1
|
Bian C, Wei M, Luo X, Sun J, Ji H. Molecular cloning, characterization and expression analysis of PINK1 (Phosphatase and tensin homolog-induced putative kinase 1) and Parkin (parkin RBR E3 ubiquitin protein ligase) in grass carp (Ctenopharyngodon idellus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 166:105349. [PMID: 40044098 DOI: 10.1016/j.dci.2025.105349] [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: 02/18/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
Phosphatase and tensin homolog-induced putative kinase 1 (PINK1) and parkin RBR E3 ubiquitin protein ligase (Parkin) emerged as mediators of mitophagy and regulators of the immune response in mammals. However, their gene characterizations and roles remain poorly understood in fish. Herein, we identified and characterized pink1 and parkin genes and studied their involvement in immune responses to lipopolysaccharide (LPS) in Ctenopharyngodon idellus kidney (CIK) cells. Bioinformatic analysis found that PINK1 and Parkin were relatively conservative during evolution. In CIK cells, LPS significantly increased the mRNA expression of the transcription factor nf-κb and its downstream proinflammatory cytokines, such as tnfα, il-6 and il-1β, along with the activation of PINK1/Parkin-mediated mitophagy (P < 0.05). Furthermore, inhibition of mitophagy aggravated LPS-induced inflammation in CIK cells. Overall, our findings suggest that PINK1/Parkin-mediated mitophagy may play a protective role in LPS-induced inflammation in fish.
Collapse
Affiliation(s)
- Chenchen Bian
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Mingkui Wei
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Xiaolong Luo
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China.
| |
Collapse
|
2
|
Sun H, Wang Y, Deng G, Gao R, Zhang M, Huang L, He W, Zhang Z, Yu D, Chen P, Lu F, Liu S. Cortex Dictamni-induced hepatotoxicity by enhanced oxidative phosphorylation: Insights from integrative transcriptomics, proteomics, and metabolomics analyses. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156511. [PMID: 39954621 DOI: 10.1016/j.phymed.2025.156511] [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: 11/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Cortex Dictamni (CD) is a traditional Chinese medicine that is commonly used to treat various skin diseases. Recently, clinical reports have highlighted its potential to induce severe hepatotoxicity. However, the underlying mechanisms of toxicity remain inadequately explored. PURPOSE The aim of this study was to elucidate the intrinsic mechanisms of CD-induced hepatotoxicity. STUDY DESIGN Hepatotoxicity was assessed in SD rats, and human primary hepatocytes (HPHs) and differentiated HepaRG (dHepaRG) cells were used for in vitro testing. METHODS The major components of CD were determined using ultra-performance liquid chromatography (UPLC). Rats were randomly divided into control, CD-high (CD-H), CD-middle (CD-M), CD-low (CD-L), and isoniazid (INH) groups and administered oral gavage for four weeks. Serum biochemical indices, histopathological changes, apoptotic markers, and liver function were evaluated to assess hepatotoxicity. A comprehensive analysis of rat liver samples was performed using transcriptomic, proteomic, and metabolomic approaches to identify key pathways involved in CD-induced hepatotoxicity. In vitro toxicity validation of CD was performed using HPHs and dHepaRG cells. The key pathway was validated in vivo and in vitro. RESULTS CD primarily contained obacunone, fraxinellone, and dictamine. Administration of CD-H (9 times the maximum daily clinical dose in adults) and CD-M (3 times the maximum daily clinical dose in adults) for 4 weeks induced varying degrees of hepatotoxicity in rats. The CD-H group presented increased absolute and relative liver weights, reduced alanine aminotransferase (ALT) and bile acid transporter levels, and increased albumin (ALB) and cytochrome P450 (CYP) 3A4 levels, indicating significant hepatotoxicity in rats. Integrated multiomics analysis revealed that NADH dehydrogenase (ubiquinone) Fe-S protein 2 (Ndufs2) is a critical regulator of CD-induced hepatotoxicity involving oxidative phosphorylation (OXPHOS). CD inhibited the viability of HPHs and dHepaRG cells, demonstrating its significant cytotoxicity. Mechanistic validation revealed that CD upregulated Ndufs2, reactive oxygen species (ROS) and mitochondrial respiratory chain complex (MRCC) I, leading to nuclear factor erythroid 2-related factor 2 (Nrf2) pathway activation, apoptosis, mitochondrial dysfunction, and hepatotoxicity. CONCLUSION In summary, our study presents a comprehensive picture of the toxicity of CD in terms of dose and sex and reveals, for the first time, the central role of Ndufs2-regulated OXPHOS in CD-induced hepatotoxicity.
Collapse
Affiliation(s)
- Huijuan Sun
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Geyu Deng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Rui Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Mengmeng Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Lin Huang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Wenjie He
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Zhendong Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Pingping Chen
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China.
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China.
| |
Collapse
|
3
|
Mejía-Guzmán JE, Belmont-Hernández RA, Chávez-Tapia NC, Uribe M, Nuño-Lámbarri N. Metabolic-Dysfunction-Associated Steatotic Liver Disease: Molecular Mechanisms, Clinical Implications, and Emerging Therapeutic Strategies. Int J Mol Sci 2025; 26:2959. [PMID: 40243565 PMCID: PMC11988898 DOI: 10.3390/ijms26072959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is a highly prevalent metabolic disorder characterized by hepatic steatosis in conjunction with at least one cardiometabolic risk factor, such as obesity, type 2 diabetes, hypertension, or dyslipidemia. As global rates of obesity and metabolic syndrome continue to rise, MASLD is becoming a major public health concern, with projections indicating a substantial increase in prevalence over the coming decades. The disease spectrum ranges from simple steatosis to metabolic-dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis, and hepatocellular carcinoma, contributing to significant morbidity and mortality worldwide. This review delves into the molecular mechanisms driving MASLD pathogenesis, including dysregulation of lipid metabolism, chronic inflammation, oxidative stress, mitochondrial dysfunction, and gut microbiota alterations. Recent advances in research have highlighted the role of genetic and epigenetic factors in disease progression, as well as novel therapeutic targets such as peroxisome proliferator-activated receptors (PPARs), fibroblast growth factors, and thyroid hormone receptor beta agonists. Given the multifaceted nature of MASLD, a multidisciplinary approach integrating early diagnosis, molecular insights, lifestyle interventions, and personalized therapies is critical. This review underscores the urgent need for continued research into innovative treatment strategies and precision medicine approaches to halt MASLD progression and improve patient outcomes.
Collapse
Affiliation(s)
- Jeysson E. Mejía-Guzmán
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
| | - Ramón A. Belmont-Hernández
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Postgraduate Program in Experimental Biology, División de Ciencias Básicas y de la Salud (DCBS), Universidad Autonoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Norberto C. Chávez-Tapia
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
| | - Misael Uribe
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
| | - Natalia Nuño-Lámbarri
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Surgery Department, Faculty of Medicine, The National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| |
Collapse
|
4
|
Lin X, Gao H, Xin M, Huang J, Li X, Zhou Y, Lv K, Huang X, Wang J, Zhou Y, Cui D, Fang C, Wu L, Shi X, Ma Z, Qian Y, Tong H, Dai J, Jin J, Huang J. α-Actinin-1 deficiency in megakaryocytes causes low platelet count, platelet dysfunction, and mitochondrial impairment. Blood Adv 2025; 9:1185-1201. [PMID: 39813624 PMCID: PMC11925533 DOI: 10.1182/bloodadvances.2024014805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/18/2025] Open
Abstract
ABSTRACT Cytoskeletal remodeling and mitochondrial bioenergetics play important roles in thrombocytopoiesis and platelet function. Recently, α-actinin-1 mutations have been reported in patients with congenital macrothrombocytopenia. However, the role and underlying mechanism of α-actinin-1 in thrombocytopoiesis and platelet function remain elusive. Using megakaryocyte (MK)-specific α-actinin-1 knockout (KO; PF4-Actn1-/-) mice, we demonstrated that PF4-Actn1-/- mice exhibited reduced platelet counts. The decreased platelet number in PF4-Actn1-/- mice was due to defects in thrombocytopoiesis. Hematoxylin and eosin staining and flow cytometry revealed a decrease in the number of MKs in the bone marrow of PF4-Actn1-/- mice. The absence of α-actinin-1 increased the proportion of 2 N-4 N MKs and decreased the proportion of 8 N-32 N MKs. Colony-forming unit-MK colony formation, the ratio of proplatelet formation-bearing MKs, and MK migration in response to stromal cell-derived factor-1 signaling were inhibited in PF4-Actn1-/- mice. Platelet spreading, clot retraction, aggregation, integrin αIIbβ3 activation, and CD62P exposure in response to various agonists were decreased in PF4-Actn1-/- platelets. Notably, PF4-Actn1-/- platelets inhibited calcium mobilization, reactive oxygen species (ROS) generation, and actin polymerization in response to collagen and thrombin. Furthermore, the PF4-Actn1-/- mice exhibited impaired hemostasis and thrombosis. Mechanistically, proteomic analysis of low-ploidy (2-4 N) and high-ploidy (≥8 N) PF4-Actn1-/- MKs revealed that α-actinin-1 deletion reduced platelet activation and mitochondrial function. PF4-Actn1-/- platelets and Actn1 KO 293T cells exhibited reduced mitochondrial membrane potential, mitochondrial ROS generation, mitochondrial calcium mobilization, and mitochondrial bioenergetics. Overall, in this study, we report that mice with α-actinin-1 deficiency in MKs exhibit low platelet count and impaired platelet function, thrombosis, and mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Xiangjie Lin
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hanchen Gao
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Xin
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Huang
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Li
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yutong Zhou
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Huang
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghan Wang
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulan Zhou
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lanlan Wu
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaofeng Shi
- Department of Hematology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixin Ma
- Clinical Prenatal Diagnosis Center, Key Laboratory of Reproductive Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Qian
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyan Tong
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Dai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Jin
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, China
| | - Jiansong Huang
- Department of Hematology, Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
6
|
Li X, Li D, Zhang R. Single-Cell RNA sequencing reveals mitochondrial dysfunction in microtia chondrocytes. Sci Rep 2025; 15:1021. [PMID: 39762337 PMCID: PMC11704343 DOI: 10.1038/s41598-025-85169-x] [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: 10/08/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Microtia is a congenital malformation characterized by underdevelopment of the external ear. While chondrocyte dysfunction has been implicated in microtia, the specific cellular abnormalities remain poorly understood. This study aimed to investigate mitochondrial dysfunction in microtia chondrocytes using single-cell RNA sequencing. Cartilage samples were obtained from patients with unilateral, non-syndromic microtia and healthy controls. Single-cell RNA sequencing was performed using the 10 × Genomics platform. Bioinformatic analyses including cell type identification, trajectory analysis, and gene co-expression network analysis were conducted. Mitochondrial function was assessed through ROS levels, membrane potential, and transmission electron microscopy. Chondrocytes from microtia samples showed lower mitochondrial function scores compared to normal samples. Trajectory analysis revealed more disorganized differentiation patterns in microtia chondrocytes. Mitochondrial dysfunction in microtia chondrocytes was confirmed by increased ROS production, decreased membrane potential, and altered mitochondrial structure. Gene co-expression network analysis identified hub genes associated with mitochondrial function, including SDHA, SIRT1, and PGC1A, which showed reduced expression in microtia chondrocytes. This study provides evidence of mitochondrial dysfunction in microtia chondrocytes and identifies potential key genes involved in this process. These findings offer new insights into the pathogenesis of microtia and may guide future therapeutic strategies.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Datao Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruhong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Du Q, Ning N, Zhao X, Liu F, Zhang S, Xia Y, Li F, Yuan S, Xie X, Zhu M, Huang Z, Tang Z, Wang J, He R, Yang XP. Acylglycerol kinase inhibits macrophage anti-tumor activity via limiting mtDNA release and cGAS-STING-type I IFN response. Theranostics 2025; 15:1304-1319. [PMID: 39816692 PMCID: PMC11729555 DOI: 10.7150/thno.101298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025] Open
Abstract
Background: Tumor associated macrophages (TAMs) are critical components in regulating the immune statuses of the tumor microenvironments. Although TAM has been intensively studied, it is unclear how mitochondrial proteins such as AGK regulate the TAMs' function. Methods: We investigated the AGK function in TAMs using macrophage-specific Agk deficient mice with B16 and LLC syngeneic tumor models. Flow cytometry was used to evaluate the stemness and activation of CD8+ T cells. The enhanced release of mtDNA into the cytosol in the Agk-deficient BMDMs was measured by RT-PCR and immunofluorescence; the cGAS-STING-type I IFN pathway was evaluated by immunoblotting. Mitochondria functions were evaluated by electron microscope and seahorse equipment. Results: We have noted an increased expression of AGK in TAMs of multiple tumor types, which was negatively correlates with the tumor tissue immune scores. In the B16 and LLC tumor models, macrophage Agk-deficient mice have reduced tumor growth and enhanced populations of CD8+ Tpex. AGK-deficient macrophages have increased mitochondrial damage and mtDNA release into the cytosol, which leads to enhanced cGAS-STING-type I IFN activation. Blockade of the type I IFN signaling pathway with anti-IFNAR reversed the phenotype in Agk-deficient mice. Conclusions: Our findings define a critical role of AGK in maintaining the macrophage mitochondrial homeostasis that is associated with mtDNA release and following cGAS-STING activation and type I IFN pathway. Targeting AGK in TAMs may represent a novel strategy to enhance anti-tumoral activity.
Collapse
Affiliation(s)
- Qiuyang Du
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Second Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Na Ning
- Department of Pathology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xiujuan Zhao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifan Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan, China
| | - Si Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan, China
| | - Yuting Xia
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Fei Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijie Yuan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaorong Xie
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengdi Zhu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehan Huang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Tang
- Division of Trauma Surgery, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran He
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Second Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
| |
Collapse
|
8
|
Peng Y, Nie H, Kang K, Li X, Tao Y, Zhou Y. The deubiquitinating enzyme ATXN3 promotes hepatocellular carcinoma progression by stabilizing TAZ. Cancer Gene Ther 2025; 32:136-145. [PMID: 39672915 DOI: 10.1038/s41417-024-00869-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Hepatocellular carcinoma (HCC) was a primary cause of cancer-related morbidity and mortality in China. ATXN3 was a deubiquitinase (DUB) associated with spinocerebellar ataxia, widely expressed in the cytoplasm and nucleus of cells in the central nervous system and other tissues. The crucial role of the Hippo pathway in tumorigenesis has been established, among which TAZ served as one of the key molecules. However, the mechanisms underlying the deubiquitinase and TAZ in HCC remained largely unknown. In the present study, we explored that ATXN3 was overexpressed in HCC. ATXN3 promoted the proliferation, migration, invasion, and tumorigenic ability of HCC in vitro and in vivo. Besides, we explored that ATXN3 was positively associated with TAZ in HCC. ATXN3 could interact with, stabilize, and deubiquitylate TAZ in a deubiquitylase-dependent manner. Specifically, this interaction was primarily mediated by the C-terminal domain of TAZ and Josephin domain of ATXN3, thereby inhibiting the K48-linked ubiquitin chain on TAZ. Furthermore, we demonstrated that ATXN3 promoted the occurrence and development of HCC by regulating TAZ. Therefore, our study revealed the oncogenic function of ATXN3 and an interesting deubiquitination mechanism of ATXN3 and TAZ in HCC, providing new insights into the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Yuanhao Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Hui Nie
- Department of Pathology, School of Basic Medicine and Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kuo Kang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuanxuan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine and Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Liu M, Fu X, Yi Q, Xu E, Dong L. Impaired mitochondrial oxidative phosphorylation induces CD8 + T cell exhaustion. Biochem Biophys Res Commun 2024; 734:150738. [PMID: 39342799 DOI: 10.1016/j.bbrc.2024.150738] [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: 09/18/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
CD8+ T cells play a crucial role in anti-tumor immunity, but their function can be impaired by exhaustion induced by prolonged antigen stimulation. Mitochondrial dysfunction, a hallmark of the tumor microenvironment (TME), has been linked to various pathologies, but its specific role in CD8+ T cell exhaustion remains underexplored. Here, we established an in vitro model of CD8+ T cell exhaustion by co-culturing OVA-specific OT1 CD8+ T cells with OVA-expressing MC38 tumor cells. Next, we investigated the impact of mitochondrial dysfunction on exhaustion using pharmacological inhibitors targeting the electron transport chain. The role of the mitochondrial complex I component NDUFA10 was further examined through genetic knockout in CD8+ T cells using CRISPR-Cas9. Inhibition of the mitochondrial electron transport chain significantly accelerated CD8+ T cell exhaustion in vitro. Knockout of NDUFA10 in CD8+ T cells led to enhanced tumor growth and increased exhaustion of tumor-infiltrating CD8+ T cells in a Rag1-/- tumor-bearing transfer model. This study highlights the critical role of mitochondrial function in regulating CD8+ T cell exhaustion and anti-tumor activity, providing new insights into the metabolic underpinnings of immune dysfunction in cancer.
Collapse
Affiliation(s)
- Min Liu
- Department of Otolaryngology, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, 200053, China
| | - Xinyue Fu
- Department of Otolaryngology, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, 200053, China
| | - Qinghe Yi
- Department of Otolaryngology, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, 200053, China
| | - Enhong Xu
- Department of Otolaryngology, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, 200053, China
| | - Longbao Dong
- Department of Otolaryngology, Naval Medical Center of People's Liberation Army of China (PLA), Shanghai, 200053, China.
| |
Collapse
|
10
|
Zhang Z, Liu Y, Wu H, Yuan Y, Liu Z, Sulaiman M, Yuan S, Yang M. Netupitant Inhibits the Proliferation of Breast Cancer Cells by Targeting AGK. Cancers (Basel) 2024; 16:3807. [PMID: 39594764 PMCID: PMC11592365 DOI: 10.3390/cancers16223807] [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: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Currently, there is a significant lack of effective pharmacological agents for the treatment of breast cancer. Acylglycerol Kinase (AGK), a lipid kinase, has been found to be aberrantly expressed in breast cancer and is closely associated with tumor proliferation, migration, and invasion. However, no clinical anti-tumor drugs specifically targeting this kinase have been developed. Methods: siRNA was utilized to knock down the AGK gene; CCK8 and colony formation assays were employed to evaluate the in vitro proliferative capacity of tumor cells. Molecular dynamics simulations and BIL assays were conducted to analyze drug binding affinity. Annexin V/PI staining was used to assess apoptotic phenomena; subcutaneous xenograft tumor experiments in nude mice were performed to confirm the in vivo anti-tumor efficacy of the drug. Results: Netupitant exhibited stable binding affinity for AGK and interacted with amino acids within the ATP-binding region of the enzyme. The IC50 values for the SK-BR-3 and MDA-MB-231 cell lines were determined as 16.15 ± 4.25 µmol/L and 24.02 ± 4.19 µmol/L, respectively; at a concentration of 2.5 µmol/L, Netupitant effectively inhibited clonogenic capacity in breast cancer cells; furthermore, treatment with 10 µmol/L significantly induced apoptosis in these cells. Doses of 50 mg/kg and 100 mg/kg Netupitant markedly suppressed growth rates of subcutaneous xenograft tumors in nude mice while also promoting apoptotic processes. Both in vivo and in vitro studies indicated that Netupitant could inhibit the activation of the PI3K/AKT/mTOR signaling pathway. Conclusions: By targeting AGK, Netupitant inhibits its kinase activity, which leads to reduced phosphorylation levels of PTEN, thereby suppressing the activation of the PI3K/AKT/mTOR signaling pathway and ultimately resulting in apoptosis in breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shengtao Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (Y.L.); (H.W.); (M.S.)
| | - Mei Yang
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (Y.L.); (H.W.); (M.S.)
| |
Collapse
|
11
|
Pi D, Liang Z, Pan M, Zhen J, Zheng C, Pan J, Fan W, Song Q, Yang Q, Zhang Y. Atractylodes lancea Rhizome Polysaccharide Alleviates MCD Diet-Induced NASH by Inhibiting the p53/mTOR Pathway. Int J Mol Sci 2024; 25:11112. [PMID: 39456893 PMCID: PMC11508089 DOI: 10.3390/ijms252011112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a form of chronic liver disease that is characterized by liver inflammation and steatosis, with possible progression to fibrosis. Currently, no drugs have been approved for the treatment of NASH. In this study, we isolated a polysaccharide from Atractylodes lancea rhizome (AP) and established a methionine- and choline-deficient (MCD) diet -induced NASH mouse model to investigate the preventive effect and potential mechanism of AP on NASH. The results showed that AP effectively reduced liver lipid accumulation and inflammation and reduced autophagy and ferroptosis in hepatocytes, thereby preventing the development of NASH. These findings suggest that AP may be a promising natural candidate for the treatment of NASH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qinhe Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yupei Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
12
|
Liu R, Ding Y, Jing F, Chen Z, Su C, Pan L. Effects of dietary glycerol monolaurate on growth and digestive performance, lipid metabolism, immune defense and gut microbiota of shrimp (Penaeus vannamei). FISH & SHELLFISH IMMUNOLOGY 2024; 151:109666. [PMID: 38838839 DOI: 10.1016/j.fsi.2024.109666] [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: 03/09/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
The advancement of the Penaeus vannamei industry in a sustainable manner necessitates the creation of eco-friendly and exceptionally effective feed additives. To achieve this, 720 similarly-sized juvenile shrimp (0.88 ± 0.02 g) were randomly divided into four groups in this study, with each group consisting of three replicates, each tank (400 L) containing 60 shrimp. Four experimental diets were formulated by adding 0, 500, 1000, and 1500 mg kg-1 glycerol monolaurate (GML) to the basal diet, and the feeding trial lasted for 42 days. Subsequently, a 72-h White Spot Syndrome Virus (WSSV) challenge test was conducted. Polynomial orthogonal contrasts analysis revealed that with the increase in the concentration of GML, those indicators related to growth, metabolism and immunity, exhibit linear or quadratic correlations (P < 0.05). The results indicate that the GML groups exhibited a significant improvement in the shrimp weight gain rate, specific growth rate, and a reduction in the feed conversion ratio (P < 0.05). Furthermore, the GML groups promoted the lipase activity and reduced lipid content of the shrimp, augmented the expression of triglyceride and fatty acid decomposition-related genes and lowered the levels of plasma triglycerides (P < 0.05). GML can also enhanced the humoral immunity of the shrimp by activating the Toll-like receptor and Immune deficiency immune pathways, improved the phagocytic capacity and antibacterial ability of shrimp hemocytes. The challenge test revealed that GML significantly reduced the mortality of the shrimp compared to control group. The 16S rRNA sequencing indicates that the GML group can increases the abundance of beneficial bacteria. However, 1500 mg kg-1 GML adversely affected the stability of the intestinal microbiota, significantly upregulating intestinal antimicrobial peptide-related genes and tumor necrosis factor-alpha levels (P < 0.05). In summary, 1000 mg kg-1 GML was proven to enhance the growth performance, lipid absorption and metabolism, humoral immune response, and gut microbiota condition of P. vannamei, with no negative physiological effects.
Collapse
Affiliation(s)
- Renzhi Liu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yanjun Ding
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Futao Jing
- Shandong Fisheries Development and Resources Conservation Center, Jinan, 250013, China
| | - Zhifei Chen
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Chen Su
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
13
|
Yagan M, Najam S, Hu R, Wang Y, Dadi P, Xu Y, Simmons AJ, Stein R, Adams CM, Jacobson DA, Lau K, Liu Q, Gu G. Atf4 protects islet β-cell identity and function under acute glucose-induced stress but promotes β-cell failure in the presence of free fatty acid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601249. [PMID: 39005465 PMCID: PMC11244863 DOI: 10.1101/2024.06.28.601249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Glucolipotoxicity, caused by combined hyperglycemia and hyperlipidemia, results in β-cell failure and type 2 diabetes (T2D) via cellular stress-related mechanisms. Activating transcription factor 4 (Atf4) is an essential effector of stress response. We show here that Atf4 expression in β-cells is dispensable for glucose homeostasis in young mice, but it is required for β-cell function during aging and under obesity-related metabolic stress. Henceforth, aged Atf4- deficient β-cells display compromised secretory function under acute hyperglycemia. In contrast, they are resistant to acute free fatty acid-induced loss-of identity and dysfunction. At molecular level, Atf4 -deficient β-cells down-regulate genes involved in protein translation, reducing β-cell identity gene products under high glucose. They also upregulate several genes involved in lipid metabolism or signaling, likely contributing to their resistance to free fatty acid-induced dysfunction. These results suggest that Atf4 activation is required for β-cell identity and function under high glucose, but this paradoxically induces β-cell failure in the presence of high levels of free fatty acids. Different branches of Atf4 activity could be manipulated for protecting β-cells from metabolic stress-induced failure. Highlights Atf4 is dispensable in β-cells in young miceAtf4 protects β-cells under high glucoseAtf4 exacerbate fatty acid-induced β-cell defectsAtf4 activates translation but depresses lipid-metabolism.
Collapse
|
14
|
Xue X, Wang L, Wu R, Li Y, Liu R, Ma Z, Jia K, Zhang Y, Li X. Si-Wu-Tang alleviates metabolic dysfunction-associated fatty liver disease by inhibiting ACSL4-mediated arachidonic acid metabolism and ferroptosis in MCD diet-fed mice. Chin Med 2024; 19:79. [PMID: 38844978 PMCID: PMC11157816 DOI: 10.1186/s13020-024-00953-7] [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: 03/19/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver disease worldwide. Si-Wu-Tang (SWT), a traditional Chinese medicine decoction has shown therapeutic effects on various liver diseases. However, the hepatoprotective effects and underlying mechanism of SWT on MAFLD remain unclear. METHODS First, a methionine-choline-deficient (MCD) diet-fed mice model was used and lipidomic analysis and transcriptomic analysis were performed. The contents of total iron ions, ferrous ions, and lipid peroxidation were detected and Prussian blue staining was performed to confirm the protective effects of SWT against ferroptosis. Finally, chemical characterization and network pharmacological analysis were employed to identify the potential active ingredients. RESULTS Serological and hepatic histopathological findings indicated SWT's discernible therapeutic impact on MCD diet-induced MAFLD. Lipidomic analysis revealed that SWT improved intrahepatic lipid accumulation by inhibiting TG synthesis and promoting TG transport. Transcriptomic analysis suggested that SWT ameliorated abnormal FA metabolism by inhibiting FA synthesis and promoting FA β-oxidation. Then, ferroptosis phenotype experiments revealed that SWT could effectively impede hepatocyte ferroptosis, which was induced by long-chain acyl-CoA synthetase 4 (ACSL4)-mediated esterification of arachidonic acid (AA). Finally, chemical characterization and network pharmacological analysis identified that paeoniflorin and other active ingredients might be responsible for the regulative effects against ferroptosis and MAFLD. CONCLUSION In conclusion, our study revealed the intricate mechanism through which SWT improved MCD diet-induced MAFLD by targeting FA metabolism and ferroptosis in hepatocytes, thus offering a novel therapeutic approach for the treatment of MAFLD and its complications.
Collapse
Affiliation(s)
- Xiaoyong Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Le Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiyu Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Zhi Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kexin Jia
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yinhao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
15
|
Guo P, Hu S, Liu X, He M, Li J, Ma T, Huang M, Fang Q, Wang Y. CAV3 alleviates diabetic cardiomyopathy via inhibiting NDUFA10-mediated mitochondrial dysfunction. J Transl Med 2024; 22:390. [PMID: 38671439 PMCID: PMC11055322 DOI: 10.1186/s12967-024-05223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The progression of diabetic cardiomyopathy (DCM) is noticeably influenced by mitochondrial dysfunction. Variants of caveolin 3 (CAV3) play important roles in cardiovascular diseases. However, the potential roles of CAV3 in mitochondrial function in DCM and the related mechanisms have not yet been elucidated. METHODS Cardiomyocytes were cultured under high-glucose and high-fat (HGHF) conditions in vitro, and db/db mice were employed as a diabetes model in vivo. To investigate the role of CAV3 in DCM and to elucidate the molecular mechanisms underlying its involvement in mitochondrial function, we conducted Liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis and functional experiments. RESULTS Our findings demonstrated significant downregulation of CAV3 in the cardiac tissue of db/db mice, which was found to be associated with cardiomyocyte apoptosis in DCM. Importantly, cardiac-specific overexpression of CAV3 effectively inhibited the progression of DCM, as it protected against cardiac dysfunction and cardiac remodeling associated by alleviating cardiomyocyte mitochondrial dysfunction. Furthermore, mass spectrometry analysis and immunoprecipitation assays indicated that CAV3 interacted with NDUFA10, a subunit of mitochondrial complex I. CAV3 overexpression reduced the degradation of lysosomal pathway in NDUFA10, restored the activity of mitochondrial complex I and improved mitochondrial function. Finally, our study demonstrated that CAV3 overexpression restored mitochondrial function and subsequently alleviated DCM partially through NDUFA10. CONCLUSIONS The current study provides evidence that CAV3 expression is significantly downregulated in DCM. Upregulation of CAV3 interacts with NDUFA10, inhibits the degradation of lysosomal pathway in NDUFA10, a subunit of mitochondrial complex I, restores the activity of mitochondrial complex I, ameliorates mitochondrial dysfunction, and thereby protects against DCM. These findings indicate that targeting CAV3 may be a promising approach for the treatment of DCM.
Collapse
Affiliation(s)
- Ping Guo
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Shuiqing Hu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Xiaohui Liu
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Miaomiao He
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jie Li
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Tingqiong Ma
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Man Huang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Qin Fang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Yan Wang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
16
|
Lei X, Peng Y, Li Y, Chen Q, Shen Z, Yin W, Lemiasheuski V, Xu S, He J. Effects of selenium nanoparticles produced by Lactobacillus acidophilus HN23 on lipid deposition in WRL68 cells. Bioorg Chem 2024; 145:107165. [PMID: 38367427 DOI: 10.1016/j.bioorg.2024.107165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/14/2024] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Selenium is an essential trace element for most organisms, protecting cells from oxidative damage caused by free radicals and serving as an adjunctive treatment for non-alcoholic fatty liver disease (NAFLD). In this study, We used the lactic acid bacterium Lactobacillus acidophilus HN23 to reduce tetra-valent sodium selenite into particulate matter, and analyzed it through inductively coupled plasma mass spectrometry (ICP-MS), scanning electron microscopy (SEM), X-ray diffraction energy dispersive spectrometry (EDS), and Fourier transform infrared spectroscopy (FTIR). We found that it consisted of selenium nanoparticles (SeNPs) with a mass composition of 65.8 % zero-valent selenium and some polysaccharide and polypeptide compounds, with particle sizes ranging from 60 to 300 nm. We also detected that SeNPs were much less toxic to cells than selenite. We further used free fatty acids (FFA)-induced WRL68 fatty liver cell model to study the therapeutic effect of SeNPs on NAFLD. The results show that SeNPs are more effective than selenite in reducing lipid deposition, increasing mitochondrial membrane potential (MMP) and antioxidant capacity of WRL68 cells, which is attributed to the chemical valence state of selenium and organic composition in SeNPs. In conclusion, SeNPs produced by probiotics L. acidophilus had the potential to alleviate NAFLD by reducing hepatocyte lipid deposition and oxidative damage. This study may open a new avenue for SeNPs drug development to treat NAFLD.
Collapse
Affiliation(s)
- Xianglan Lei
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; College of Tropical Agricultural Technology, Hainan Vocational University, Haikou 570100, China
| | - Yuxuan Peng
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; College of Tropical Agricultural Technology, Hainan Vocational University, Haikou 570100, China; Faculty of Biology, Belarusian State University, 220030 Minsk, Belarus
| | - Yan Li
- International Sakharov Environmental Institute, Belarusian State University, 220030 Minsk, Belarus
| | - Qianyuan Chen
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenguo Shen
- College of Tropical Agricultural Technology, Hainan Vocational University, Haikou 570100, China
| | - Wen Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Indus-trial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Viktar Lemiasheuski
- International Sakharov Environmental Institute, Belarusian State University, 220030 Minsk, Belarus; All-Russian Research Institute of Physiology, Biochemistry and Nutrition of Animals - Branch of the Federal Research Center for Animal Husbandry Named After Academy Member L. K. Ernst, Institute, 249013, Borovsk, Russian Federation
| | - Siyang Xu
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Jin He
- National Key Laboratory of Agricultural Microbiology & Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
17
|
Zhong H, Jin Y, Abdullah, Hussain M, Liu X, Feng F, Guan R. Recent advances of hepatoprotective peptides: Production, structure, mechanisms, and interactions with intestinal microbiota. FOOD BIOSCI 2024; 58:103744. [DOI: 10.1016/j.fbio.2024.103744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Dong J, Li M, Peng R, Zhang Y, Qiao Z, Sun N. ACACA reduces lipid accumulation through dual regulation of lipid metabolism and mitochondrial function via AMPK- PPARα- CPT1A axis. J Transl Med 2024; 22:196. [PMID: 38395901 PMCID: PMC10885411 DOI: 10.1186/s12967-024-04942-0] [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: 12/09/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a multifaceted metabolic disorder, whose global prevalence is rapidly increasing. Acetyl CoA carboxylases 1 (ACACA) is the key enzyme that controls the rate of fatty acid synthesis. Hence, it is crucial to investigate the function of ACACA in regulating lipid metabolism during the progress of NAFLD. METHODS Firstly, a fatty liver mouse model was established by high-fat diet at 2nd, 12th, and 20th week, respectively. Then, transcriptome analysis was performed on liver samples to investigate the underlying mechanisms and identify the target gene of the occurrence and development of NAFLD. Afterwards, lipid accumulation cell model was induced by palmitic acid and oleic acid (PA ∶ OA molar ratio = 1∶2). Next, we silenced the target gene ACACA using small interfering RNAs (siRNAs) or the CMS-121 inhibitor. Subsequently, experiments were performed comprehensively the effects of inhibiting ACACA on mitochondrial function and lipid metabolism, as well as on AMPK- PPARα- CPT1A pathway. RESULTS This data indicated that the pathways significantly affected by high-fat diet include lipid metabolism and mitochondrial function. Then, we focus on the target gene ACACA. In addition, the in vitro results suggested that inhibiting of ACACA in vitro reduces intracellular lipid accumulation, specifically the content of TG and TC. Furthermore, ACACA ameliorated mitochondrial dysfunction and alleviate oxidative stress, including MMP complete, ATP and ROS production, as well as the expression of mitochondria respiratory chain complex (MRC) and AMPK proteins. Meanwhile, ACACA inhibition enhances lipid metabolism through activation of PPARα/CPT1A, leading to a decrease in intracellular lipid accumulation. CONCLUSION Targeting ACACA can reduce lipid accumulation by mediating the AMPK- PPARα- CPT1A pathway, which regulates lipid metabolism and alleviates mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jian Dong
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Muzi Li
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Runsheng Peng
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Key Laboratory of Biotechnology & Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Yuchuan Zhang
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Zilin Qiao
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou, China
- Gansu Provincial Bioengineering Materials Engineering Research Center, Lanzhou, China
| | - Na Sun
- Gansu Technology Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou, China.
- Key Laboratory of Biotechnology & Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
| |
Collapse
|
19
|
Guo L, Liu JJ, Long SY, Wang PY, Li S, Wang JL, Wei XF, Li J, Lei L, Huang AL, Hu JL. TIM22 and TIM29 inhibit HBV replication by up-regulating SRSF1 expression. J Med Virol 2024; 96:e29439. [PMID: 38294104 DOI: 10.1002/jmv.29439] [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: 09/13/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
Hepatitis B virus (HBV) infection is a serious global health problem. After the viruses infect the human body, the host can respond to the virus infection by coordinating various cellular responses, in which mitochondria play an important role. Evidence has shown that mitochondrial proteins are involved in host antiviral responses. In this study, we found that the overexpression of TIM22 and TIM29, the members of the inner membrane translocase TIM22 complex, significantly reduced the level of intracellular HBV DNA and RNA and secreted HBV surface antigens and E antigen. The effects of TIM22 and TIM29 on HBV replication and transcription is attributed to the reduction of core promoter activity mediated by the increased expression of SRSF1 which acts as a suppressor of HBV replication. This study provides new evidence for the critical role of mitochondria in the resistance of HBV infection and new targets for the development of treatment against HBV infection.
Collapse
Affiliation(s)
- Lin Guo
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Clinical Laboratory, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Jia-Jun Liu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shao-Yuan Long
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Pei-Yun Wang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shan Li
- Department of Clinical Laboratory, the Sixth Hospital of Chengdu, Chengdu, China
| | - Jin-Lan Wang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xia-Fei Wei
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Jie Li
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ling Lei
- Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jie-Li Hu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Cui X, Yang Y, Zhang M, Bao L, Jiao F, Liu S, Wang H, Wei X, Qian W, Shi X, Su C, Qian Y. Mulberry leaves supplementation alters lipid metabolism and promotes fatty acid β oxidation in growing mutton sheep. J Anim Sci 2024; 102:skae076. [PMID: 38908013 PMCID: PMC11196999 DOI: 10.1093/jas/skae076] [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: 03/23/2023] [Accepted: 06/21/2024] [Indexed: 06/24/2024] Open
Abstract
Mulberry leaves (MLs) are an unconventional feed with fiber and various active ingredients, and are acknowledged as likely to regulate lipid metabolism, while the molecular mechanism remains undefined. Therefore, our objective was to define the role of MLs on the overall lipid metabolism. We conducted a feeding experiment of three groups on growing mutton sheep fed with dried mulberry leaves (DMLs), with fermented mulberry leaves (FMLs), or without MLs (as control). Analyses of transcriptome and widely target lipids demonstrated the addition of MLs triggered big perturbations in genes and metabolites related to glycerolipid, phospholipid, ether lipid, and sphingolipid metabolism. Additionally, the variations of the above lipids in the treatment of MLs possibly facilitate immunity enhancement of growing mutton sheep via the activation of complement and coagulation cascades. Furthermore, treatments with MLs could expedite proceedings of lipid degradation and fatty acid β oxidation in mitochondria, thereby to achieve the effect of lipid reduction. Besides, added DMLs also fuel fatty acid β-oxidation in peroxisomes and own much stronger lipolysis than added FMLs, possibly attributed to high fiber content in DMLs. These findings establish the novel lipid-lowering role and immune protection of MLs, which lays the foundation for the medicinal application of MLs.
Collapse
Affiliation(s)
- Xiaopeng Cui
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212000, China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Minjuan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lijun Bao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Feng Jiao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shuang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hexin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xinlan Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wei Qian
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiang Shi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chao Su
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yonghua Qian
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- Shenzhen Fengnong Holding Co., Ltd, Shenzhen, Guangdong 518000, China
| |
Collapse
|
21
|
Meng K, Lu S, Li Y, Hu L, Zhang J, Cao Y, Wang Y, Zhang CZ, He Q. LINC00493-encoded microprotein SMIM26 exerts anti-metastatic activity in renal cell carcinoma. EMBO Rep 2023; 24:e56282. [PMID: 37009826 PMCID: PMC10240204 DOI: 10.15252/embr.202256282] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 04/04/2023] Open
Abstract
Human microproteins encoded by long non-coding RNAs (lncRNA) have been increasingly discovered, however, complete functional characterization of these emerging proteins is scattered. Here, we show that LINC00493-encoded SMIM26, an understudied microprotein localized in mitochondria, is tendentiously downregulated in clear cell renal cell carcinoma (ccRCC) and correlated with poor overall survival. LINC00493 is recognized by RNA-binding protein PABPC4 and transferred to ribosomes for translation of a 95-amino-acid protein SMIM26. SMIM26, but not LINC00493, suppresses ccRCC growth and metastatic lung colonization by interacting with acylglycerol kinase (AGK) and glutathione transport regulator SLC25A11 via its N-terminus. This interaction increases the mitochondrial localization of AGK and subsequently inhibits AGK-mediated AKT phosphorylation. Moreover, the formation of the SMIM26-AGK-SCL25A11 complex maintains mitochondrial glutathione import and respiratory efficiency, which is abrogated by AGK overexpression or SLC25A11 knockdown. This study functionally characterizes the LINC00493-encoded microprotein SMIM26 and establishes its anti-metastatic role in ccRCC, and therefore illuminates the importance of hidden proteins in human cancers.
Collapse
Affiliation(s)
- Kun Meng
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan UniversityGuangzhouChina
| | - Shaohua Lu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
- Sino‐French Hoffmann Institute, School of Basic Medical Sciences, State Key Laboratory of Respiratory DiseaseGuangzhou Medical UniversityGuangzhouChina
| | - Yu‐Ying Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
| | - Li‐Ling Hu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
| | - Jing Zhang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan UniversityGuangzhouChina
| | - Yun Cao
- Department of Pathology, State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
| | - Chris Zhiyi Zhang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
| | - Qing‐Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan UniversityGuangzhouChina
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan UniversityGuangzhouChina
| |
Collapse
|
22
|
Chen L, Wang Y. Interdisciplinary advances reshape the delivery tools for effective NASH treatment. Mol Metab 2023; 73:101730. [PMID: 37142161 DOI: 10.1016/j.molmet.2023.101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH), a severe systemic and inflammatory subtype of nonalcoholic fatty liver disease, eventually develops into cirrhosis and hepatocellular carcinoma with few options for effective treatment. Currently potent small molecules identified in preclinical studies are confronted with adverse effects and long-term ineffectiveness in clinical trials. Nevertheless, highly specific delivery tools designed from interdisciplinary concepts may address the significant challenges by either effectively increasing the concentrations of drugs in target cell types, or selectively manipulating the gene expression in liver to resolve NASH. SCOPE OF REVIEW We focus on dissecting the detailed principles of the latest interdisciplinary advances and concepts that direct the design of future delivery tools to enhance the efficacy. Recent advances have indicated that cell and organelle-specific vehicles, non-coding RNA research (e.g. saRNA, hybrid miRNA) improve the specificity, while small extracellular vesicles and coacervates increase the cellular uptake of therapeutics. Moreover, strategies based on interdisciplinary advances drastically elevate drug loading capacity and delivery efficiency and ameliorate NASH and other liver diseases. MAJOR CONCLUSIONS The latest concepts and advances in chemistry, biochemistry and machine learning technology provide the framework and strategies for the design of more effective tools to treat NASH, other pivotal liver diseases and metabolic disorders.
Collapse
Affiliation(s)
- Linshan Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health.
| |
Collapse
|
23
|
Fang J, Yu CH, Li XJ, Yao JM, Fang ZY, Yoon SH, Yu WY. Gut dysbiosis in nonalcoholic fatty liver disease: pathogenesis, diagnosis, and therapeutic implications. Front Cell Infect Microbiol 2022; 12:997018. [PMID: 36425787 PMCID: PMC9679376 DOI: 10.3389/fcimb.2022.997018] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/19/2022] [Indexed: 07/21/2023] Open
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) is increasing recently and has become one of the most common clinical liver diseases. Since the pathogenesis of NAFLD has not been completely elucidated, few effective therapeutic drugs are available. As the "second genome" of human body, gut microbiota plays an important role in the digestion, absorption and metabolism of food and drugs. Gut microbiota can act as an important driver to advance the occurrence and development of NAFLD, and to accelerate its progression to cirrhosis and hepatocellular carcinoma. Growing evidence has demonstrated that gut microbiota and its metabolites directly affect intestinal morphology and immune response, resulting in the abnormal activation of inflammation and intestinal endotoxemia; gut dysbiosis also causes dysfunction of gut-liver axis via alteration of bile acid metabolism pathway. Because of its composition diversity and disease-specific expression characteristics, gut microbiota holds strong promise as novel biomarkers and therapeutic targets for NAFLD. Intervening intestinal microbiota, such as antibiotic/probiotic treatment and fecal transplantation, has been a novel strategy for preventing and treating NAFLD. In this article, we have reviewed the emerging functions and association of gut bacterial components in different stages of NAFLD progression and discussed its potential implications in NAFLD diagnosis and therapy.
Collapse
Affiliation(s)
- Jie Fang
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chen-Huan Yu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Cancer Hospital, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xue-Jian Li
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jin-Mei Yao
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng-Yu Fang
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Soo-Hyun Yoon
- Institute of Medical Science, Wonkwang University, Iksan, South Korea
| | - Wen-Ying Yu
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Song C, Li W, Wang Z. The Landscape of Liver Chromatin Accessibility and Conserved Non-coding Elements in Larimichthys crocea, Nibea albiflora, and Lateolabrax maculatus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:763-775. [PMID: 35895229 DOI: 10.1007/s10126-022-10142-y] [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: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Large yellow croaker (Larimichthys crocea), yellow drum (Nibea albiflora), and Chinese seabass (Lateolabrax maculatus) are important economic marine fishes in China. The conserved non-coding elements (CNEs) in the liver tissues of the three kinds of fish are directly or indirectly involved in the regulation of gene expression and affect liver functions. However, the fishes' CNEs and even chromatin accessibility landscape have not been effectively investigated. Hence, this study established the landscapes of the fishes' genome-wide chromatin accessibility and CNEs by detecting regions of the open chromatin in their livers using an assay for transposase-accessible chromatin by high-throughput sequencing (ATAC-seq) and comparative genomics approach. The results showed that Smad1, Sp1, and Foxl1 transcription factor binding motifs were considerably enriched in the chromatin accessibility landscape in the liver of the three species, and the three transcription factors (TFs) had a wide range of common targets. The hypothetical gene set was targeted by one, two, or all three TFs, which was much higher than would be expected for an accidental outcome. The gene sets near the CNEs were mainly enriched through processes such as a macromolecule metabolic process and ribonucleoprotein complex biogenesis. The active CNEs were found in the promoter regions of genes such as ap1g1, hax1, and ndufs2. And 5 CNEs were predicted to be highly conserved active enhancers. These results demonstrated that Smad1, Sp1, and Foxl1 might be related to the liver function in the three fishes. In addition, we found a series of ATAC-seq-labeled CNEs located in the gene promoter regions, and highly conserved H3k27ac + -labeled CNEs located in the liver function genes. The highly conserved nature of these regulatory elements suggests that they play important roles in the liver in fish. This study mined the landscape of chromatin accessibility and CNEs of three important economic fishes to fill the knowledge gaps in this field. Moreover, the work provides useful data for the industrial application and theoretical research of these three fish species.
Collapse
Affiliation(s)
- Chaowei Song
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Wanbo Li
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiyong Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|