, Gok K, Ekmekci H. Evaluation of vascular peroxidase 1, humanin, MOTS-c and miR-200c expression levels in untreated preeclampsia patients. Mol Biol Rep 2024; 52:66. [PMID: 39704971 DOI: 10.1007/s11033-024-10148-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: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The objective of this study was to evaluate the levels of Vascular Peroxidase 1 (VPO1), humanin, and MOTS-c in relation to miR-200c expression in untreated preeclamptic pregnancies, and to compare these findings with endoglin levels. METHODS AND RESULTS In this study, blood samples were collected from preeclamptic patients presenting to the clinic prior to the initiation of treatment. The levels of endoglin, VPO1, humanin, and MOTS-c were measured using enzyme-linked immunosorbent assay (ELISA), while miR-200c expression was quantified using reverse transcription polymerase chain reaction (RT-PCR). Receiver operating characteristic (ROC) analysis was performed to assess diagnostic accuracy. Statistical significance was determined at p < 0.05. The levels of endoglin, VPO1, and miR-200c were found to be significantly elevated in the preeclampsia group compared to the control group (p < 0.05), whereas MOTS-c levels were significantly reduced (p < 0.05). No significant difference was observed in humanin levels between the two groups. A positive correlation was identified between endoglin levels and VPO1 (r = 0.943, p < 0.001), humanin (r = 0.421, p < 0.01), and uric acid (r = 0.314, p = 0.02) in the preeclamptic group. CONCLUSIONS Our findings suggest that the elevation of VPO1 and miR-200c levels, along with the reduction of humanin and MOTS-c levels, may contribute to the increased endoglin levels and subsequent endothelial dysfunction observed in preeclampsia. These changes may be associated with the pathogenesis and severity of the disease.
Collapse
Affiliation(s)
- Erkam Coskun
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ozlem Balci Ekmekci
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Gungor
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Abdullah Tuten
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mahmut Oncul
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kubra Hamzaoğlu
- Department of Obstetrics and Gynecology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Koray Gok
- Department of Perinatology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Hakan Ekmekci
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
34
|
Zhang M, Wei J, He C, Sui L, Jiao C, Zhu X, Pan X. Inter- and intracellular mitochondrial communication: signaling hubs in aging and age-related diseases. Cell Mol Biol Lett 2024; 29:153. [PMID: 39695918 DOI: 10.1186/s11658-024-00669-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
Mitochondria are versatile and complex organelles that can continuously communicate and interact with the cellular milieu. Deregulated communication between mitochondria and host cells/organelles has significant consequences and is an underlying factor of many pathophysiological conditions, including the process of aging. During aging, mitochondria lose function, and mitocellular communication pathways break down; mitochondrial dysfunction interacts with mitochondrial dyscommunication, forming a vicious circle. Therefore, strategies to protect mitochondrial function and promote effective communication of mitochondria can increase healthy lifespan and longevity, which might be a new treatment paradigm for age-related disorders. In this review, we comprehensively discuss the signal transduction mechanisms of inter- and intracellular mitochondrial communication, as well as the interactions between mitochondrial communication and the hallmarks of aging. This review emphasizes the indispensable position of inter- and intracellular mitochondrial communication in the aging process of organisms, which is crucial as the cellular signaling hubs. In addition, we also specifically focus on the status of mitochondria-targeted interventions to provide potential therapeutic targets for age-related diseases.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liutao Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chucheng Jiao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
35
|
Yang L, Huang Y, Ding Y, Hu Z, Zhang XS, Su YH, Liu X. Impact of mtDNA-encoded proteins arising from cytosolic translation (mPACT). Sci Bull (Beijing) 2024; 69:3648-3651. [PMID: 39419670 DOI: 10.1016/j.scib.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Affiliation(s)
- Liang Yang
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510000, China
| | - Yile Huang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China
| | - Yingzhe Ding
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China
| | - Zhijuan Hu
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510000, China
| | - Xian Sheng Zhang
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an 271000, China
| | - Ying Hua Su
- State Key Laboratory of Wheat Improvement, College of Life Sciences, Shandong Agricultural University, Tai'an 271000, China.
| | - Xingguo Liu
- Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510000, China.
| |
Collapse
|
36
|
Ahmad EM, Abdelsamad A, El-Shabrawi HM, El-Awady MAM, Aly MAM, El-Soda M. In-silico identification of putatively functional intergenic small open reading frames in the cucumber genome and their predicted response to biotic and abiotic stresses. PLANT, CELL & ENVIRONMENT 2024; 47:5330-5342. [PMID: 39189930 DOI: 10.1111/pce.15104] [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: 01/07/2024] [Revised: 07/13/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024]
Abstract
The availability of high-throughput sequencing technologies increased our understanding of different genomes. However, the genomes of all living organisms still have many unidentified coding sequences. The increased number of missing small open reading frames (sORFs) is due to the length threshold used in most gene identification tools, which is true in the genic and, more importantly and surprisingly, in the intergenic regions. Scanning the cucumber genome intergenic regions revealed 420 723 sORF. We excluded 3850 sORF with similarities to annotated cucumber proteins. To propose the functionality of the remaining 416 873 sORF, we calculated their codon adaptation index (CAI). We found 398 937 novel sORF (nsORF) with CAI ≥ 0.7 that were further used for downstream analysis. Searching against the Rfam database revealed 109 nsORFs similar to multiple RNA families. Using SignalP-5.0 and NLS, identified 11 592 signal peptides. Five predicted proteins interacting with Meloidogyne incognita and Powdery mildew proteins were selected using published transcriptome data of host-pathogen interactions. Gene ontology enrichment interpreted the function of those proteins, illustrating that nsORFs' expression could contribute to the cucumber's response to biotic and abiotic stresses. This research highlights the importance of previously overlooked nsORFs in the cucumber genome and provides novel insights into their potential functions.
Collapse
Affiliation(s)
- Esraa M Ahmad
- Department of Genetics, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Ahmed Abdelsamad
- Department of Genetics, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Hattem M El-Shabrawi
- Plant Biotechnology Department, Genetic Engineering & Biotechnology Division, National Research Center, Giza, Egypt
| | | | - Mohammed A M Aly
- Department of Genetics, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Mohamed El-Soda
- Department of Genetics, Faculty of Agriculture, Cairo University, Giza, Egypt
| |
Collapse
|
37
|
Morena F, Cabrera AR, Jones RG, Schrems ER, Muhyudin R, Washington TA, Murach KA, Greene NP. Transcriptional analysis of cancer cachexia: conserved and unique features across preclinical models and biological sex. Am J Physiol Cell Physiol 2024; 327:C1514-C1531. [PMID: 39466180 PMCID: PMC11684872 DOI: 10.1152/ajpcell.00647.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Studies suggest heterogeneity in cancer cachexia (CC) among models and biological sexes, yet examinations comparing models and sexes are scarce. We compared the transcriptional landscape of skeletal muscle across murine CC models and biological sexes during early and late CC. Global gene expression analyses were performed on gastrocnemius [Lewis lung carcinoma (LLC)], quadriceps (KPC-pancreatic), and tibialis anterior [Colon-26 (C26)-colorectal and ApcMin/+] muscles across biological sexes. Differentially expressed genes (DEGs) were identified using an adj-P value of <0.05, followed by pathway and computational cistrome analyses. Integrating all controls, early and late stages of all models and sexes revealed up to 68% of DEGs and pathways were enriched at early and late CC, indicating a conserved transcriptional profile during CC development. Comparing DEGs and pathways within sexes and across models, in early CC, the transcriptional response was highly heterogeneous. At late stage, 11.5% of upregulated and 10% of downregulated genes were shared between models in males, whereas 18.9% of upregulated and 7% of downregulated DEGs were shared in females. Shared DEGs were enriched in proteasome and mitophagy/autophagy pathways (upregulated), and downregulation of energy metabolism pathways in males only. Between sexes, though the proportion of shared DEGs was low (<16%), similar pathway enrichment was observed, including proteasome and mitophagy at late-stage CC. In early CC, oncostatin M receptor (Osmr) upregulation was the only commonality across all models and sexes, whereas CLOCK and ARNTL/BMAL1 were predicted transcriptional factors associated with dysregulations in all three male models. This study highlights sex and model differences in CC progression and suggests conserved transcriptional changes as potential therapeutic targets.NEW & NOTEWORTHY This study is among the first to integrate and compare the skeletal muscle transcriptional landscape across multiple preclinical models and biological sexes. We highlight that 1) early CC transcriptional changes are two-thirds conserved at late stages, 2) DEGs are largely model and sex specific, and 3) transcriptional factors including CLOCK and ARNTL/BMAL1, which influence early CC gene expression, might represent a global therapeutic target with a chance of efficacy across various cancer types.
Collapse
Affiliation(s)
- Francielly Morena
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ronald G Jones
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ruqaiza Muhyudin
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
38
|
Xiao X, Wang Y, Li T, Wang Q, Luo X, Li J, Gao L. Microproteins encoded by short open reading frames: Vital regulators in neurological diseases. Prog Neurobiol 2024; 243:102694. [PMID: 39586488 DOI: 10.1016/j.pneurobio.2024.102694] [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: 07/08/2024] [Revised: 10/18/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Short open reading frames (sORFs) are frequently overlooked because of their historical classification as non-coding elements or dismissed as "transcriptional noise". However, advanced genomic and proteomic technologies have allowed for screening and validating sORFs-encoded peptides, revealing their fundamental regulatory roles in cellular processes and sparking a growing interest in microprotein biology. In neuroscience, microproteins serve as neurotransmitters in signal transmission and regulate metabolism and emotions, exerting pivotal effects on neurological conditions such as nerve injury, neurogenic tumors, inflammation, and neurodegenerative diseases. This review summarizes the origins, characteristics, classifications, and functions of microproteins, focusing on their molecular mechanisms in neurological disorders. Potential applications, future perspectives, and challenges are discussed.
Collapse
Affiliation(s)
- Xiao Xiao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Yitian Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Tingyu Li
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Qiang Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Xiaolei Luo
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Jingdong Li
- Institute of Hepato-Biliary-Pancreatic-Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637100, PR China.
| | - Linbo Gao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
39
|
Chen HX, Ma YZ, Xie PP, Huang JY, Li LQ, Zhang W, Zhu Y, Zhuang SM, Lin YF. Micropeptide MPM regulates cardiomyocyte proliferation and heart growth via the AKT pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119820. [PMID: 39163918 DOI: 10.1016/j.bbamcr.2024.119820] [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: 04/12/2024] [Revised: 07/26/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
The role of micropeptide in cardiomyocyte proliferation remains unknown. We found that MPM (micropeptide in mitochondria) was highly expressed in cardiomyocytes. Compared to MPM+/+ mice, MPM knockout (MPM-/-) mice exhibited reduction in left ventricular (LV) mass, myocardial thickness and LV fractional shortening. RNA-sequencing analysis in H9c2, a rat cardiomyocyte cell line, identified downregulation of cell cycle-promoting genes as the most significant alteration in MPM-silencing cells. Consistently, gain- and loss-of-function analyses in H9c2 cells revealed that cardiomyocyte proliferation was repressed by silencing MPM but was promoted by overexpressing MPM. Moreover, the cardiomyocytes in the hearts of MPM-/- mice displayed reduced proliferation rates. Mechanism investigations disclosed that MPM is crucial for AKT activation in cardiomyocytes. We also identified an interaction between MPM and PTPMT1, and found that silencing PTPMT1 attenuated the effect of MPM in activating the AKT pathway, whereas inhibition of the AKT pathway abrogated the role of MPM in promoting cardiomyocyte proliferation. Collectively, these results indicate that MPM may promote cardiomyocyte proliferation and thus heart growth by interacting with PTPMT1 to activate the AKT pathway. Our findings identify the novel function and regulatory network of MPM and highlight the importance of micropeptides in cardiomyocyte proliferation and heart growth.
Collapse
Affiliation(s)
- Hua-Xing Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yan-Zhen Ma
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Peng-Peng Xie
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jie-Yi Huang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Lan-Qi Li
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Wei Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Ying Zhu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shi-Mei Zhuang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China.
| | - Yi-Fang Lin
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China.
| |
Collapse
|
40
|
Kumagai H, Kim SJ, Miller B, Zempo H, Tanisawa K, Natsume T, Lee SH, Wan J, Leelaprachakul N, Kumagai ME, Ramirez R, Mehta HH, Cao K, Oh TJ, Wohlschlegel JA, Sha J, Nishida Y, Fuku N, Dobashi S, Miyamoto-Mikami E, Takaragawa M, Fuku M, Yoshihara T, Naito H, Kawakami R, Torii S, Midorikawa T, Oka K, Hara M, Iwasaka C, Yamada Y, Higaki Y, Tanaka K, Yen K, Cohen P. MOTS-c modulates skeletal muscle function by directly binding and activating CK2. iScience 2024; 27:111212. [PMID: 39559755 PMCID: PMC11570452 DOI: 10.1016/j.isci.2024.111212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/16/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
MOTS-c is a mitochondrial microprotein that improves metabolism. Here, we demonstrate CK2 is a direct and functional target of MOTS-c. MOTS-c directly binds to CK2 and activates it in cell-free systems. MOTS-c administration to mice prevented skeletal muscle atrophy and enhanced muscle glucose uptake, which were blunted by suppressing CK2 activity. Interestingly, the effects of MOTS-c are tissue-specific. Systemically administered MOTS-c binds to CK2 in fat and muscle, yet stimulates CK2 activity in muscle while suppressing it in fat by differentially modifying CK2-interacting proteins. Notably, a naturally occurring MOTS-c variant, K14Q MOTS-c, has reduced binding to CK2 and does not activate it or elicit its effects. Male K14Q MOTS-c carriers exhibited a higher risk of sarcopenia and type 2 diabetes (T2D) in an age- and physical-activity-dependent manner, whereas females had an age-specific reduced risk of T2D. Altogether, these findings provide evidence that CK2 is required for MOTS-c effects.
Collapse
Affiliation(s)
- Hiroshi Kumagai
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Su-Jeong Kim
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Brendan Miller
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hirofumi Zempo
- Department of Administrative Nutrition, Faculty of Health and Nutrition, Tokyo Seiei College, Tokyo, Japan
| | - Kumpei Tanisawa
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | | | - Shin Hyung Lee
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Junxiang Wan
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Naphada Leelaprachakul
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Michi Emma Kumagai
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ricardo Ramirez
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hemal H. Mehta
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kevin Cao
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Tae Jung Oh
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - James A. Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jihui Sha
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Shohei Dobashi
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Eri Miyamoto-Mikami
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Mizuki Takaragawa
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Mizuho Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Tsudanuma Central General Hospital, Chiba, Japan
| | - Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Ryoko Kawakami
- Physical Fitness Research Institute, Meiji Yasuda Life Foundation of Health and Welfare, Tokyo, Japan
| | - Suguru Torii
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | - Taishi Midorikawa
- College of Health and Welfare, J.F. Oberlin University, Tokyo, Japan
| | - Koichiro Oka
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Chiharu Iwasaka
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Yosuke Yamada
- Sports and Health Sciences, Graduate School of Biomedical Engineering, Tohoku University, Miyagi, Japan
- Medicine and Science in Sports and Exercise, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Yasuki Higaki
- Laboratory of Exercise Physiology, Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Keitaro Tanaka
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Kelvin Yen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
41
|
Asil H, Demiryürek AT, Düzen IV, Büyükcelebi O, Saracaloglu A, Demirkiran C, Demiryürek Ş. Effects of empagliflozin and dapagliflozin on serum humanin, MOTS-c levels, nitrosative stress, and ferroptosis parameters in diabetic patients with heart failure. Eur J Pharmacol 2024; 982:176934. [PMID: 39182552 DOI: 10.1016/j.ejphar.2024.176934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors produce cardioprotective effects on heart failure (HF), even in the absence of diabetes. However, the underlying mechanisms of this cardioprotective effect remain unexplored. The purpose of this study was to examine the effects of SGLT2 inhibitors on serum MOTS-c, humanin levels, nitrosative stress, and ferroptosis parameters in diabetic patients with HF with reduced ejection fraction (HFrEF). A total of 74 adult diabetic patients with HFrEF and 37 healthy controls were included in this prospective study. Half of the patients were using SGLT2 inhibitors (empagliflozin or dapagliflozin) for at least two months. Serum nitric oxide and 3-nitrotyrosine levels were markedly higher in diabetic patients with HFrEF than the control (P < 0.001), but these elevations were inhibited with SGLT2 inhibitors. Although SGLT2 inhibitors had no marked effect on humanin levels, they significantly augmented MOTS-c levels when compared to the control. SGLT2 inhibitors augmented GPX4 but inhibited ACSL4 levels when compared to diabetic patients with HF. However, TFRC levels were increased in the patient group (P < 0.001 for all) but not modified with SGLT2 inhibitors. Our results suggest that increased nitrosative stress is significantly depressed by SGLT2 inhibitors. This study was the first to show that SGLT2 inhibitors can stimulate MOTS-c, but not humanin, in diabetic patients with HFrEF. SGLT2 inhibitors reduced ferroptosis through elevation of GPX4 and suppression of ACSL4 levels. Our data suggest that SGLT2 inhibitors could produce cardioprotective effects through relieving ferroptosis, inhibiting nitosative stress, and stimulating mitochondrial MOTS-c release.
Collapse
Affiliation(s)
- Hatice Asil
- Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | | | - Irfan Veysel Düzen
- Department of Cardiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Osman Büyükcelebi
- Department of Cardiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Ahmet Saracaloglu
- Department of Medical Pharmacology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Cahit Demirkiran
- Department of Medical Pharmacology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Şeniz Demiryürek
- Department of Physiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
42
|
Rice MC, Imun M, Jung SW, Park CY, Kim JS, Lai RW, Barr CR, Son JM, Tor K, Kim E, Lu RJ, Cohen I, Benayoun BA, Lee C. The Human Mitochondrial Genome Encodes for an Interferon-Responsive Host Defense Peptide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.02.530691. [PMID: 39553971 PMCID: PMC11565950 DOI: 10.1101/2023.03.02.530691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The mitochondrial DNA (mtDNA) can trigger immune responses and directly entrap pathogens, but it is not known to encode for active immune factors. The immune system is traditionally thought to be exclusively nuclear-encoded. Here, we report the identification of a mitochondrial-encoded host defense peptide (HDP) that presumably derives from the primordial proto-mitochondrial bacteria. We demonstrate that MOTS-c (mitochondrial open reading frame from the twelve S rRNA type-c) is a mitochondrial-encoded amphipathic and cationic peptide with direct antibacterial and immunomodulatory functions, consistent with the peptide chemistry and functions of known HDPs. MOTS-c targeted E. coli and methicillin-resistant S. aureus (MRSA), in part, by targeting their membranes using its hydrophobic and cationic domains. In monocytes, IFNγ, LPS, and differentiation signals each induced the expression of endogenous MOTS-c. Notably, MOTS-c translocated to the nucleus to regulate gene expression during monocyte differentiation and programmed them into macrophages with unique transcriptomic signatures related to antigen presentation and IFN signaling. MOTS-c-programmed macrophages exhibited enhanced bacterial clearance and shifted metabolism. Our findings support MOTS-c as a first-in-class mitochondrial-encoded HDP and indicates that our immune system is not only encoded by the nuclear genome, but also by the co-evolved mitochondrial genome.
Collapse
|
43
|
Yin Y, Li Y, Ma B, Ren C, Zhao S, Li J, Gong Y, Yang H, Li J. Mitochondrial-Derived Peptide MOTS-c Suppresses Ovarian Cancer Progression by Attenuating USP7-Mediated LARS1 Deubiquitination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405620. [PMID: 39321430 DOI: 10.1002/advs.202405620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Mitochondrial-nuclear communication plays a vital role in maintaining cellular homeostasis. MOTS-c, a short peptide derived from the 12S rRNA of mitochondrial DNA, has been suggested as a retrograde mitochondrial signal. Although recent clinical studies have suggested a possible link between MOTS-c and human cancer, the role of MOTS-c in tumorigenesis has yet to be investigated. Here, MOTS-c levels are found to be reduced in both serum and tumor tissues from ovarian cancer (OC) patients, which are associated with poor patients' prognosis. Exogenous MOTS-c inhibits the proliferation, migration and invasion of OC cells, and induces cell cycle arrest and apoptosis. Mechanistically, MOTS-c interacts with LARS1 and promotes its ubiquitination and proteasomal degradation. In addition, USP7 was identified as a deubiquitinase of LARS1, and MOTS-c can attenuates USP7-mediated LARS1 deubiquitination by competing with USP7 for binding to LARS1. Besides, LARS1 was found to be increased and play an important oncogenic function in OC. More importantly, MOTS-c displays a marked anti-tumor effect on OC growth without systemic toxicity in vivo. In conclusion, this study reveals a crucial role of MOTS-c in OC and provides a possibility for MOTS-c as a therapeutic target for the treatment of this manlignacy.
Collapse
Affiliation(s)
- Yadong Yin
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yujie Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Boyi Ma
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Chenlu Ren
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shuhua Zhao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jia Li
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yun Gong
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jibin Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, 710032, China
| |
Collapse
|
44
|
Kim S. The Relationship Between MOTS-c K14Q Polymorphism and Sarcopenia, Blood Lipids, and Mental Health in Older Korean Adults. Biomedicines 2024; 12:2384. [PMID: 39457696 PMCID: PMC11504729 DOI: 10.3390/biomedicines12102384] [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: 09/23/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/objectives: An East Asian-specific 1382A>C polymorphism in the mitochondrial open reading frame of the 12S rRNA type-c results in an amino acid substitution from Lys (K) to Gln (Q) at the 14th amino acid residue. This study investigated the association between m.1382A>C polymorphism and sarcopenia, blood lipids, and mental health in older Korean adults. Methods: The study included 683 community-dwelling Korean adults (345 men and 338 women) aged 65 years and older. The m.1382A>C polymorphism was genotyped with a 7500 real-time PCR system. Handgrip strength (HGS) was measured, and appendicular skeletal muscle (ASM) mass was calculated. Demographics, blood lipids, falling risk, nutritional intake, cognition function, and depression were additionally measured. Results: Men carrying the C allele had significantly higher ASM (21.6 ± 3.0 vs. 19.5 ± 2.2 kg, p = 0.018), ASM/height2 (7.76 ± 0.76 vs. 7.14 ± 0.62 kg/m2, p = 0.012), lean mass (53.3 ± 6.2 vs. 46.5 ± 4.0 kg, p < 0.001), left HGS (33.3 ± 5.0 vs. 28.9 ± 4.0 kg, p = 0.010), and right HGS (35.6 ± 5.3 vs. 30.9 ± 4.3 kg, p = 0.009) than men carrying the A allele. The genotype differences in ASM (p = 0.017), ASM/height2 (p = 0.011), lean mass (p < 0.001), left HGS (p = 0.010), and right HGS (p = 0.009) remained significant even after adjusting for all measured covariates. By contrast, no significant differences in other measured parameters were found between women carrying the A and C alleles. Conclusions: Our study findings indicate that the m.1382A>C polymorphism may be used as a genetic biomarker of age-related sarcopenia in older Korean men.
Collapse
Affiliation(s)
- Shinuk Kim
- Gyedang College of General Education, Sangmyung University, Cheonan 31066, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
45
|
Dashti M, Ali NM, Alsaleh H, John SE, Nizam R, Al-Mulla F, Thanaraj TA. Mitochondrial haplogroup R offers protection against obesity in Kuwaiti and Qatari populations. Front Endocrinol (Lausanne) 2024; 15:1449374. [PMID: 39464187 PMCID: PMC11502345 DOI: 10.3389/fendo.2024.1449374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024] Open
Abstract
Background The Kuwaiti and Qatari populations have a high prevalence of obesity, a major risk factor for various metabolic disorders. Previous studies have independently explored mitochondrial DNA (mtDNA) variations and their association with obesity in these populations. This study aims to investigate the role of mtDNA haplogroups and variants in obesity risk among these Gulf populations. Methods Whole exome sequencing data from 1,112 participants (348 Kuwaitis and 764 Qataris) were analyzed for mtDNA variants. Participants were classified as obese or non-obese based on body mass index (BMI). Association analyses were performed to examine the relationship between mtDNA haplogroups and obesity, adjusting for covariates such as age and sex. Results Haplogroup R was found to be protective against obesity, with an odds ratio (OR) of 0.69 (p = 0.045). This association remained significant after adjusting for age and sex (OR = 0.694; 95% CI: 0.482-0.997; p = 0.048). Several mtDNA variants, particularly those involved in mitochondrial energy metabolism, showed nominal associations with obesity, but these did not remain significant after correcting for multiple testing. Conclusion Haplogroup R consistently demonstrates a protective association against obesity in both Kuwaiti and Qatari populations, highlighting its potential as a biomarker for obesity risk in the Gulf region. However, further research with larger sample sizes is needed to validate these findings and clarify the role of mtDNA variants in obesity.
Collapse
Affiliation(s)
- Mohammed Dashti
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Naser M. Ali
- Department of Medical Laboratories, Ahmadi Hospital, Kuwait Oil Company (KOC), Ahmadi, Kuwait
| | - Hussain Alsaleh
- Saad Al-Abdullah Academy for Security Sciences, Ministry of Interior, Shuwaikh, Kuwait
| | - Sumi Elsa John
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | |
Collapse
|
46
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
47
|
Mcleod JC, Lim C, Stokes T, Sharif JA, Zeynalli V, Wiens L, D’Souza AC, Colenso-Semple L, McKendry J, Morton RW, Mitchell CJ, Oikawa SY, Wahlestedt C, Paul Chapple J, McGlory C, Timmons JA, Phillips SM. Network-based modelling reveals cell-type enriched patterns of non-coding RNA regulation during human skeletal muscle remodelling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.11.606848. [PMID: 39416175 PMCID: PMC11482748 DOI: 10.1101/2024.08.11.606848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
A majority of human genes produce non-protein-coding RNA (ncRNA), and some have roles in development and disease. Neither ncRNA nor human skeletal muscle is ideally studied using short-read sequencing, so we used a customised RNA pipeline and network modelling to study cell-type specific ncRNA responses during muscle growth at scale. We completed five human resistance-training studies (n=144 subjects), identifying 61% who successfully accrued muscle-mass. We produced 288 transcriptome-wide profiles and found 110 ncRNAs linked to muscle growth in vivo, while a transcriptome-driven network model demonstrated interactions via a number of discrete functional pathways and single-cell types. This analysis included established hypertrophy-related ncRNAs, including CYTOR - which was leukocyte-associated (FDR = 4.9 ×10-7). Novel hypertrophy-linked ncRNAs included PPP1CB-DT (myofibril assembly genes, FDR = 8.15 × 10-8), and EEF1A1P24 and TMSB4XP8 (vascular remodelling and angiogenesis genes, FDR = 2.77 × 10-5). We also discovered that hypertrophy lncRNA MYREM shows a specific myonuclear expression pattern in vivo. Our multi-layered analyses established that single-cell-associated ncRNA are identifiable from bulk muscle transcriptomic data and that hypertrophy-linked ncRNA genes mediate their association with muscle growth via multiple cell types and a set of interacting pathways.
Collapse
Affiliation(s)
- Jonathan C. Mcleod
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Changhyun Lim
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
- Population Health Sciences Institute, Faculty of Medicial Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Tanner Stokes
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Jalil-Ahmad Sharif
- Faculty of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Vagif Zeynalli
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Lucas Wiens
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Alysha C D’Souza
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - James McKendry
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
- Faculty of Land and Food Systems, Food, Nutrition & Health, University of British Columbia, BC, Canada
| | - Robert W. Morton
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - Sara Y. Oikawa
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - J Paul Chapple
- Faculty of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Chris McGlory
- School of Kinesiology and Health Studies, Queens University, Kingston, ON, Canada
| | - James A. Timmons
- Faculty of Medicine and Dentistry, Queen Mary University London, London, UK
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stuart M. Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
48
|
Tzani I, Castro-Rivadeneyra M, Kelly P, Strasser L, Zhang L, Clynes M, Karger BL, Barron N, Bones J, Clarke C. Detection of host cell microprotein impurities in antibody drug products. Nat Commun 2024; 15:8605. [PMID: 39366928 PMCID: PMC11452709 DOI: 10.1038/s41467-024-51870-0] [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: 04/12/2023] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
Chinese hamster ovary (CHO) cells are used to produce almost 90% of therapeutic monoclonal antibodies (mAbs) and antibody fusion proteins (Fc-fusion). The annotation of non-canonical translation events in these cellular factories remains incomplete, limiting our ability to study CHO cell biology and detect host cell protein (HCP) impurities in the final antibody drug product. We utilised ribosome footprint profiling (Ribo-seq) to identify novel open reading frames (ORFs) including N-terminal extensions and thousands of short ORFs (sORFs) predicted to encode microproteins. Mass spectrometry-based HCP analysis of eight commercial antibody drug products (7 mAbs and 1 Fc-fusion protein) using the extended protein sequence database revealed the presence of microprotein impurities. We present evidence that microprotein abundance varies with growth phase and can be affected by the cell culture environment. In addition, our work provides a vital resource to facilitate future studies of non-canonical translation and the regulation of protein synthesis in CHO cell lines.
Collapse
Affiliation(s)
- Ioanna Tzani
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
| | - Marina Castro-Rivadeneyra
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin, Ireland
| | - Paul Kelly
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
| | - Lisa Strasser
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
| | - Lin Zhang
- Bioprocess R&D, Pfizer Inc. Andover, Massachusetts, USA
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Barry L Karger
- Barnett Institute, Northeastern University, 360 Huntington Ave, Boston, MA, USA
| | - Niall Barron
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin, Ireland
| | - Jonathan Bones
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin, Ireland
| | - Colin Clarke
- National Institute for Bioprocessing Research and Training, Fosters Avenue, Blackrock, Co, Dublin, Ireland.
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
49
|
Li Y, Li Z, Ren Y, Lei Y, Yang S, Shi Y, Peng H, Yang W, Guo T, Yu Y, Xiong Y. Mitochondrial-derived peptides in cardiovascular disease: Novel insights and therapeutic opportunities. J Adv Res 2024; 64:99-115. [PMID: 38008175 PMCID: PMC11464474 DOI: 10.1016/j.jare.2023.11.018] [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: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Mitochondria-derived peptides (MDPs) represent a recently discovered family of peptides encoded by short open reading frames (ORFs) found within mitochondrial genes. This group includes notable members including humanin (HN), mitochondrial ORF of the 12S rDNA type-c (MOTS-c), and small humanin-like peptides 1-6 (SHLP1-6). MDPs assume pivotal roles in the regulation of diverse cellular processes, encompassing apoptosis, inflammation, and oxidative stress, which are all essential for sustaining cellular viability and normal physiological functions. Their emerging significance extends beyond this, prompting a deeper exploration into their multifaceted roles and potential applications. AIM OF REVIEW This review aims to comprehensively explore the biogenesis, various types, and diverse functions of MDPs. It seeks to elucidate the central roles and underlying mechanisms by which MDPs participate in the onset and development of cardiovascular diseases (CVDs), bridging the connections between cell apoptosis, inflammation, and oxidative stress. Furthermore, the review highlights recent advancements in clinical research related to the utilization of MDPs in CVD diagnosis and treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW MDPs levels are diminished with aging and in the presence of CVDs, rendering them potential new indicators for the diagnosis of CVDs. Also, MDPs may represent a novel and promising strategy for CVD therapy. In this review, we delve into the biogenesis, various types, and diverse functions of MDPs. We aim to shed light on the pivotal roles and the underlying mechanisms through which MDPs contribute to the onset and advancement of CVDs connecting cell apoptosis, inflammation, and oxidative stress. We also provide insights into the current advancements in clinical research related to the utilization of MDPs in the treatment of CVDs. This review may provide valuable information with MDPs for CVD diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Ying Lei
- School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Silong Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuqi Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Han Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Weijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Tiantian Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
50
|
Mcleod J, Lim C, Stokes T, Sharif JA, Zeynalli V, Wiens L, D’Souza A, Colenso-Semple L, McKendry J, Morton R, Mitchell C, Oikawa S, Wahlestedt C, Chapple J, McGlory C, Timmons J, Phillips S. Network-based modelling reveals cell-type enriched patterns of non-coding RNA regulation during human skeletal muscle remodelling. NAR MOLECULAR MEDICINE 2024; 1:ugae016. [PMID: 39669123 PMCID: PMC11632610 DOI: 10.1093/narmme/ugae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 12/14/2024]
Abstract
A majority of human genes produce non-protein-coding RNA (ncRNA), and some have roles in development and disease. Neither ncRNA nor human skeletal muscle is ideally studied using short-read sequencing, so we used a customized RNA pipeline and network modelling to study cell-type specific ncRNA responses during muscle growth at scale. We completed five human resistance-training studies (n = 144 subjects), identifying 61% who successfully accrued muscle-mass. We produced 288 transcriptome-wide profiles and found 110 ncRNAs linked to muscle growth in vivo, while a transcriptome-driven network model demonstrated interactions via a number of discrete functional pathways and single-cell types. This analysis included established hypertrophy-related ncRNAs, including CYTOR-which was leukocyte-associated (false discovery rate [FDR] = 4.9 × 10-7). Novel hypertrophy-linked ncRNAs included PPP1CB-DT (myofibril assembly genes, FDR = 8.15 × 10-8), and EEF1A1P24 and TMSB4XP8 (vascular remodelling and angiogenesis genes, FDR = 2.77 × 10-5). We also discovered that hypertrophy lncRNA MYREM shows a specific myonuclear expression pattern in vivo. Our multi-layered analyses established that single-cell-associated ncRNA are identifiable from bulk muscle transcriptomic data and that hypertrophy-linked ncRNA genes mediate their association with muscle growth via multiple cell types and a set of interacting pathways.
Collapse
Affiliation(s)
- Jonathan C Mcleod
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Changhyun Lim
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Population Health Sciences Institute, Faculty of Medicial Sciences, Newcastle University, Newcastle upon Tyne, NE2 4AX, UK
| | - Tanner Stokes
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Jalil-Ahmad Sharif
- Faculty of Medicine and Dentistry, Queen Mary University London, London, E1 4NS, UK
| | - Vagif Zeynalli
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Lucas Wiens
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Alysha C D’Souza
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | | | - James McKendry
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Faculty of Land and Food Systems, Food, Nutrition & Health, University of British Columbia, BC, V6T 1Z4, Canada
| | - Robert W Morton
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Cameron J Mitchell
- School of Kinesiology, University of British Columbia, BC, V6T 1Z1, Canada
| | - Sara Y Oikawa
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Claes Wahlestedt
- University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - J Paul Chapple
- Faculty of Medicine and Dentistry, Queen Mary University London, London, E1 4NS, UK
| | - Chris McGlory
- School of Kinesiology and Health Studies, Queens University, Kingston, ON, K7L 3N6, Canada
| | - James A Timmons
- Faculty of Medicine and Dentistry, Queen Mary University London, London, E1 4NS, UK
- University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| |
Collapse
|