1
|
Gospodaryov DV. Alternative NADH dehydrogenase: A complex I backup, a drug target, and a tool for mitochondrial gene therapy. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149529. [PMID: 39615731 DOI: 10.1016/j.bbabio.2024.149529] [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: 03/28/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Alternative NADH dehydrogenase, also known as type II NADH dehydrogenase (NDH-2), catalyzes the same redox reaction as mitochondrial respiratory chain complex I. Specifically, it oxidizes reduced nicotinamide adenine dinucleotide (NADH) while simultaneously reducing ubiquinone to ubiquinol. However, unlike complex I, this enzyme is non-proton pumping, comprises of a single subunit, and is resistant to rotenone. Initially identified in bacteria, fungi and plants, NDH-2 was subsequently discovered in protists and certain animal taxa including sea squirts. The gene coding for NDH-2 is also present in the genomes of some annelids, tardigrades, and crustaceans. For over two decades, NDH-2 has been investigated as a potential substitute for defective complex I. In model organisms, NDH-2 has been shown to ameliorate a broad spectrum of conditions associated with complex I malfunction, including symptoms of Parkinson's disease. Recently, lifespan extension has been observed in animals expressing NDH-2 in a heterologous manner. A variety of mechanisms have been put forward by which NDH-2 may extend lifespan. Such mechanisms include the activation of pro-longevity pathways through modulation of the NAD+/NADH ratio, decreasing production of reactive oxygen species (ROS) in mitochondria, or then through moderate increases in ROS production followed by activation of defense pathways (mitohormesis). This review gives an overview of the latest research on NDH-2, including the structural peculiarities of NDH-2, its inhibitors, its role in the pathogenicity of mycobacteria and apicomplexan parasites, and its function in bacteria, fungi, and animals.
Collapse
Affiliation(s)
- Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka, 76018, Ivano-Frankivsk, Ukraine.
| |
Collapse
|
2
|
McGill Percy KC, Liu Z, Qi X. Mitochondrial dysfunction in Alzheimer's disease: Guiding the path to targeted therapies. Neurotherapeutics 2025; 22:e00525. [PMID: 39827052 DOI: 10.1016/j.neurot.2025.e00525] [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: 08/31/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration, marked by the accumulation of amyloid-β (Aβ) plaques and tau tangles. Emerging evidence suggests that mitochondrial dysfunction plays a pivotal role in AD pathogenesis, driven by impairments in mitochondrial quality control (MQC) mechanisms. MQC is crucial for maintaining mitochondrial integrity through processes such as proteostasis, mitochondrial dynamics, mitophagy, and precise communication with other subcellular organelles. In AD, disruptions in these processes lead to bioenergetic failure, gene dysregulation, the accumulation of damaged mitochondria, neuroinflammation, and lipid homeostasis impairment, further exacerbating neurodegeneration. This review elucidates the molecular pathways involved in MQC and their pathological relevance in AD, highlighting recent discoveries related to mitochondrial mechanisms underlying neurodegeneration. Furthermore, we explore potential therapeutic strategies targeting mitochondrial dysfunction, including gene therapy and pharmacological interventions, offering new avenues for slowing AD progression. The complex interplay between mitochondrial health and neurodegeneration underscores the need for innovative approaches to restore mitochondrial function and mitigate the onset and progression of AD.
Collapse
Affiliation(s)
- Kyle C McGill Percy
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zunren Liu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Center for Mitochondrial Research and Therapeutics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
3
|
Parkhitko AA, Cracan V. Xenotopic synthetic biology: Prospective tools for delaying aging and age-related diseases. SCIENCE ADVANCES 2025; 11:eadu1710. [PMID: 40153513 DOI: 10.1126/sciadv.adu1710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
Metabolic dysregulation represents one of the major driving forces in aging. Although multiple genetic and pharmacological manipulations are known to extend longevity in model organisms, aging is a complex trait, and targeting one's own genes may be insufficient to prevent age-dependent deterioration. An alternative strategy could be to use enzymes from other species to reverse age-associated metabolic changes. In this review, we discuss a set of enzymes from lower organisms that have been shown to affect various metabolic parameters linked to age-related processes. These enzymes include modulators of steady-state levels of amino acids (METase, ASNase, and ADI), NADPH/NADP+ and/or reduced form of coenzyme Q (CoQH2)/CoQ redox potentials (NDI1, AOX, LbNOX, TPNOX, EcSTH, RquA, LOXCAT, Grubraw, and ScURA), GSH (StGshF), mitochondrial membrane potential (mtON and mito-dR), or reactive oxygen species (DAAO and KillerRed-SOD1). We propose that leveraging non-mammalian enzymes represents an untapped resource that can be used to delay aging and age-related diseases.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
4
|
Zhang X, Chen J, Sheng X, Ding CF, Yan Y. Preparation and characterization of a biomimetic honeycomb cross-linked chitosan membrane and its application in the serum of gastric cancer patients. Int J Biol Macromol 2024; 279:135367. [PMID: 39244117 DOI: 10.1016/j.ijbiomac.2024.135367] [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: 06/20/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Chitosan, as a biological macromolecule with excellent biocompatibility, has great potential for application in immobilized metal affinity chromatography (IMAC) strategies. In-depth analysis of low-abundance phosphopeptides in organisms can help reveal the pathological mechanisms of diseases. Here, we developed an IMAC material based on a biomimetic honeycomb chitosan membrane. The material demonstrates excellent biocompatibility, good hydrophilicity, and strong metal chelating capacity, which collectively confer outstanding enrichment properties. The material has high sensitivity (0.05 fmol), great selectivity (1:2000), excellent cycling stability (at least 10 cycles) and acid-base stability. In addition, the material was employed in human serum, successfully enriching 129 phosphopeptides from the serum of gastric cancer patients and 146 phosphopeptides from healthy controls. Sequence logo suggests a potential association between gastric cancer and glutamine. Ultimately, an in-depth gene ontology analysis was carried out on the phosphopeptides that were enriched in the serum samples. Compared to normal controls, our results demonstrated dysregulated expression of biological process, cellular component, and molecular function in gastric cancer patients. This suggests that the disease involves, such as blood coagulation pathways, cholesterol metabolism, and heparin binding. All experimental outcomes converge to demonstrate the substantial promise of the material for applications within proteomics research.
Collapse
Affiliation(s)
- Xiaoya Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Jiakai Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Xiuqin Sheng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Chuan-Fan Ding
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, PR China.
| | - Yinghua Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
5
|
Li H, Chen Z, Shen Y, Xiong T, Chen A, Chen L, Ye Y, Jiang Q, Zhang Y, Sun J, Shen L. Gene therapy in Aβ-induced cell and mouse models of Alzheimer's disease through compensating defective mitochondrial complex I function. J Transl Med 2024; 22:760. [PMID: 39143479 PMCID: PMC11323700 DOI: 10.1186/s12967-024-05571-3] [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: 05/09/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurogenerative disorder without effective treatments. Defects in mitochondrial complex I are thought to contribute to AD pathogenesis. The aim of this study is to explore whether a novel gene therapy transducing yeast complex I gene NDI1 can be used to treat AD with severely reduced complex I function in cell and animal models. METHODS The differentiated human neural cells were induced by Aβ1-42 to establish the AD cell model, and adeno-associated virus serotype 9 (AAV9) was used to transduce yeast NDI1 into the cell model. Aβ1-42 was injected into the hippocampus area of the brain to establish the AD mouse model. AAV9-NDI1 was injected stereotaxically into the hippocampus area to test the therapeutic effect. RESULTS The expressed yeast complex I had an ameliorating effect on the defective function of human complex I and cellular pathological characteristics in the AD cell model. Furthermore, AAV9-NDI1 gene therapy in the hippocampus had a therapeutic effect on various aspects of mitochondrial function, histopathological characteristics and neurological defects in the AD mouse model. In addition, AAV9-NDI1 injection into the hippocampus of normal mice did not cause any adverse effect. CONCLUSIONS Compensating mitochondrial complex I function with yeast NDI1 is effective for gene therapy in Aβ-induced AD cell and mouse models. The results of this study offer a novel strategy and approach for treating AD types characterized by complex I abnormalities.
Collapse
Affiliation(s)
- Hongzhi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Northern Zhongxin Road, Chashan University Town, Wenzhou, Zhejiang, 325035, China.
| | - Zhuo Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuqi Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ting Xiong
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Andong Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lixia Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yifan Ye
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qingyou Jiang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yaxi Zhang
- Brain Center, Wenzhou Central Hospital, Wenzhou, 325000, China
| | - Jun Sun
- Brain Center, Wenzhou Central Hospital, Wenzhou, 325000, China.
| | - Luxi Shen
- Department of Internal Neurology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
6
|
Shang X, Wang H, Gu J, Zhao X, Zhang J, Sun B, Zhu X. Ferroptosis-related gene transferrin receptor protein 1 expression correlates with the prognosis and tumor immune microenvironment in cervical cancer. PeerJ 2024; 12:e17842. [PMID: 39131609 PMCID: PMC11313409 DOI: 10.7717/peerj.17842] [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: 04/02/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Background Ferroptosis is a non-apoptotic iron-dependent form of cell death implicated in various cancer pathologies. However, its precise role in tumor growth and progression of cervical cancer (CC) remains unclear. Transferrin receptor protein 1 (TFRC), a key molecule associated with ferroptosis, has been identified as influencing a broad range of pathological processes in different cancers. However, the prognostic significance of TFRC in CC remains unclear. The present study utilized bioinformatics to explore the significance of the ferroptosis-related gene TFRC in the progression and prognosis of CC. Methods We obtained RNA sequencing data and corresponding clinical information on patients with CC from The Cancer Genome Atlas (TCGA), Genotype Tissue Expression (GTEx) and Gene Expression Omnibus (GEO) databases. Using least absolute shrinkage and selection operator (LASSO) Cox regression, we then generated a multigene signature of five ferroptosis-related genes (FRGs) for the prognostic prediction of CC. We investigated the relationship between TFRC gene expression and immune cell infiltration by employing single-sample GSEA (ssGSEA) analysis. The potential functional role of the TFRC gene was evaluated through gene set enrichment analysis (GSEA). Immunohistochemistry and qPCR was employed to assess TFRC mRNA and protein expression in 33 cases of cervical cancer. Furthermore, the relationship between TFRC mRNA expression and overall survival (OS) was investigated in patients. Results CC samples had significantly higher TFRC gene expression levels than normal tissue samples. Higher TFRC gene expression levels were strongly associated with higher cancer T stages and OS events. The findings of multivariate analyses illustrated that the OS in CC patients with high TFRC expression is shorter than in patients with low TFRC expression. Significant increases were observed in the levels of TFRC mRNA and protein expression in patients diagnosed with CC. Conclusion Increased TFRC expression in CC was associated with disease progression, an unfavorable prognosis, and dysregulated immune cell infiltration. In addition, it highlights ferroptosis as a promising therapeutic target for CC.
Collapse
Affiliation(s)
- Xiujuan Shang
- Department of Laboratory Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang, Jiangsu, China
| | - Hongdong Wang
- Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Jin Gu
- Department of Laboratory Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang, Jiangsu, China
| | - Xiaohui Zhao
- Department of Laboratory Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang, Jiangsu, China
| | - Jing Zhang
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bohao Sun
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinming Zhu
- Department of Laboratory Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang, Jiangsu, China
| |
Collapse
|
7
|
Jin X, Dong W, Chang K, Yan Y. Research on the signaling pathways related to the intervention of traditional Chinese medicine in Parkinson's disease:A literature review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117850. [PMID: 38331124 DOI: 10.1016/j.jep.2024.117850] [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: 10/05/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is the most common progressive neurodegenerative disorder affecting more than 10 million people worldwide and is characterized by the progressive loss of Daergic (DA) neurons in the substantia nigra pars compacta. It has been reported that signaling pathways play a crucial role in the pathogenesis of PD, while the active ingredients of traditional Chinese medicine (TCM) have been found to possess a protective effect against PD. TCM has demonstrated significant potential in mitigating oxidative stress (OS), neuroinflammation, and apoptosis of DA neurons via the regulation of signaling pathways associated with PD. AIM OF THE REVIEW This study discussed and analyzed the signaling pathways involved in the occurrence and development of PD and the mechanism of active ingredients of TCM regulating PD via signaling pathways, with the aim of providing a basis for the development and clinical application of therapeutic strategies for TCM in PD. MATERIALS AND METHODS With "Parkinson's disease", "Idiopathic Parkinson's Disease", "Lewy Body Parkinson's Disease", "Parkinson's Disease, Idiopathic", "Parkinson Disease, Idiopathic", "Parkinson's disorders", "Parkinsonism syndrome", "Traditional Chinese medicine", "Chinese herbal medicine", "active ingredients", "medicinal plants" as the main keywords, PubMed, Web of Science and other online search engines were used for literature retrieval. RESULTS PD exhibits a close association with various signaling pathways, including but not limited to MAPKs, NF-κB, PI3K/Akt, Nrf2/ARE, Wnt/β-catenin, TLR/TRIF, NLRP3, Notch. The therapeutic potential of TCM lies in its ability to regulate these signaling pathways. In addition, the active ingredients of TCM have shown significant effects in improving OS, neuroinflammation, and DA neuron apoptosis in PD. CONCLUSION The active ingredients of TCM have unique advantages in regulating PD-related signaling pathways. It is suggested to combine network pharmacology and bioinformatics to study the specific targets of TCM. This not only provides a new way for the prevention and treatment of PD with the active ingredients of TCM, but also provides a scientific basis for the selection and development of TCM preparations.
Collapse
Affiliation(s)
- Xiaxia Jin
- National Key Laboratory of Quality Assurance and Sustainable Utilization of Authentic Medicinal Materials, Chinese Medicine Resource Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wendi Dong
- Foshan Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Foshan 528000, China
| | - Kaile Chang
- Shaanxi University of Traditional Chinese Medicine, Xianyang, 712046, China
| | - Yongmei Yan
- National Key Laboratory of Quality Assurance and Sustainable Utilization of Authentic Medicinal Materials, Chinese Medicine Resource Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang 712000, China.
| |
Collapse
|
8
|
Liu ZY, Lin LC, Liu ZY, Yang JJ, Tao H. m6A epitranscriptomic and epigenetic crosstalk in cardiac fibrosis. Mol Ther 2024; 32:878-889. [PMID: 38311850 PMCID: PMC11163196 DOI: 10.1016/j.ymthe.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiac fibrosis, a crucial pathological characteristic of various cardiac diseases, presents a significant treatment challenge. It involves the deposition of the extracellular matrix (ECM) and is influenced by genetic and epigenetic factors. Prior investigations have predominantly centered on delineating the substantial influence of epigenetic and epitranscriptomic mechanisms in driving the progression of fibrosis. Recent studies have illuminated additional avenues for modulating the progression of fibrosis, offering potential solutions to the challenging issues surrounding fibrosis treatment. In the context of cardiac fibrosis, an intricate interplay exists between m6A epitranscriptomic and epigenetics. This interplay governs various pathophysiological processes: mitochondrial dysfunction, mitochondrial fission, oxidative stress, autophagy, apoptosis, pyroptosis, ferroptosis, cell fate switching, and cell differentiation, all of which affect the advancement of cardiac fibrosis. In this comprehensive review, we meticulously analyze pertinent studies, emphasizing the interplay between m6A epitranscriptomics and partial epigenetics (including histone modifications and noncoding RNA), aiming to provide novel insights for cardiac fibrosis treatment.
Collapse
Affiliation(s)
- Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| |
Collapse
|
9
|
Albadawi E, El-Tokhy A, Albadrani M, Adel M, El-Gamal R, Zaarina W, El-Agawy MSED, Elsayed HRH. The role of stinging nettle (Urtica dioica L.) in the management of rotenone-induced Parkinson's disease in rats. Tissue Cell 2024; 87:102328. [PMID: 38387425 DOI: 10.1016/j.tice.2024.102328] [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: 10/17/2023] [Revised: 01/27/2024] [Accepted: 02/11/2024] [Indexed: 02/24/2024]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative conditions. Alpha-synuclein deposition, Lewy bodies (LBs) formation, disruption of the autophagic machinery, apoptosis of substantia nigra dopaminergic neurons, oxidative stress, and neuroinflammation are all pathologic hallmarks of PD. The leaves of the stinging Nettle (Urtica dioica L.) have a long history as an herbal cure with antioxidant, anti-inflammatory, anti-cancer, immunomodulatory, and neuroprotective properties. The current study aims for the first time to investigate the role of Nettle supplementation on Rotenone-induced PD. Rats were divided into five groups; a Saline control, Nettle control (100 mg/kg/day), Rotenone control (2 mg/kg/day), Rotenone + Nettle (50 mg /kg/day), and Rotenone + Nettle (100 mg/kg). After four weeks, the rats were examined for behavioral tests. The midbrains were investigated for histopathological alteration and immunohistochemical reaction for Tyrosine hydroxylase in the dopaminergic neurons, α-synuclein for Lewy bodies, caspase 3 for apoptotic neurons, LC3 and P62 for autophagic activity. Midbrain homogenates were examined for oxidative stress markers. mRNA expression of TNFα and Il6; inflammatory markers, Bcl-2, BAX and Caspase 3; apoptosis markers, were detected in midbrains. The results showed that Nettle caused recovery of midbrain dopaminergic neurons, by inhibiting apoptosis, inflammation, and oxidative stress and by restoring the autophagic machinery with clearance of α-synuclein deposits. We can conclude that Nettle is a potentially effective adjuvant in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Emad Albadawi
- Department of Anatomy, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Ahmed El-Tokhy
- Plant Protection Department, Faculty of Agriculture, New Valley University, El-Kharga, Egypt
| | - Muayad Albadrani
- Department of Family and Community Medicine, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Mohammed Adel
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Egypt; Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Medical Biochemistry, Horus University in Egypt (HUE), New Damietta, Damietta, Egypt
| | - Wael Zaarina
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Anatomy, Faculty of Medicine, Mansoura National University, Gamasa, Egypt
| | - Mosaab Salah El-Din El-Agawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Hassan Reda Hassan Elsayed
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Department of Anatomy and Neurobiology, College of Medicine and Health Sciences, National University of Science and Technology, Sohar, Oman.
| |
Collapse
|
10
|
Liu X, Yan Q, Liu X, Wei W, Zou L, Zhao F, Zeng S, Yi L, Ding H, Zhao M, Chen J, Fan S. PKM2 induces mitophagy through the AMPK-mTOR pathway promoting CSFV proliferation. J Virol 2024; 98:e0175123. [PMID: 38319105 PMCID: PMC10949426 DOI: 10.1128/jvi.01751-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/14/2023] [Indexed: 02/07/2024] Open
Abstract
Viruses exploit the host cell's energy metabolism system to support their replication. Mitochondria, known as the powerhouse of the cell, play a critical role in regulating cell survival and virus replication. Our prior research indicated that the classical swine fever virus (CSFV) alters mitochondrial dynamics and triggers glycolytic metabolic reprogramming. However, the role and mechanism of PKM2, a key regulatory enzyme of glycolytic metabolism, in CSFV replication remain unclear. In this study, we discovered that CSFV enhances PKM2 expression and utilizes PKM2 to inhibit pyruvate production. Using an affinity purification coupled mass spectrometry system, we successfully identified PKM as a novel interaction partner of the CSFV non-structural protein NS4A. Furthermore, we validated the interaction between PKM2 and both CSFV NS4A and NS5A through co-immunoprecipitation and confocal analysis. PKM2 was found to promote the expression of both NS4A and NS5A. Moreover, we observed that PKM2 induces mitophagy by activating the AMPK-mTOR signaling pathway, thereby facilitating CSFV proliferation. In summary, our data reveal a novel mechanism whereby PKM2, a metabolic enzyme, promotes CSFV proliferation by inducing mitophagy. These findings offer a new avenue for developing antiviral strategies. IMPORTANCE Viruses rely on the host cell's material-energy metabolic system for replication, inducing host metabolic disorders and subsequent immunosuppression-a major contributor to persistent viral infections. Classical swine fever virus (CSFV) is no exception. Classical swine fever is a severe acute infectious disease caused by CSFV, resulting in significant economic losses to the global pig industry. While the role of the metabolic enzyme PKM2 (pyruvate dehydrogenase) in the glycolytic pathway of tumor cells has been extensively studied, its involvement in viral infection remains relatively unknown. Our data unveil a new mechanism by which the metabolic enzyme PKM2 mediates CSFV infection, offering novel avenues for the development of antiviral strategies.
Collapse
Affiliation(s)
- Xiaodi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Quanhui Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Xueyi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Wenkang Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Linke Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Feifan Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guang Dong, China
| |
Collapse
|
11
|
Hang PZ, Ge FQ, Zhang MR, Li QH, Yu HQ, Song YC, Guo DD, Zhao J, Zhu H. BDNF mimetic 7,8-dihydroxyflavone rescues rotenone-induced cytotoxicity in cardiomyocytes by ameliorating mitochondrial dysfunction. Free Radic Biol Med 2023; 198:83-91. [PMID: 36764626 DOI: 10.1016/j.freeradbiomed.2023.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The relationship between mitochondrial dysfunction and cardiovascular disease pathogenesis is well recognized. 7,8-Dihydroxyflavone (7,8-DHF), a mimetic of brain-derived neurotrophic factor, inhibits mitochondrial impairments and improves cardiac function. However, the regulatory role of 7,8-DHF in the mitochondrial function of cardiomyocytes is not fully understood. To investigate the potential mito-protective effects of 7,8-DHF in cardiomyocytes, we treated H9c2 or HL-1 cells with the mitochondrial respiratory complex I inhibitor rotenone (Rot) as an in vitro model of mitochondrial dysfunction. We found that 7,8-DHF effectively eliminated various concentrations of Rot-induced cell death and reduced lactate dehydrogenase release. 7,8-DHF significantly improved mitochondrial membrane potential and inhibited mitochondrial reactive oxygen species. Moreover, 7,8-DHF decreased routine and leak respiration, restored protein levels of mitochondrial complex I-IV, and increased ATP production in Rot-treated H9c2 cells. The protective role of 7,8-DHF in Rot-induced damage was validated in HL-1 cells. Nuclear phosphorylation protein expression of signal transducer and activator of transcription 3 (STAT3) was significantly increased by 7,8-DHF. The present study suggests that 7,8-DHF rescues Rot-induced cytotoxicity by inhibiting mitochondrial dysfunction and promoting nuclear translocation of p-STAT3 in cardiomyocytes, thus nominating 7,8-DHF as a new pharmacological candidate agent against mitochondrial dysfunction in cardiac diseases.
Collapse
Affiliation(s)
- Peng-Zhou Hang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Feng-Qin Ge
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Man-Ru Zhang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qi-Hang Li
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Hua-Qing Yu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yu-Chen Song
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Dan-Dan Guo
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Jing Zhao
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| | - Hua Zhu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
12
|
Rehman MU, Sehar N, Dar NJ, Khan A, Arafah A, Rashid S, Rashid SM, Ganaie MA. Mitochondrial dysfunctions, oxidative stress and neuroinflammation as therapeutic targets for neurodegenerative diseases: An update on current advances and impediments. Neurosci Biobehav Rev 2023; 144:104961. [PMID: 36395982 DOI: 10.1016/j.neubiorev.2022.104961] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer disease (AD), Parkinson disease (PD), and Huntington disease (HD) represent a major socio-economic challenge in view of their high prevalence yet poor treatment outcomes affecting quality of life. The major challenge in drug development for these NDs is insufficient clarity about the mechanisms involved in pathogenesis and pathophysiology. Mitochondrial dysfunction, oxidative stress and inflammation are common pathways that are linked to neuronal abnormalities and initiation of these diseases. Thus, elucidating the shared initial molecular and cellular mechanisms is crucial for recognizing novel remedial targets, and developing therapeutics to impede or stop disease progression. In this context, use of multifunctional compounds at early stages of disease development unclogs new avenues as it acts on act on multiple targets in comparison to single target concept. In this review, we summarize overview of the major findings and advancements in recent years focusing on shared mechanisms for better understanding might become beneficial in searching more potent pharmacological interventions thereby reducing the onset or severity of various NDs.
Collapse
Affiliation(s)
- Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Nawab John Dar
- School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78992 USA
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Srinagar, Jammu and Kashmir, India
| | - Majid Ahmad Ganaie
- Department of Pharmacology & Toxicology, College of Dentistry and Pharmacy, Buraydah Colleges, Buraydah, Saudi Arabia
| |
Collapse
|
13
|
Liu L, Su R, Huang P, Li X, Xiong J, Xiao Y, Mao D, Liu L. Case Report: Evidences of myasthenia and cerebellar atrophy in a chinese patient with novel compound heterozygous MSTO1 variants. Front Genet 2022; 13:947886. [PMID: 36035138 PMCID: PMC9402982 DOI: 10.3389/fgene.2022.947886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022] Open
Abstract
Misato Mitochondrial Distribution and Morphology Regulator 1 (MSTO1) is a soluble cytoplasmic protein that regulates mitochondrial dynamics by promoting mitochondrial fusion. Variants in the MSTO1 gene cause a rare disease characterized by early-onset myopathy and cerebellar ataxia, with almost 30 cases reported worldwide. Here we report a case of a 3-year-old boy with novel heterozygous variants of the MSTO1 gene (c.1A>G (p.M1?) and c.727G>C(p.Ala243Pro)). Sequencing data and subsequent validation show that the two variants were inherited from the mother and father of the patient (both were heterozygous). The clinical features are infancy-onset mental and motor retardation, language disorder, dysarthria, scoliosis, cerebellar atrophy, tremor, lower-extremity muscle weakness, elevated muscle enzymes, extensive myopathy with chronic atrophy, hyperventilation lungs, and previously unreported hairy back and enlarged gastrocnemius. Finally, novel heterozygous MSTO1 variants were discovered in this case, which expands the gene spectrum and clinical phenotype of this type of disease, and provides a new direction for future treatment and research. Then we summarize the mutational spectrum, pathological, clinical features and imaging of MSTO1 variants in a cohort of reported 31 patients and discuss the pathogenesis of MSTO1 in humans.
Collapse
Affiliation(s)
- Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ruiting Su
- Department of Clinical Medicine, Xiangya Medical College, Central South University, Changsha, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xingfang Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Xiong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Dingan Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatric Neurology, Patientren’s Medical Center, Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Lingjuan Liu,
| |
Collapse
|
14
|
Burtscher J, Romani M, Bernardo G, Popa T, Ziviani E, Hummel FC, Sorrentino V, Millet GP. Boosting mitochondrial health to counteract neurodegeneration. Prog Neurobiol 2022; 215:102289. [DOI: 10.1016/j.pneurobio.2022.102289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 12/22/2022]
|
15
|
Wang B, Yan Y, Ding CF. Metal-organic framework-based sample preparation in proteomics. J Chromatogr A 2022; 1671:462971. [DOI: 10.1016/j.chroma.2022.462971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 01/05/2023]
|