1
|
Yu H, Liang J, Bao Y, Chen K, Jin Y, Li X, Chen H, Gou Y, Lu K, Lin Z. Ginkgolide A enhances FoxO1 expression and reduces endoplasmic reticulum stress to mitigate osteoarthritis in mice. Int Immunopharmacol 2024; 142:113116. [PMID: 39288630 DOI: 10.1016/j.intimp.2024.113116] [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: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
This study aimed to investigate the effects of Ginkgolide A (GA) on chondrocytes under oxidative stress and to elucidate its potential molecular mechanisms. Using a destabilization of the medial meniscus (DMM) model in mice and an in vitro osteoarthritis (OA) model induced by tert-butyl hydroperoxide (TBHP) in chondrocytes, we validated the therapeutic efficacy and underlying mechanisms of GA. Potential OA targets of GA were identified through network pharmacology, Gene Ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Further exploration into the effects on endoplasmic reticulum stress (ERS), apoptosis, extracellular matrix (ECM) degradation, and Forkhead Box O1 (FoxO1) related pathways was conducted using Western blotting, immunofluorescence, TUNEL staining, flow cytometry, X-ray, micro-computed tomography (Micro-CT) analysis, and histological staining. The results demonstrated that GA upregulated FoxO1 expression and inhibited ERS-related signaling pathways, thereby reducing apoptosis and ECM degradation. In conclusion, GA significantly alleviated OA symptoms both in vitro and in vivo, suggesting its potential as a therapeutic agent for OA.
Collapse
Affiliation(s)
- Heng Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jinghao Liang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yingying Bao
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Kaiye Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yangcan Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiang Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hao Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yong Gou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Keyu Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhongke Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
2
|
Diniz LP, Araujo APB, Carvalho CF, Matias I, de Sá Hayashide L, Marques M, Pessoa B, Andrade CBV, Vargas G, Queiroz DD, de Carvalho JJ, Galina A, Gomes FCA. Accumulation of damaged mitochondria in aging astrocytes due to mitophagy dysfunction: Implications for susceptibility to mitochondrial stress. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167470. [PMID: 39153665 DOI: 10.1016/j.bbadis.2024.167470] [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: 05/03/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Aging disrupts brain function, leading to cognitive decline and neurodegenerative diseases. Senescent astrocytes, a hallmark of aging, contribute to this process through unknown mechanisms. This study investigates how senescence impacts astrocytic mitochondrial dynamics, which are critical for brain health. Our research, conducted using aged mouse brains, represents the first evidence of morphologically damaged mitochondria in astrocytes, along with functional alterations in mitochondrial respiration. In vitro experiments revealed that senescent astrocytes exhibit an increase in mitochondrial fragmentation and impaired mitophagy. Concurrently, there was an upregulation of mitochondrial biogenesis, indicating a compensatory response to mitochondrial damage. Importantly, these senescent astrocytes were more susceptible to mitochondrial stress, a vulnerability reversed by rapamycin treatment. These findings suggest a potential link between senescence, impaired mitochondrial quality control, and increased susceptibility to mitochondrial stress in astrocytes. Overall, our study highlights the importance of addressing mitochondrial dysfunction and senescence-related changes in astrocytes as a promising approach for developing therapies to counter age-related neurodegeneration and improve brain health.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Ana Paula Bergamo Araujo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clara Fernandes Carvalho
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isadora Matias
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lívia de Sá Hayashide
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Marques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Pessoa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cherley Borba Vieira Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Departamento de Histologia e Embriologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriele Vargas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniela Dias Queiroz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge José de Carvalho
- Departamento de Histologia e Embriologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Galina
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
3
|
Kalykaki M, Rubio-Tomás T, Tavernarakis N. The role of mitochondria in cytokine and chemokine signalling during ageing. Mech Ageing Dev 2024; 222:111993. [PMID: 39307464 DOI: 10.1016/j.mad.2024.111993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Ageing is accompanied by a persistent, low-level inflammation, termed "inflammageing", which contributes to the pathogenesis of age-related diseases. Mitochondria fulfil multiple roles in host immune responses, while mitochondrial dysfunction, a hallmark of ageing, has been shown to promote chronic inflammatory states by regulating the production of cytokines and chemokines. In this review, we aim to disentangle the molecular mechanisms underlying this process. We describe the role of mitochondrial signalling components such as mitochondrial DNA, mitochondrial RNA, N-formylated peptides, ROS, cardiolipin, cytochrome c, mitochondrial metabolites, potassium efflux and mitochondrial calcium in the age-related immune system activation. Furthermore, we discuss the effect of age-related decline in mitochondrial quality control mechanisms, including mitochondrial biogenesis, dynamics, mitophagy and UPRmt, in inflammatory states upon ageing. In addition, we focus on the dynamic relationship between mitochondrial dysfunction and cellular senescence and its role in regulating the secretion of pro-inflammatory molecules by senescent cells. Finally, we review the existing literature regarding mitochondrial dysfunction and inflammation in specific age-related pathological conditions, including neurodegenerative diseases (Alzheimer's and Parkinson's disease, and amyotrophic lateral sclerosis), osteoarthritis and sarcopenia.
Collapse
Affiliation(s)
- Maria Kalykaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Crete GR-70013, Greece
| | - Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Crete GR-70013, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Crete GR-70013, Greece; Division of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete GR-71003, Greece.
| |
Collapse
|
4
|
Zuo X, Gao L, Peng X, Dong L, Huang M, Hu T, Deng L, Zhu Q, Zhang J. Unveiling the role of mtDNA in Liver-Kidney Crosstalk: Insights from trichloroethylene hypersensitivity syndrome. Int Immunopharmacol 2024; 138:112513. [PMID: 38917520 DOI: 10.1016/j.intimp.2024.112513] [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: 05/15/2024] [Revised: 06/16/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024]
Abstract
In specific pathological conditions, addressing liver injury may yield favorable effects on renal function through the phenomenon of liver-kidney crosstalk. Mitochondrial DNA (mtDNA) possesses the capability to trigger downstream pathways of inflammatory cytokines, ultimately leading to immune-mediated organ damage. Consequently, understanding the intricate molecular mechanisms governing mtDNA involvement in diseases characterized by liver-kidney crosstalk is of paramount significance. This study seeks to elucidate the role of mtDNA in conditions marked by liver-kidney crosstalk. In previous clinical cases, it has been observed that patients with Trichloroethylene Hypersensitivity Syndrome (TCE-HS) who experience severe liver injury often also exhibit renal injury. In this study, patients diagnosed with trichloroethylene hypersensitivity syndrome were recruited from Shenzhen Occupational Disease Control Center. And Balb/c mice were treated with trichloroethylene. The correlation between liver and kidney injuries in patients with TCE-HS was assessed using Enzyme-Linked Immunosorbent Assay (ELISA). Alterations in mtDNA levels were examined in mouse hepatocytes, red blood cells (RBCs), and renal tubular epithelial cells utilizing immunofluorescence and PCR techniques. TCE-sensitized mice exhibited a significant increase in reactive oxygen species (ROS) and the opening of the mitochondrial permeability transition pore in hepatocytes, resulting in the release of mtDNA. Furthermore, heightened levels of mtDNA and Toll-like Receptor 9 (TLR9) expression were observed in RBCs. Additional experiments demonstrated elevated expression of TLR9 and its downstream mediator MyD88 in renal tubule epithelial cells of TCE-sensitized mice. In vitro investigations confirmed that mtDNA activates the TLR9 pathway in TCMK-1 cells. Collectively, these results suggest that mtDNA released from mitochondrial damage in hepatocytes is carried by RBCs to renal tubular epithelial cells and mediates inflammatory injury in renal tubular epithelial cells through activation of the TLR9 receptor.
Collapse
Affiliation(s)
- Xulei Zuo
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Lei Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Xinyu Peng
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Luolun Dong
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Meng Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Tingting Hu
- Shenzhen Prevention and Treatment Center for Occupational Disease, Shenzhen, PR China
| | - Lihua Deng
- Shenzhen Prevention and Treatment Center for Occupational Disease, Shenzhen, PR China.
| | - Qixing Zhu
- Institute of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| | - Jiaxiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| |
Collapse
|
5
|
Lin Y, Huang Y, Yang B, Zhang Y, Ji N, Li J, Zhou Y, Shen YQ, Chen Q. Precision therapy targeting CAMK2 to overcome resistance to EGFR inhibitors in FAT1-mutated oral squamous cell carcinoma. Chin Med J (Engl) 2024:00029330-990000000-01204. [PMID: 39227322 DOI: 10.1097/cm9.0000000000003217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a prevalent type of cancer with a high mortality rate in its late stages. One of the major challenges in OSCC treatment is the resistance to epidermal growth factor receptor (EGFR) inhibitors. Therefore, it is imperative to elucidate the mechanism underlying drug resistance and develop appropriate precision therapy strategies to enhance clinical efficacy. METHODS To evaluate the efficacy of the combination of the Ca2+/calmodulin-dependent protein kinase II (CAMK2) inhibitor KN93 and EGFR inhibitors, we performed in vitro and in vivo experiments using two FAT atypical cadherin 1 (FAT1)-deficient (SCC9 and SCC25) and two FAT1 wild-type (SCC47 and HN12) OSCC cell lines. We assessed the effects of EGFR inhibitors (afatinib or cetuximab), KN93, or their combination on the malignant phenotype of OSCC in vivo and in vitro. The alterations in protein expression levels of members of the EGFR signaling pathway and SRY-box transcription factor 2 (SOX2) were analyzed. Changes in the yes-associated protein 1 (YAP1) protein were characterized. Moreover, we analyzed mitochondrial dysfunction. Besides, the effects of combination therapy on mitochondrial dynamics were also evaluated. RESULTS OSCC with FAT1 mutations exhibited resistance to EGFR inhibitors treatment. The combination of KN93 and EGFR inhibitors significantly inhibited the proliferation, survival, and migration of FAT1-mutated OSCC cells and suppressed tumor growth in vivo. Mechanistically, combination therapy enhanced the therapeutic sensitivity of FAT1-mutated OSCC cells to EGFR inhibitors by modulating the EGFR pathway and downregulated tumor stemness-related proteins. Furthermore, combination therapy induced reactive oxygen species (ROS)-mediated mitochondrial dysfunction and disrupted mitochondrial dynamics, ultimately resulting in tumor suppression. CONCLUSION Combination therapy with EGFR inhibitors and KN93 could be a novel precision therapeutic strategy and a potential clinical solution for EGFR-resistant OSCC patients with FAT1 mutations.
Collapse
Affiliation(s)
- Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Scalise S, Zannino C, Lucchino V, Lo Conte M, Abbonante V, Benedetto GL, Scalise M, Gambardella A, Parrotta EI, Cuda G. Ascorbic acid mitigates the impact of oxidative stress in a human model of febrile seizure and mesial temporal lobe epilepsy. Sci Rep 2024; 14:5941. [PMID: 38467734 PMCID: PMC10928078 DOI: 10.1038/s41598-024-56680-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/09/2024] [Indexed: 03/13/2024] Open
Abstract
Prolonged febrile seizures (FS) in children are linked to the development of temporal lobe epilepsy (MTLE). The association between these two pathologies may be ascribed to the long-term effects that FS exert on neural stem cells, negatively affecting the generation of new neurons. Among the insults associated with FS, oxidative stress is noteworthy. Here, we investigated the consequences of exposure to hydrogen peroxide (H2O2) in an induced pluripotent stem cell-derived neural stem cells (iNSCs) model of a patient affected by FS and MTLE. In our study, we compare the findings from the MTLE patient with those derived from iNSCs of a sibling exhibiting a milder phenotype defined only by FS, as well as a healthy individual. In response to H2O2 treatment, iNSCs derived from MTLE patients demonstrated an elevated production of reactive oxygen species and increased apoptosis, despite the higher expression levels of antioxidant genes and proteins compared to other cell lines analysed. Among the potential causative mechanisms of enhanced vulnerability of MTLE patient iNSCs to oxidative stress, we found that these cells express low levels of the heat shock protein HSPB1 and of the autophagy adaptor SQSTM1/p62. Pre-treatment of diseased iNSCs with the antioxidant molecule ascorbic acid restored HSBP1 and p62 expression and simultaneously reduced the levels of ROS and apoptosis. Our findings suggest the potential for rescuing the impaired oxidative stress response in diseased iNSCs through antioxidant treatment, offering a promising mechanism to prevent FS degeneration in MTLE.
Collapse
Affiliation(s)
- Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Vittorio Abbonante
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Giorgia Lucia Benedetto
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Antonio Gambardella
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy.
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
7
|
Joshi CS, Salazar AM, Wang C, Ligon MM, Chappidi RR, Fashemi BE, Felder PA, Mora A, Grimm SL, Coarfa C, Mysorekar IU. D-Mannose reduces cellular senescence and NLRP3/GasderminD/IL-1β-driven pyroptotic uroepithelial cell shedding in the murine bladder. Dev Cell 2024; 59:33-47.e5. [PMID: 38101412 PMCID: PMC10872954 DOI: 10.1016/j.devcel.2023.11.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 07/24/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
Aging is a risk factor for disease via increased susceptibility to infection, decreased ability to maintain homeostasis, inefficiency in combating stress, and decreased regenerative capacity. Multiple diseases, including urinary tract infection (UTI), are more prevalent with age; however, the mechanisms underlying the impact of aging on the urinary tract mucosa and the correlation between aging and disease remain poorly understood. Here, we show that, relative to young (8-12 weeks) mice, the urothelium of aged (18-24 months) female mice accumulates large lysosomes with reduced acid phosphatase activity and decreased overall autophagic flux in the aged urothelium, indicative of compromised cellular homeostasis. Aged bladders also exhibit basal accumulation of reactive oxygen species (ROS) and a dampened redox response, implying heightened oxidative stress. Furthermore, we identify a canonical senescence-associated secretory phenotype (SASP) in the aged urothelium, along with continuous NLRP3-inflammasome- and Gasdermin-D-dependent pyroptotic cell death. Consequently, aged mice chronically exfoliate urothelial cells, further exacerbating age-related urothelial dysfunction. Upon infection with uropathogenic E. coli, aged mice harbor increased bacterial reservoirs and are more prone to spontaneous recurrent UTI. Finally, we discover that treatment with D-mannose, a natural bioactive monosaccharide, rescues autophagy flux, reverses the SASP, and mitigates ROS and NLRP3/Gasdermin/interleukin (IL)-1β-driven pyroptotic epithelial cell shedding in aged mice. Collectively, our results demonstrate that normal aging affects bladder physiology, with aging alone increasing baseline cellular stress and susceptibility to infection, and suggest that mannose supplementation could serve as a senotherapeutic to counter age-associated urothelial dysfunction.
Collapse
Affiliation(s)
- Chetanchandra S Joshi
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Arnold M Salazar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Caihong Wang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marianne M Ligon
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rayvanth R Chappidi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bisiayo E Fashemi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul A Felder
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amy Mora
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sandra L Grimm
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center of Aging, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Chen Q, Zhou D, Wang C, Ye M, Jia Y, Liu B, Bukulmez O, Norman RJ, Hu H, Yeung SB, Teng X, Liu W, Chen M. The adverse effects of vitrification on mouse embryo development and metabolic phenotype in offspring. FASEB J 2024; 38:e23372. [PMID: 38102977 DOI: 10.1096/fj.202301774rr] [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: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
Embryo vitrification is a standard procedure in assisted reproductive technology. Previous studies have shown that frozen embryo transfer is associated with an elevated risk of adverse maternal and neonatal outcomes. This study aimed to explore the effects of mouse blastocyst vitrification on the phenotype of vitrified-warmed blastocysts, their intrauterine and postnatal development, and the long-term metabolic health of the derived offspring. The vitrified-warmed blastocysts (IVF + VT group) exhibited reduced mitochondrial activity, increased apoptotic levels, and decreased cell numbers when compared to the fresh blastocysts (IVF group). Implantation rates, live pup rates, and crown-rump length at E18.5 were not different between the two groups. However, there was a significant decrease in fetal weight and fetal/placental weight ratio in the IVF + VT group. Furthermore, the offspring of the IVF + VT group at an age of 36 weeks had reduced whole energy consumption, impaired glucose and lipid metabolism when compared with the IVF group. Notably, RNA-seq results unveiled disturbed hepatic gene expression in the offspring from vitrified-warmed blastocysts. This study revealed the short-term negative impacts of vitrification on embryo and fetal development and the long-term influence on glucose and lipid metabolism that persist from the prenatal stage into adulthood in mice.
Collapse
Affiliation(s)
- Qiaoyu Chen
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Dan Zhou
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Changxin Wang
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingming Ye
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanping Jia
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Binya Liu
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Orhan Bukulmez
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert J Norman
- Robinson Research Institute, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Hanxin Hu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shu-Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, the University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Xiaoming Teng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenqiang Liu
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Miaoxin Chen
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Robledo E, Benito Rodriguez PG, Vega IA, Colombo MI, Aguilera MO. Staphylococcus aureus phagocytosis is affected by senescence. FRONTIERS IN AGING 2023; 4:1198241. [PMID: 37584054 PMCID: PMC10423838 DOI: 10.3389/fragi.2023.1198241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/03/2023] [Indexed: 08/17/2023]
Abstract
Senescent cells accumulate in multicellular animals with aging, resulting in organ or tissue dysfunction. These alterations increase the incidence of a variety of illnesses, including infectious diseases, and, in certain instances, its severity. In search of a rationale for this phenomenon, we focused on the endophagocytic pathway in senescent cells. We first described the endocytic vesicle populations at different stages of maturation using confocal microscopy. There was an increase in the number of vacuoles per cell, which was partially explained by an increase in cell size. No changes in vesicle maturation or degradation capacities were determined by microscopy or Western blot assays. Also, we studied the internalization of various endophagocytic cargoes in senescent cells and observed only a decrease in the intracellular recovery of bacteria such as Staphylococcus aureus. Afterwards, we studied the intracellular traffic of S. aureus, and observed no differences in the infection between control and senescent cells. In addition we quantified the recovery of bacteria from control and senescent cells infected in the presence of several inhibitors of endophagosomal maturation, and no changes were observed. These results suggest that bacterial internalization is affected in senescent cells. Indeed, we confirmed this hypothesis by determining minor bacterial adherence and internalization by confocal microscopy. Furthermore, it is important to highlight that we found very similar results with cells from aged animals, specifically BMDMs. This alteration in senescent cells enlightens the diminished bacterial clearance and may be a factor that increases the propensity to suffer severe infectious conditions in the elderly.
Collapse
Affiliation(s)
- Esteban Robledo
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Paula Guadalupe Benito Rodriguez
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
| | - Israel Aníbal Vega
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Isabel Colombo
- Instituto de Histología y Embriología (IHEM) “Dr. Mario H. Burgos” CONICET, Universidad Nacional de Cuyo Mendoza, Mendoza, Argentina
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Milton Osmar Aguilera
- Departamento Bases Científicas en Salud-Facultad de Ciencias Médicas, Facultad de Medicina, Biología Celular y Molecular, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Odontología, Microbiología, Parasitología e Inmunología, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
10
|
Li M, Yu Y, Xue K, Li J, Son G, Wang J, Qian W, Wang S, Zheng J, Yang C, Ge J. Genistein mitigates senescence of bone marrow mesenchymal stem cells via ERRα-mediated mitochondrial biogenesis and mitophagy in ovariectomized rats. Redox Biol 2023; 61:102649. [PMID: 36871183 PMCID: PMC9995482 DOI: 10.1016/j.redox.2023.102649] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023] Open
Abstract
Senescence of bone marrow mesenchymal stem cells (BMMSCs) induced by chronic oxidative stress is an important factor contributes to the postmenopausal osteoporosis (PMOP). Mitochondrial quality control takes a pivotal role in regulating oxidative stress and cell senescence. Genistein is a major isoflavone in soy products, which is best known for its ability to inhibit bone loss in both postmenopausal women and ovariectomized (OVX) rodents. Here we show that OVX-BMMSCs displayed premature senescence, elevated reactive oxygen species (ROS) level and mitochondria dysfunction, while genistein rescued these phenotypes. Using network pharmacology and molecular docking, we identified estrogen-related receptor α (ERRα) as the potential target of genistein. Knockdown of ERRα greatly abolished the anti-senescence effect of genistein on OVX-BMMSCs. Further, the mitochondrial biogenesis and mitophagy induced by genistein were inhibited by ERRα knockdown in OVX-BMMSCs. In vivo, genistein inhibited trabecular bone loss and p16INK4a expression, upregulated sirtuin 3 (SIRT3) and peroxisome proliferator-activated receptor gamma coactivator one alpha (PGC1α) expression in the trabecular bone area of proximal tibia in OVX rats. Together, this study revealed that genistein ameliorates senescence of OVX-BMMSCs through ERRα-mediated mitochondrial biogenesis and mitophagy, which provided a molecular basis for advancement and development of therapeutic strategies against PMOP.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yejia Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ke Xue
- Department of Pastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayi Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Geehun Son
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiajia Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wentao Qian
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shaoyi Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiawei Zheng
- Department of Oromaxillofacial Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China.
| | - Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
11
|
Chang CF, Gunawan AL, Liparulo I, Zushin PJH, Bertholet AM, Kirichok Y, Stahl A. CoQ Regulates Brown Adipose Tissue Respiration and Uncoupling Protein 1 Expression. Antioxidants (Basel) 2022; 12:14. [PMID: 36670876 PMCID: PMC9854525 DOI: 10.3390/antiox12010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Coenzyme Q (CoQ, aka ubiquinone) is a key component of the mitochondrial electron transport chain (ETC) and membrane-incorporated antioxidant. CoQ10 deficiencies encompass a heterogeneous spectrum of clinical phenotypes and can be caused by hereditary mutations in the biosynthesis pathway or result from pharmacological interventions such as HMG-CoA Reductase inhibitors, and statins, which are widely used to treat hypercholesterolemia and prevent cardiovascular disease. How CoQ deficiency affects individual tissues and cell types, particularly mitochondrial-rich ones such as brown adipose tissue (BAT), has remained poorly understood. Here we show that pharmacological and genetic models of BAT CoQ deficiency show altered respiration that can only in part be explained by classical roles of CoQ in the respiration chain. Instead, we found that CoQ strongly impacts brown and beige adipocyte respiration via the regulation of uncoupling protein 1 (UCP1) expression. CoQ deficiency in BAT robustly decreases UCP1 protein levels and uncoupled respiration unexpectedly, resulting in increased inner mitochondrial membrane potential and decreased ADP/ATP ratios. Suppressed UCP1 expression was also observed in a BAT-specific in vivo model of CoQ deficiency and resulted in enhanced cold sensitivity. These findings demonstrate an as yet unappreciated role of CoQ in the transcriptional regulation of key thermogenic genes and functions.
Collapse
Affiliation(s)
- Ching-Fang Chang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Amanda L. Gunawan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Irene Liparulo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Peter-James H. Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Ambre M. Bertholet
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuriy Kirichok
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
12
|
Langeh U, Kumar V, Kumar A, Kumar P, Singh C, Singh A. Cellular and mitochondrial quality control mechanisms in maintaining homeostasis in ageing. Rejuvenation Res 2022; 25:208-222. [PMID: 35850516 DOI: 10.1089/rej.2022.0027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aging is a natural process in all living organisms defined as destruction of cell function as a result of long-term accumulation of damages. Autophagy is a cellular house safeguard pathway which responsible for degrading damaged cellular organelles. Moreover, it maintains cellular homeostasis, control lifetime, and longevity. Damaged mitochondrial accumulation is a characteristic of aging which associated with neurodegeneration. Mitochondria functions as a principal energy source via supplying ATP through oxidative phosphorylation which serves as fuel for neuronal function. Mitophagy and mitochondrial specific autophagy plays an important role in maintenance of neuronal health via the removal of dysfunctional and aged mitochondria. The mitochondrial QC system involves different strategies for protecting against mitochondrial dysfunction and maintaining healthy mitochondria in cells. Mitochondrial function protection could be a strategy for the promotion of neuroprotection. Mitophagy, could be an effective target for drug discovery. Therefore, further detailed studies for mechanism of mitophagy will advance our mitochondrial phenotype knowledge and understanding to disease pathogenesis. This review mainly focuses on ageing mediated mechanism of autophagy and mitophagy for maintaining the cellular homeostasis and longevity.
Collapse
Affiliation(s)
- Urvashi Langeh
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | - Vishal Kumar
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | | | - Pradeep Kumar
- University of the Witwatersrand, 37707, Department of Pharmacy and Pharmacology, Johannesburg-Braamfontein, Gauteng, South Africa;
| | - Charan Singh
- ISF College of Pharmacy, 75126, Pharmacology, Moga, Punjab, India;
| | - Arti Singh
- ISF College of Pharmacy, 75126, Pharmacology, ISF College of Pharmacy, Department of Pharmacology, Moga, Moga, Punjab, India, 142001;
| |
Collapse
|
13
|
Zhang Q, Fu H, Gong W, Cao F, Wu T, Hu F. Plumbagin protects H9c2 cardiomyocytes against TBHP‑induced cytotoxicity by alleviating ROS‑induced apoptosis and modulating autophagy. Exp Ther Med 2022; 24:501. [PMID: 35837065 DOI: 10.3892/etm.2022.11428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/24/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Qianrui Zhang
- Department of Pharmacy, General Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, P.R. China
| | - Haitan Fu
- Department of Pharmacy, General Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, P.R. China
| | - Wenjuan Gong
- Department of Pharmacy, General Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, P.R. China
| | - Feng Cao
- Department of Pharmacy, General Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, P.R. China
| | - Tao Wu
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, Hubei 430000, P.R. China
| | - Fei Hu
- Department of Neurosurgery, General Hospital of the Yangtze River Shipping, Wuhan Brain Hospital, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
14
|
Mitochondrial protein import determines lifespan through metabolic reprogramming and de novo serine biosynthesis. Nat Commun 2022; 13:651. [PMID: 35115503 PMCID: PMC8814026 DOI: 10.1038/s41467-022-28272-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Sustained mitochondrial fitness relies on coordinated biogenesis and clearance. Both processes are regulated by constant targeting of proteins into the organelle. Thus, mitochondrial protein import sets the pace for mitochondrial abundance and function. However, our understanding of mitochondrial protein translocation as a regulator of longevity remains enigmatic. Here, we targeted the main protein import translocases and assessed their contribution to mitochondrial abundance and organismal physiology. We find that reduction in cellular mitochondrial load through mitochondrial protein import system suppression, referred to as MitoMISS, elicits a distinct longevity paradigm. We show that MitoMISS triggers the mitochondrial unfolded protein response, orchestrating an adaptive reprogramming of metabolism. Glycolysis and de novo serine biosynthesis are causatively linked to longevity, whilst mitochondrial chaperone induction is dispensable for lifespan extension. Our findings extent the pro-longevity role of UPRmt and provide insight, relevant to the metabolic alterations that promote or undermine survival and longevity. Mitochondrial function is linked to lifespan. Here the authors show that inhibition of mitochondrial protein import leads to a reduction in mitochondrial abundance and extends lifespan in Caenorhabditis elegans via activation of glycolysis and de novo serine biosynthesis.
Collapse
|
15
|
Chandrasekaran A, Dittlau KS, Corsi GI, Haukedal H, Doncheva NT, Ramakrishna S, Ambardar S, Salcedo C, Schmidt SI, Zhang Y, Cirera S, Pihl M, Schmid B, Nielsen TT, Nielsen JE, Kolko M, Kobolák J, Dinnyés A, Hyttel P, Palakodeti D, Gorodkin J, Muddashetty RS, Meyer M, Aldana BI, Freude KK. Astrocytic reactivity triggered by defective autophagy and metabolic failure causes neurotoxicity in frontotemporal dementia type 3. Stem Cell Reports 2021; 16:2736-2751. [PMID: 34678206 PMCID: PMC8581052 DOI: 10.1016/j.stemcr.2021.09.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022] Open
Abstract
Frontotemporal dementia type 3 (FTD3), caused by a point mutation in the charged multivesicular body protein 2B (CHMP2B), affects mitochondrial ultrastructure and the endolysosomal pathway in neurons. To dissect the astrocyte-specific impact of mutant CHMP2B expression, we generated astrocytes from human induced pluripotent stem cells (hiPSCs) and confirmed our findings in CHMP2B mutant mice. Our data provide mechanistic insights into how defective autophagy causes perturbed mitochondrial dynamics with impaired glycolysis, increased reactive oxygen species, and elongated mitochondrial morphology, indicating increased mitochondrial fusion in FTD3 astrocytes. This shift in astrocyte homeostasis triggers a reactive astrocyte phenotype and increased release of toxic cytokines, which accumulate in nuclear factor kappa b (NF-κB) pathway activation with increased production of CHF, LCN2, and C3 causing neurodegeneration. FTD3 iPSC-derived astrocytes display impaired autophagy Impaired autophagy affects mitochondria turnover, glucose hypometabolism and TCA cycle FTD3 astrocytes contribute to reactive gliosis by increased C3, LCN2, IL6, and IL8 Reactive astrocyte phenotypes are present in both in vitro and in vivo models
Collapse
Affiliation(s)
- Abinaya Chandrasekaran
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Katarina Stoklund Dittlau
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Giulia I Corsi
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark; Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg 1871, Denmark
| | - Henriette Haukedal
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Nadezhda T Doncheva
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark; Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg 1871, Denmark; Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sarayu Ramakrishna
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore 560065, India; The University of Trans-Disciplinary Health Sciences and Technology, Bangalore 560064, India
| | - Sheetal Ambardar
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore 560065, India; National Center for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Claudia Salcedo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sissel I Schmidt
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Yu Zhang
- Department of Experimental Medical Science, Wallenberg Center for Molecular Medicine and Lund Stem Cell Center, Lund University, Lund 22184, Sweden
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Maria Pihl
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | | | - Troels Tolstrup Nielsen
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen E Nielsen
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen 2100, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | | | | | - Poul Hyttel
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Dasaradhi Palakodeti
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore 560065, India
| | - Jan Gorodkin
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark; Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg 1871, Denmark
| | - Ravi S Muddashetty
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore 560065, India
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark.
| |
Collapse
|
16
|
Thu VT, Yen NTH, Ly NTH. Liquiritin from Radix Glycyrrhizae Protects Cardiac Mitochondria from Hypoxia/Reoxygenation Damage. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2021; 2021:1857464. [PMID: 34413986 PMCID: PMC8369190 DOI: 10.1155/2021/1857464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/09/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
AIMS The purpose of this study was to evaluate the protective effect of liquiritin (LIQ) from Radix Glycyrrhizae on cardiac mitochondria against hypoxia/reoxygenation (HR) injury. METHODS H9C2 cells were subject to the HR model. LIQ purified from Radix Glycyrrhizae (purity > 95%) was administrated to reoxygenation period. Cell viability, mitochondrial mass, mitochondrial membrane potential, reactive oxygen species, and mitochondrial Ca2⁺ level were then assessed by using Cell Counting kit-8 and suitable fluorescence probe kits. RESULTS LIQ administration remarkably reduced the rate of HR damage via increasing H9C2 cell viability level and preserving mitochondria after HR. Particularly, 60 μM of LIQ posthypoxic treatment markedly reduced cell death in HR-subjected H9C2 cell groups (p < 0.05). Interestingly, posthypoxic treatment of LIQ significantly prevented the loss of mitochondrial membrane potential, the decrease in mitochondrial mass, the increase in reactive oxygen species production, and the elevation of mitochondrial Ca2⁺ level in HR-treated H9C2 cells. CONCLUSION The present study provides for the first time the cardioprotective of LIQ posthypoxic treatment via reducing H9C2 cell death and protecting cardiac mitochondria against HR damage.
Collapse
Affiliation(s)
- Vu Thi Thu
- Center for Life Science Research, Faculty of Biology, VNU University of Science, Vietnam National University, 334 Nguyen Trai, Hanoi, Vietnam
- The Key Laboratory of Enzyme and Protein Technology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Ngo Thi Hai Yen
- Center for Life Science Research, Faculty of Biology, VNU University of Science, Vietnam National University, 334 Nguyen Trai, Hanoi, Vietnam
| | | |
Collapse
|
17
|
Xi H, He Z, Lv C. FOXG1 improves mitochondrial function and promotes the progression of nasopharyngeal carcinoma. Mol Med Rep 2021; 24:651. [PMID: 34278485 PMCID: PMC8299199 DOI: 10.3892/mmr.2021.12290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Forkhead‑box gene 1 (FOXG1) has been reported to serve an important role in various malignancies, but its effects on nasopharyngeal cancer (NPC) remain unknown. Thus, the present study aimed to investigate the specific regulatory relationship between FOXG1 and NPC progression. Tumor tissues and matching para‑carcinoma tissues were obtained from patients with NPC. Small interfering (si)RNA‑FOXG1 and pcDNA3.1‑FOXG1 were transfected into SUNE‑1 and C666‑1 cells to knockdown and overexpress FOXG1 expression, respectively. FOXG1 expression was detected using reverse transcription‑quantitative PCR and immunohistochemistry. Cell proliferation was detected using MTT and 5‑ethynyl‑20‑deoxyuridine assays. Transwell invasion assay, wound healing assay and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. Western blotting was conducted to detect the expression levels of mitochondrial markers (succinate dehydrogenase complex flavoprotein subunit A, heat shock protein 60 and pyruvate dehydrogenase), epithelial‑mesenchymal transition (EMT) related proteins (N‑cadherin, Snail and E‑cadherin) and apoptosis‑related proteins [Bax, Bcl‑2, poly(ADP‑ribose) polymerase 1 (PARP), cleaved PARP, cleaved caspase‑3, cleaved caspase‑8, cleaved caspase‑9, caspase‑3, caspase‑8 and caspase‑9]. The mitochondrial membrane potential was detected via flow cytometry, while the ATP/ADP ratio was determined using the ADP/ATP ratio assay kit. The present results demonstrated that FOXG1 expression was upregulated in NPC tissues and cells, and was associated with distant metastasis and TNM stage. Moreover, knockdown of FOXG1 inhibited the proliferation, migration, invasion, EMT and mitochondrial function of SUNE‑1 cells, as well as promoted cell apoptosis, while the opposite results were observed in C666‑1 cells. In conclusion, FOXG1 enhanced proliferation, migration and invasion, induced EMT and improved mitochondrial function in NPC cells. The current findings provide an adequate theoretical basis for the treatment of NPC.
Collapse
Affiliation(s)
- Huajun Xi
- Department of Otolaryngology and Stomatology, Shouguang People's Hospital, Shouguang, Shandong 262700, P.R. China
| | - Zhengxiang He
- Department of Otolaryngology and Maxillofacial Surgery, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Cao Lv
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
18
|
Haghi A, Salemi M, Fakhimahmadi A, Mohammadi Kian M, Yousefi H, Rahmati M, Mohammadi S, Ghavamzadeh A, Moosavi MA, Nikbakht M. Effects of different autophagy inhibitors on sensitizing KG-1 and HL-60 leukemia cells to chemotherapy. IUBMB Life 2020; 73:130-145. [PMID: 33205598 DOI: 10.1002/iub.2411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/15/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
A little number of current autophagy inhibitors may have beneficial effects on the acute myeloid leukemia (AML) patients. However, there is a strong need to figure out which settings should be activated or inhibited in autophagy pathway to prevail drug resistance and also to improve current treatment options in leukemia. Therefore, this study aimed to compare the effects of well-known inhibitors of autophagy (as 3-MA, BafA1, and HCQ) in leukemia KG-1 and HL-60 cells exposed to arsenic trioxide (ATO) and/or all-trans retinoic acid (ATRA). Cell proliferation and cytotoxicity of cells were examined by MTT assay. Autophagy was studied by evaluating the development of acidic vesicular organelles, and the autophagosomes formation was investigated by acridine orange staining and transmission electron microscopy. Moreover, the gene and protein expressions levels of autophagy markers (ATGs, p62/SQSTM1, and LC-3B) were also performed by qPCR and western blotting, respectively. The rate of apoptosis and cell cycle were evaluated using flow cytometry. We compared the cytotoxic and apoptotic effects of ATO and/or ATRA in both cell lines and demonstrated that some autophagy markers upregulated in this context. Also, it was shown that autophagy blockers HCQ and/or BafA1 could potentiate the cytotoxic effects of ATO/ATRA, which were more pronounced in KG-1 cells compared to HL-60 cell line. This study showed the involvement of autophagy during the treatment of KG-1 and HL-60 cells by ATO/ATRA. This study proposed that therapy of ATO/ATRA in combination with HCQ can be considered as a more effective strategy for targeting leukemic KG-1 cells.
Collapse
Affiliation(s)
- Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdieh Salemi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aila Fakhimahmadi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mahnaz Mohammadi Kian
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, Louisiana, USA
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Mohammadi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Liu JX, Yang C, Liu ZJ, Su HY, Zhang WH, Pan Q, Liu HF. Protection of procyanidin B2 on mitochondrial dynamics in sepsis associated acute kidney injury via promoting Nrf2 nuclear translocation. Aging (Albany NY) 2020; 12:15638-15655. [PMID: 32805725 PMCID: PMC7467384 DOI: 10.18632/aging.103726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
In septic acute kidney injury (SAKI), the positive feedback between damaged mitochondria and accumulation of reactive oxygen species results in cell and tissue damage through multiple mechanisms. Removing the damaged mitochondria or neutralizing the reactive oxygen species has been considered beneficial to alleviating cell damage. The antioxidant Procyanidin B2 has been reported to inhibits reactive oxygen species and thereby reduces cell injury. However, it is unclear whether this effect is associated with clearance of damaged mitochondria. Here, we evaluated the efficacy of procyanidin B2 on SAKI, and focused on its effects on mitochondrial dynamics and removing damaged mitochondria via mitophagy. The results showed that the renal function, renal tubular cell vacuolization and oxidative stress were decreased in SAKI mice treated with procyanidin B2, moreover, skewed mitochondrial fusion/fission, mitochondrial mediated apoptosis and impaired mitophagy were improved in SAKI mice treated with procyanidin B2. In mechanism, the improvement of procyanidin B2 on mitochondrial dynamics were associated with increased nuclear translocation of the transcription factor, Nrf2. In summary, our findings highlighted that the protective efficacy of procyanidin B2 in reducing cellular damage in SAKI, and mechanisms improving mitochondrial dynamics and quality control at least in part by promoting Nrf2 translocation into the nucleus.
Collapse
Affiliation(s)
- Jian-Xing Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Ze-Jian Liu
- Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Hong-Yong Su
- Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Wei-Huang Zhang
- Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| |
Collapse
|
20
|
Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of Autophagy in Oxidative Stress. Int J Mol Sci 2020; 21:ijms21093289. [PMID: 32384691 PMCID: PMC7246723 DOI: 10.3390/ijms21093289] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a catabolic process for unnecessary or dysfunctional cytoplasmic contents by lysosomal degradation pathways. Autophagy is implicated in various biological processes such as programmed cell death, stress responses, elimination of damaged organelles and development. The role of autophagy as a crucial mediator has been clarified and expanded in the pathological response to redox signalling. Autophagy is a major sensor of the redox signalling. Reactive oxygen species (ROS) are highly reactive molecules that are generated as by-products of cellular metabolism, principally by mitochondria. Mitochondrial ROS (mROS) are beneficial or detrimental to cells depending on their concentration and location. mROS function as redox messengers in intracellular signalling at physiologically low level, whereas excessive production of mROS causes oxidative damage to cellular constituents and thus incurs cell death. Hence, the balance of autophagy-related stress adaptation and cell death is important to comprehend redox signalling-related pathogenesis. In this review, we attempt to provide an overview the basic mechanism and function of autophagy in the context of response to oxidative stress and redox signalling in pathology.
Collapse
Affiliation(s)
- Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jieun Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| |
Collapse
|
21
|
Folkerts H, Wierenga AT, van den Heuvel FA, Woldhuis RR, Kluit DS, Jaques J, Schuringa JJ, Vellenga E. Elevated VMP1 expression in acute myeloid leukemia amplifies autophagy and is protective against venetoclax-induced apoptosis. Cell Death Dis 2019; 10:421. [PMID: 31142733 PMCID: PMC6541608 DOI: 10.1038/s41419-019-1648-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Vacuole membrane protein (VMP1) is a putative autophagy protein, which together with Beclin-1 acts as a molecular switch in activating autophagy. In the present study the role of VMP1 was analysed in CD34+ cells of cord blood (CB) and primary acute myeloid leukemia (AML) cells and cell lines. An increased expression of VMP1 was observed in a subset of AML patients. Functional studies in normal CB CD34+ cells indicated that inhibiting VMP1 expression reduced autophagic-flux, coinciding with reduced expansion of hematopoietic stem and progenitor cells (HSPC), delayed differentiation, increased apoptosis and impaired in vivo engraftment. Comparable results were observed in leukemic cell lines and primary AML CD34+ cells. Ultrastructural analysis indicated that leukemic cells overexpressing VMP1 displayed a reduced number of mitochondrial structures, while the number of lysosomal degradation structures was increased. The overexpression of VMP1 did not affect cell proliferation and differentiation, but increased autophagic-flux and improved mitochondrial quality, which coincided with an increased threshold for venetoclax-induced loss of mitochondrial outer membrane permeabilization (MOMP) and apoptosis. In conclusion, our data indicate that in leukemic cells high VMP1 is involved with mitochondrial quality control.
Collapse
Affiliation(s)
- Hendrik Folkerts
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albertus T Wierenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Fiona A van den Heuvel
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Roy R Woldhuis
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Darlyne S Kluit
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jennifer Jaques
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Jacob Schuringa
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
22
|
Ge C, An N, Li L, Wei W, Ji L, Yuan N, Fang Y, Xu L, Song L, Zhang J, Song C, Wang J, Zhang S. Autophagy-deficient mice are more susceptible to engrafted leukemogenesis. Blood Cells Mol Dis 2019; 77:129-136. [PMID: 31059942 DOI: 10.1016/j.bcmd.2019.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 04/27/2019] [Indexed: 12/14/2022]
Abstract
Autophagy is primarily considered as an important survival mechanism for both normal cells and cancer cells in response to metabolic stress or chemotherapy; but the role of autophagy in leukemogenesis is not fully understood. The aim of this study is to explore the role of intrinsic autophagy in the leukemogenesis of B-cell acute lymphoblastic leukemia (B-ALL). In this study, conditional knockout mice Atg7f/f;Ubc-Cre, in which an autophagy-essential gene Atg7 is universally deleted, were used as recipients, B-ALL cell line 697 was used as donor cells to generate leukemia mouse model. Compared to wild-type mice, Atg7 knockout mice were more susceptible to engrafted leukemogenesis, shown by increase in white blood cells, lymphocytes, and platelets, decrease in HSPC number and its colony-forming unit (CFU). The liver and spleen displayed hepatosplenomegaly and inflammatory cell infiltration. Furthermore, second competitive transplantation revealed dysfunction of the HSPC in Atg7-knockout leukemia mice represented by destructive self-renew ability (CFU) and reconstitution ability including decreased B220, Ter 119 cells, and increased Gr-1 cell percentage. In summary, Mice with universal deletion of Atg7 are more inclined to the occurrence of engrafted human leukemia, which is largely attributed to the deterioration of the function of HSPC in autophagy deficient mice.
Collapse
Affiliation(s)
- Chaorong Ge
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Ni An
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Lei Li
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Wen Wei
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Li Ji
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Na Yuan
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Yixuan Fang
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Li Xu
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Lin Song
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Jingyi Zhang
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Chenglin Song
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China
| | - Jianrong Wang
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China.
| | - Suping Zhang
- Hematology Center of Cyrus Tang Medical Institute, Jiangsu Institute of Hematology, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, State Key Laboratory of Radiation Medicine and Radioprotection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
23
|
Mazumder S, De R, Debsharma S, Bindu S, Maity P, Sarkar S, Saha SJ, Siddiqui AA, Banerjee C, Nag S, Saha D, Pramanik S, Mitra K, Bandyopadhyay U. Indomethacin impairs mitochondrial dynamics by activating the PKCζ-p38-DRP1 pathway and inducing apoptosis in gastric cancer and normal mucosal cells. J Biol Chem 2019; 294:8238-8258. [PMID: 30940726 DOI: 10.1074/jbc.ra118.004415] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
The subcellular mechanism by which nonsteroidal anti-inflammatory drugs (NSAIDs) induce apoptosis in gastric cancer and normal mucosal cells is elusive because of the diverse cyclooxygenase-independent effects of these drugs. Using human gastric carcinoma cells (AGSs) and a rat gastric injury model, here we report that the NSAID indomethacin activates the protein kinase Cζ (PKCζ)-p38 MAPK (p38)-dynamin-related protein 1 (DRP1) pathway and thereby disrupts the physiological balance of mitochondrial dynamics by promoting mitochondrial hyper-fission and dysfunction leading to apoptosis. Notably, DRP1 knockdown or SB203580-induced p38 inhibition reduced indomethacin-induced damage to AGSs. Indomethacin impaired mitochondrial dynamics by promoting fissogenic activation and mitochondrial recruitment of DRP1 and down-regulating fusogenic optic atrophy 1 (OPA1) and mitofusins in rat gastric mucosa. Consistent with OPA1 maintaining cristae architecture, its down-regulation resulted in EM-detectable cristae deformity. Deregulated mitochondrial dynamics resulting in defective mitochondria were evident from enhanced Parkin expression and mitochondrial proteome ubiquitination. Indomethacin ultimately induced mitochondrial metabolic and bioenergetic crises in the rat stomach, indicated by compromised fatty acid oxidation, reduced complex I- associated electron transport chain activity, and ATP depletion. Interestingly, Mdivi-1, a fission-preventing mito-protective drug, reversed indomethacin-induced DRP1 phosphorylation on Ser-616, mitochondrial proteome ubiquitination, and mitochondrial metabolic crisis. Mdivi-1 also prevented indomethacin-induced mitochondrial macromolecular damage, caspase activation, mucosal inflammation, and gastric mucosal injury. Our results identify mitochondrial hyper-fission as a critical and common subcellular event triggered by indomethacin that promotes apoptosis in both gastric cancer and normal mucosal cells, thereby contributing to mucosal injury.
Collapse
Affiliation(s)
- Somnath Mazumder
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Rudranil De
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Subhashis Debsharma
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 736101
| | - Pallab Maity
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Souvik Sarkar
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Shubhra Jyoti Saha
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Asim Azhar Siddiqui
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Chinmoy Banerjee
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Shiladitya Nag
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Debanjan Saha
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Saikat Pramanik
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032
| | - Kalyan Mitra
- Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Uday Bandyopadhyay
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032.
| |
Collapse
|
24
|
Sharma LK, Tiwari M, Rai NK, Bai Y. Mitophagy activation repairs Leber's hereditary optic neuropathy-associated mitochondrial dysfunction and improves cell survival. Hum Mol Genet 2019; 28:422-433. [PMID: 30304398 PMCID: PMC6489411 DOI: 10.1093/hmg/ddy354] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/31/2018] [Accepted: 09/26/2018] [Indexed: 12/24/2022] Open
Abstract
Leber's hereditary optic neuropathy (LHON) is a classical mitochondrial disease caused by mutations in the mitochondrial DNA encoding complex I subunits. Oxidative stress associated with complex I defect has been implicated in developing LHON phenotype such as retinal ganglion cell (RGC) death and loss of vision. However, the mechanism of LHON pathogenesis is still not very clear and thus no effective therapies are available to date. Using cybrid models for LHON, we show that autophagy is significantly compromised in cells carrying LHON-specific mtDNA mutations, which results in reduced clearance of dysfunctional mitochondria contributing to cell death. We further show that pharmacological activation of autophagy selectively clears the damaged mitochondria and thus repairs mitochondrial defects and improves overall cell survival in LHON cell models. Our results suggest that compromised autophagy is the missing link from oxidative stress to LHON pathogenesis. Activation of mitophagy ameliorates mitochondrial defects and exerts a protective role by improving cell survival in cells carrying LHON mutations that could be utilized as a potential therapeutic target for LHON treatment.
Collapse
Affiliation(s)
- Lokendra Kumar Sharma
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biotechnology, Centre for Biological Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Meenakshi Tiwari
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathology / Lab Medicine, All India Institute of Medical Sciences-Patna, Phulwarisharif, Patna, Bihar, India
| | - Neeraj Kumar Rai
- Department of Biotechnology, Centre for Biological Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Yidong Bai
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
25
|
Korski KI, Kubli DA, Wang BJ, Khalafalla FG, Monsanto MM, Firouzi F, Echeagaray OH, Kim T, Adamson RM, Dembitsky WP, Gustafsson ÅB, Sussman MA. Hypoxia Prevents Mitochondrial Dysfunction and Senescence in Human c-Kit + Cardiac Progenitor Cells. Stem Cells 2019; 37:555-567. [PMID: 30629785 DOI: 10.1002/stem.2970] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/10/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
Senescence-associated dysfunction deleteriously affects biological activities of human c-Kit+ cardiac progenitor cells (hCPCs), particularly under conditions of in vitro culture. In comparison, preservation of self-renewal and decreases in mitochondrial reactive oxygen species (ROS) are characteristics of murine CPCs in vivo that reside within hypoxic niches. Recapitulating hypoxic niche oxygen tension conditions of ∼1% O2 in vitro for expansion of hCPCs rather than typical normoxic cell culture conditions (21% O2 ) could provide significant improvement of functional and biological activities of hCPCs. hCPCs were isolated and expanded under permanent hypoxic (hCPC-1%) or normoxic (hCPC-21%) conditions from left ventricular tissue explants collected during left ventricular assist device implantation. hCPC-1% exhibit increased self-renewal and suppression of senescence characteristics relative to hCPC-21%. Oxidative stress contributed to higher susceptibility to apoptosis, as well as decreased mitochondrial function in hCPC-21%. Hypoxia prevented accumulation of dysfunctional mitochondria, supporting higher oxygen consumption rates and mitochondrial membrane potential. Mitochondrial ROS was an upstream mediator of senescence since treatment of hCPC-1% with mitochondrial inhibitor antimycin A recapitulated mitochondrial dysfunction and senescence observed in hCPC-21%. NAD+ /NADH ratio and autophagic flux, which are key factors for mitochondrial function, were higher in hCPC-1%, but hCPC-21% were highly dependent on BNIP3/NIX-mediated mitophagy to maintain mitochondrial function. Overall, results demonstrate that supraphysiological oxygen tension during in vitro expansion initiates a downward spiral of oxidative stress, mitochondrial dysfunction, and cellular energy imbalance culminating in early proliferation arrest of hCPCs. Senescence is inhibited by preventing ROS through hypoxic culture of hCPCs. Stem Cells 2019;37:555-567.
Collapse
Affiliation(s)
- Kelli I Korski
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Dieter A Kubli
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Bingyan J Wang
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Farid G Khalafalla
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Megan M Monsanto
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Fareheh Firouzi
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Oscar H Echeagaray
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Taeyong Kim
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Robert M Adamson
- Division of Cardiology, Sharp Hospital, San Diego, California, USA
| | | | - Åsa B Gustafsson
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Mark A Sussman
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| |
Collapse
|
26
|
Onyango AN. Cellular Stresses and Stress Responses in the Pathogenesis of Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4321714. [PMID: 30116482 PMCID: PMC6079365 DOI: 10.1155/2018/4321714] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/18/2018] [Indexed: 12/14/2022]
Abstract
Insulin resistance (IR), a key component of the metabolic syndrome, precedes the development of diabetes, cardiovascular disease, and Alzheimer's disease. Its etiological pathways are not well defined, although many contributory mechanisms have been established. This article summarizes such mechanisms into the hypothesis that factors like nutrient overload, physical inactivity, hypoxia, psychological stress, and environmental pollutants induce a network of cellular stresses, stress responses, and stress response dysregulations that jointly inhibit insulin signaling in insulin target cells including endothelial cells, hepatocytes, myocytes, hypothalamic neurons, and adipocytes. The insulin resistance-inducing cellular stresses include oxidative, nitrosative, carbonyl/electrophilic, genotoxic, and endoplasmic reticulum stresses; the stress responses include the ubiquitin-proteasome pathway, the DNA damage response, the unfolded protein response, apoptosis, inflammasome activation, and pyroptosis, while the dysregulated responses include the heat shock response, autophagy, and nuclear factor erythroid-2-related factor 2 signaling. Insulin target cells also produce metabolites that exacerbate cellular stress generation both locally and systemically, partly through recruitment and activation of myeloid cells which sustain a state of chronic inflammation. Thus, insulin resistance may be prevented or attenuated by multiple approaches targeting the different cellular stresses and stress responses.
Collapse
Affiliation(s)
- Arnold N. Onyango
- Department of Food Science and Technology, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, Nairobi 00200, Kenya
| |
Collapse
|
27
|
Wang YH, Twu YC, Wang CK, Lin FZ, Lee CY, Liao YJ. Niemann-Pick Type C2 Protein Regulates Free Cholesterol Accumulation and Influences Hepatic Stellate Cell Proliferation and Mitochondrial Respiration Function. Int J Mol Sci 2018; 19:ijms19061678. [PMID: 29874879 PMCID: PMC6032364 DOI: 10.3390/ijms19061678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/23/2022] Open
Abstract
Liver fibrosis is the first step toward the progression to cirrhosis, portal hypertension, and hepatocellular carcinoma. A high-cholesterol diet is associated with liver fibrosis via the accumulation of free cholesterol in hepatic stellate cells (HSCs). Niemann-Pick type C2 (NPC2) plays an important role in the regulation of intracellular free cholesterol homeostasis via direct binding with free cholesterol. Previously, we reported that NPC2 was downregulated in liver cirrhosis tissues. Loss of NPC2 enhanced the accumulation of free cholesterol in HSCs and made them more susceptible to transforming growth factor (TGF)-β1. In this study, we showed that knockdown of NPC2 resulted in marked increases in platelet-derived growth factor BB (PDGF-BB)-induced HSC proliferation through enhanced extracellular signal-regulated kinases (ERK), p38, c-Jun N-terminal kinases (JNK), and protein kinase B (AKT) phosphorylation. In contrast, NPC2 overexpression decreased PDGF-BB-induced cell proliferation by inhibiting p38, JNK, and AKT phosphorylation. Although NPC2 expression did not affect caspase-related apoptosis, the autophagy marker light chain 3β (LC3B) was decreased in NPC2 knockdown, and free cholesterol accumulated in the HSCs. The mitochondrial respiration functions (such as oxygen consumption rate, ATP production, and maximal respiratory capacity) were decreased in NPC2 knockdown, and free cholesterol accumulated in the HSCs, while NPC2-overexpressed cells remained normal. In addition, NPC2 expression did not affect the susceptibility of HSCs to lipopolysaccharides (LPS), and U18666A treatment induced free cholesterol accumulation, which enhanced LPS-induced Toll-like receptor 4 (TLR4), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 phosphorylation, interleukin (IL)-1 and IL-6 expression. Our study demonstrated that NPC2-mediated free cholesterol homeostasis controls HSC proliferation and mitochondrial function.
Collapse
Affiliation(s)
- Yuan-Hsi Wang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan.
| | - Yuh-Ching Twu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan.
| | - Chung-Kwe Wang
- Department of International Medicine, Taipei City Hospital Ranai Branch, Taipei 106, Taiwan.
| | - Fu-Zhen Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Chun-Ya Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| | - Yi-Jen Liao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
28
|
DNAJB1-PRKACA fusion kinase interacts with β-catenin and the liver regenerative response to drive fibrolamellar hepatocellular carcinoma. Proc Natl Acad Sci U S A 2017; 114:13076-13084. [PMID: 29162699 DOI: 10.1073/pnas.1716483114] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A segmental deletion resulting in DNAJB1-PRKACA gene fusion is now recognized as the signature genetic event of fibrolamellar hepatocellular carcinoma (FL-HCC), a rare but lethal liver cancer that primarily affects adolescents and young adults. Here we implement CRISPR-Cas9 genome editing and transposon-mediated somatic gene transfer to demonstrate that expression of either the endogenous fusion protein or a chimeric cDNA leads to the formation of indolent liver tumors in mice that closely resemble human FL-HCC. Notably, overexpression of the wild-type PRKACA was unable to fully recapitulate the oncogenic activity of DNAJB1-PRKACA, implying that FL-HCC does not simply result from enhanced PRKACA expression. Tumorigenesis was significantly enhanced by genetic activation of β-catenin, an observation supported by evidence of recurrent Wnt pathway mutations in human FL-HCC, as well as treatment with the hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which causes tissue injury, inflammation, and fibrosis. Our study validates the DNAJB1-PRKACA fusion kinase as an oncogenic driver and candidate drug target for FL-HCC, and establishes a practical model for preclinical studies to identify strategies to treat this disease.
Collapse
|
29
|
Folkerts H, Hilgendorf S, Wierenga ATJ, Jaques J, Mulder AB, Coffer PJ, Schuringa JJ, Vellenga E. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis 2017; 8:e2927. [PMID: 28703806 PMCID: PMC5550863 DOI: 10.1038/cddis.2017.317] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Here we have explored whether inhibition of autophagy can be used as a treatment strategy for acute myeloid leukemia (AML). Steady-state autophagy was measured in leukemic cell lines and primary human CD34+ AML cells with a large variability in basal autophagy between AMLs observed. The autophagy flux was higher in AMLs classified as poor risk, which are frequently associated with TP53 mutations (TP53mut), compared with favorable- and intermediate-risk AMLs. In addition, the higher flux was associated with a higher expression level of several autophagy genes, but was not affected by alterations in p53 expression by knocking down p53 or overexpression of wild-type p53 or p53R273H. AML CD34+ cells were more sensitive to the autophagy inhibitor hydroxychloroquine (HCQ) than normal bone marrow CD34+ cells. Similar, inhibition of autophagy by knockdown of ATG5 or ATG7 triggered apoptosis, which coincided with increased expression of p53. In contrast to wild-type p53 AML (TP53wt), HCQ treatment did not trigger a BAX and PUMA-dependent apoptotic response in AMLs harboring TP53mut. To further characterize autophagy in the leukemic stem cell-enriched cell fraction AML CD34+ cells were separated into ROSlow and ROShigh subfractions. The immature AML CD34+-enriched ROSlow cells maintained higher basal autophagy and showed reduced survival upon HCQ treatment compared with ROShigh cells. Finally, knockdown of ATG5 inhibits in vivo maintenance of AML CD34+ cells in NSG mice. These results indicate that targeting autophagy might provide new therapeutic options for treatment of AML since it affects the immature AML subfraction.
Collapse
Affiliation(s)
- Hendrik Folkerts
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Susan Hilgendorf
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albertus T J Wierenga
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Jennifer Jaques
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - André B Mulder
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul J Coffer
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands.,Center of Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edo Vellenga
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
30
|
Correia M, Pinheiro P, Batista R, Soares P, Sobrinho-Simões M, Máximo V. Etiopathogenesis of oncocytomas. Semin Cancer Biol 2017; 47:82-94. [PMID: 28687249 DOI: 10.1016/j.semcancer.2017.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 01/01/2023]
Abstract
Oncocytomas are distinct tumors characterized by an abnormal accumulation of defective and (most probably) dysfunctional mitochondria in cell cytoplasm of such tumors. This particular phenotype has been studied for the last decades and the clarification of the etiopathogenic causes are still needed. Several mechanisms involved in the formation and maintenance of oncocytomas are accepted as reasonable causes, but the relevance and contribution of each one for oncocytic transformation may depend on different cancer etiopathogenic contexts. In this review, we describe the current knowledge of the etiopathogenic events that may lead to oncocytic transformation and discuss their contribution for tumor progression and mitochondrial accumulation.
Collapse
Affiliation(s)
- Marcelo Correia
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Pedro Pinheiro
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Rui Batista
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal
| | - Paula Soares
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal; Department of Pathology, Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal
| | - Manuel Sobrinho-Simões
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal; Department of Pathology, Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal; Department of Pathology, Centro Hospitalar São João, Porto, Portugal
| | - Valdemar Máximo
- Cancer Signalling and Metabolism Research Group, Instituto de Investigação e Inovação em Saúde - i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal; Cancer Signalling and Metabolism Research Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal; Department of Pathology, Faculdade de Medicina da Universidade do Porto - FMUP (Medical Faculty of University of Porto), Porto, Portugal.
| |
Collapse
|
31
|
MITA modulated autophagy flux promotes cell death in breast cancer cells. Cell Signal 2017; 35:73-83. [DOI: 10.1016/j.cellsig.2017.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/12/2017] [Accepted: 03/29/2017] [Indexed: 12/28/2022]
|
32
|
Edens BM, Miller N, Ma YC. Impaired Autophagy and Defective Mitochondrial Function: Converging Paths on the Road to Motor Neuron Degeneration. Front Cell Neurosci 2016; 10:44. [PMID: 26973461 PMCID: PMC4776126 DOI: 10.3389/fncel.2016.00044] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/08/2016] [Indexed: 12/12/2022] Open
Abstract
Selective motor neuron degeneration is a hallmark of amyotrophic lateral sclerosis (ALS). Around 10% of all cases present as familial ALS (FALS), while sporadic ALS (SALS) accounts for the remaining 90%. Diverse genetic mutations leading to FALS have been identified, but the underlying causes of SALS remain largely unknown. Despite the heterogeneous and incompletely understood etiology, different types of ALS exhibit overlapping pathology and common phenotypes, including protein aggregation and mitochondrial deficiencies. Here, we review the current understanding of mechanisms leading to motor neuron degeneration in ALS as they pertain to disrupted cellular clearance pathways, ATP biogenesis, calcium buffering and mitochondrial dynamics. Through focusing on impaired autophagic and mitochondrial functions, we highlight how the convergence of diverse cellular processes and pathways contributes to common pathology in motor neuron degeneration.
Collapse
Affiliation(s)
- Brittany M. Edens
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| | - Nimrod Miller
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| | - Yong-Chao Ma
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| |
Collapse
|
33
|
Choi SI, Kim EK. Autophagy in granular corneal dystrophy type 2. Exp Eye Res 2016; 144:14-21. [DOI: 10.1016/j.exer.2015.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 08/28/2015] [Accepted: 09/15/2015] [Indexed: 01/07/2023]
|
34
|
Gao J, Zou X, Yang L, Feng Z, Liu J. Hydroxytyrosol protects against acrolein induced preosteoblast cell toxicity: Involvement of Nrf2/Keap1 pathway. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
35
|
Li R, Jen N, Wu L, Lee J, Fang K, Quigley K, Lee K, Wang S, Zhou B, Vergnes L, Chen YR, Li Z, Reue K, Ann DK, Hsiai TK. Disturbed Flow Induces Autophagy, but Impairs Autophagic Flux to Perturb Mitochondrial Homeostasis. Antioxid Redox Signal 2015; 23:1207-19. [PMID: 26120766 PMCID: PMC4657520 DOI: 10.1089/ars.2014.5896] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIM Temporal and spatial variations in shear stress are intimately linked with vascular metabolic effects. Autophagy is tightly regulated in intracellular bulk degradation/recycling system for maintaining cellular homeostasis. We postulated that disturbed flow modulates autophagy with an implication in mitochondrial superoxide (mtO2(•-)) production. RESULTS In the disturbed flow or oscillatory shear stress (OSS)-exposed aortic arch, we observed prominent staining of p62, a reverse marker of autophagic flux, whereas in the pulsatile shear stress (PSS)-exposed descending aorta, p62 was attenuated. OSS significantly increased (i) microtubule-associated protein light chain 3 (LC3) II to I ratios in human aortic endothelial cells, (ii) autophagosome formation as quantified by green fluorescent protein (GFP)-LC3 dots per cell, and (iii) p62 protein levels, whereas manganese superoxide dismutase (MnSOD) overexpression by recombinant adenovirus, N-acetyl cysteine treatment, or c-Jun N-terminal kinase (JNK) inhibition reduced OSS-mediated LC3-II/LC3-I ratios and mitochondrial DNA damage. Introducing bafilomycin to Earle's balanced salt solution or to OSS condition incrementally increased both LC3-II/LC3-I ratios and p62 levels, implicating impaired autophagic flux. In the OSS-exposed aortic arch, both anti-phospho-JNK and anti-8-hydroxy-2'-deoxyguanosine (8-OHdG) staining for DNA damage were prominent, whereas in the PSS-exposed descending aorta, the staining was nearly absent. Knockdown of ATG5 with siRNA increased OSS-mediated mtO2(•-), whereas starvation or rapamycin-induced autophagy reduced OSS-mediated mtO2(•-), mitochondrial respiration, and complex II activity. INNOVATION Disturbed flow-mediated oxidative stress and JNK activation induce autophagy. CONCLUSION OSS impairs autophagic flux to interfere with mitochondrial homeostasis. Antioxid. Redox Signal. 23, 1207-1219.
Collapse
Affiliation(s)
- Rongsong Li
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Nelson Jen
- 2 Department of Bioengineering, UCLA Henry Samueli School of Engineering and Applied Science , Los Angeles, California
| | - Lan Wu
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Juhyun Lee
- 2 Department of Bioengineering, UCLA Henry Samueli School of Engineering and Applied Science , Los Angeles, California
| | - Karen Fang
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Katherine Quigley
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Katherine Lee
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Sky Wang
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Bill Zhou
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Laurent Vergnes
- 3 Department of Human Genetics, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Yun-Ru Chen
- 4 Department of Molecular Pharmacology, Beckman Research Institute, City of Hope National Medical Center , Duarte, California
| | - Zhaoping Li
- 5 Department of Medicine, VA Greater Los Angeles Healthcare System, UCLA David Geffen School of Medicine , Los Angeles, California
| | - Karen Reue
- 3 Department of Human Genetics, UCLA David Geffen School of Medicine , Los Angeles, California
| | - David K Ann
- 4 Department of Molecular Pharmacology, Beckman Research Institute, City of Hope National Medical Center , Duarte, California
| | - Tzung K Hsiai
- 1 Division of Cardiology, Department of Medicine, UCLA David Geffen School of Medicine , Los Angeles, California.,2 Department of Bioengineering, UCLA Henry Samueli School of Engineering and Applied Science , Los Angeles, California.,5 Department of Medicine, VA Greater Los Angeles Healthcare System, UCLA David Geffen School of Medicine , Los Angeles, California
| |
Collapse
|
36
|
Kamat PK, Kalani A, Tyagi SC, Tyagi N. Hydrogen Sulfide Epigenetically Attenuates Homocysteine-Induced Mitochondrial Toxicity Mediated Through NMDA Receptor in Mouse Brain Endothelial (bEnd3) Cells. J Cell Physiol 2015; 230:378-94. [PMID: 25056869 DOI: 10.1002/jcp.24722] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/18/2014] [Indexed: 01/08/2023]
Abstract
Previously we have shown that homocysteine (Hcy) caused oxidative stress and altered mitochondrial function. Hydrogen sulfide (H2S) has potent anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Therefore, in the present study we examined whether H2S ameliorates Hcy-induced mitochondrial toxicity which led to endothelial dysfunction in part, by epigenetic alterations in mouse brain endothelial cells (bEnd3). The bEnd3 cells were exposed to 100 μM Hcy treatment in the presence or absence of 30 μM NaHS (donor of H2S) for 24 h. Hcy-activate NMDA receptor and induced mitochondrial toxicity by increased levels of Ca(2+), NADPH-oxidase-4 (NOX-4) expression, mitochondrial dehydrogenase activity and decreased the level of nitrate, superoxide dismutase (SOD-2) expression, mitochondria membrane potentials, ATP production. To confirm the role of epigenetic, 5'-azacitidine (an epigenetic modulator) treatment was given to the cells. Pretreatment with NaHS (30 μM) attenuated the Hcy-induced increased expression of DNMT1, DNMT3a, Ca(2+), and decreased expression of DNMT3b in bEND3 cells. Furthermore, NaHS treatment also mitigated mitochondrial oxidative stress (NOX4, ROS, and NO) and restored ATP that indicates its protective effects against mitochondrial toxicity. Additional, NaHS significantly alleviated Hcy-induced LC3-I/II, CSE, Atg3/7, and low p62 expression which confirm its effect on mitophagy. Likewise, NaHS also restored level of eNOS, CD31, VE-cadherin and ET-1 and maintains endothelial function in Hcy treated cells. Molecular inhibition of NMDA receptor by using small interfering RNA showed protective effect whereas inhibition of H2S production by propargylglycine (PG) (inhibitor of enzyme CSE) showed mitotoxic effect. Taken together, results demonstrate that, administration of H2S protected the cells from HHcy-induced mitochondrial toxicity and endothelial dysfunction.
Collapse
Affiliation(s)
- Pradip K Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
37
|
Panda PK, Mukhopadhyay S, Das DN, Sinha N, Naik PP, Bhutia SK. Mechanism of autophagic regulation in carcinogenesis and cancer therapeutics. Semin Cell Dev Biol 2015; 39:43-55. [PMID: 25724561 DOI: 10.1016/j.semcdb.2015.02.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/12/2015] [Accepted: 02/19/2015] [Indexed: 12/15/2022]
Abstract
Autophagy in cancer is an intensely debated concept in the field of translational research. The dual nature of autophagy implies that it can potentially modulate the pro-survival and pro-death mechanisms in tumor initiation and progression. There is a prospective molecular relationship between defective autophagy and tumorigenesis that involves the accumulation of damaged mitochondria and protein aggregates, which leads to the production of reactive oxygen species (ROS) and ultimately causes DNA damage that can lead to genomic instability. Moreover, autophagy regulates necrosis and is followed by inflammation, which limits tumor metastasis. On the other hand, autophagy provides a survival advantage to detached, dormant metastatic cells through nutrient fueling by tumor-associated stromal cells. Manipulating autophagy for induction of cell death, inhibition of protective autophagy at tissue-and context-dependent for apoptosis modulation has therapeutic implications. This review presents a comprehensive overview of the present state of knowledge regarding autophagy as a new approach to treat cancer.
Collapse
Affiliation(s)
- Prashanta Kumar Panda
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Subhadip Mukhopadhyay
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Durgesh Nandini Das
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Niharika Sinha
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Prajna Paramita Naik
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India.
| |
Collapse
|
38
|
Duan JL, Yin J, Ren WK, Wu MM, Chen S, Cui ZJ, Wu X, Huang RL, Li TJ, Yin YL. Pyrrolidine dithiocarbamate restores gastric damages and suppressive autophagy induced by hydrogen peroxide. Free Radic Res 2015; 49:210-8. [PMID: 25471085 DOI: 10.3109/10715762.2014.993627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well known that gastric barrier is very important for protecting host from various insults. Simultaneously, autophagy serving as a prominent cytoprotective and survival pathway under oxidative stress conditions is being increasingly recognized. Thus, this study was conducted for investigating the effect of pyrrolidine dithiocarbamate (PDTC) on gastric barrier function and autophagy under oxidative stress induced by intragastric administration of hydrogen peroxide (H2O2). The gastric tight junction proteins [zonula occludens-1 (ZO1), occludin, and claudin1], autophagic proteins [microtubule-associated protein light chain 3I(LC3I), LC3II, and beclin1], and nuclear factor kappa B (NF-κB) signaling pathway (p65 and IκB kinase α/β) were determined by Western blot. The results showed that H2O2 exposure disturbed gastric barrier function with decreased expression of ZO1, occludin, and claudin1, and reduced gastric autophagy with decreased conversion of LC3I into LC3II in mice. However, treatment with PDTC restored these adverse effects evidenced by increased expression of ZO1 and claudin1 and increased conversion of LC3I into LC3II. Meanwhile, H2O2 exposure decreased normal human gastric epithelial mucosa cell line (GES-1) viability in a concentration-dependent way. However, after being exposed to H2O2, GES-1 exhibited autophagic response which was inconsistent with our in vivo results in mice, while PDTC failed to decrease autophagy in GES-1 induced by H2O2. Simultaneously, the beneficial effect of PDTC on gastric damage and autophagy in mice might be independent of inhibition of NF-κB. In conclusion, PDTC treatment restores gastric damages and reduced autophagy induced by H2O2. Therefore, PDTC may serve as a potential adjuvant therapy for gastric damages.
Collapse
Affiliation(s)
- J L Duan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha, Hunan , P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Datta K, Suman S, Fornace AJ. Radiation persistently promoted oxidative stress, activated mTOR via PI3K/Akt, and downregulated autophagy pathway in mouse intestine. Int J Biochem Cell Biol 2014; 57:167-76. [PMID: 25449263 DOI: 10.1016/j.biocel.2014.10.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/14/2014] [Accepted: 10/17/2014] [Indexed: 12/19/2022]
Abstract
While acute effects of toxic radiation doses on intestine are well established, we are yet to acquire a complete spectrum of sub-lethal radiation-induced chronic intestinal perturbations at the molecular level. We investigated persistent effects of a radiation dose (2 Gy) commonly used as a daily fraction in radiotherapy on oxidants and anti-oxidants, and autophagy pathways, which are interlinked processes affecting intestinal homeostasis. Six to eight weeks old C57BL/6J mice (n=10) were exposed to 2 Gy γ-ray. Mice were euthanized two or twelve months after radiation, intestine surgically removed, and flushed using sterile PBS. Parts of the intestine from jejunal-ilial region were fixed, frozen, or used for intestinal epithelial cell (IEC) isolation. While oxidant levels and mitochondrial status were assessed in isolated IEC, autophagy and oxidative stress related signaling pathways were probed in frozen and fixed samples using PCR-based expression arrays and immunoprobing. Radiation exposure caused significant alterations in the expression level of 26 autophagy and 17 oxidative stress related genes. Immunoblot results showed decreased Beclin1 and LC3-II and increased p62, PI3K/Akt, and mTOR. Flow cytometry data showed increased oxidant production and compromised mitochondrial integrity in irradiated samples. Immunoprobing of intestinal sections showed increased 8-oxo-dG and nuclear PCNA, and decreased autophagosome marker LC3-II in IEC after irradiation. We show that sub-lethal radiation could persistently downregulate anti-oxidants and autophagy signaling, and upregulate oxidant production and proliferative signaling. Radiation-induced promotion of oxidative stress and downregulation of autophagy could work in tandem to alter intestinal functions and have implications for post-radiation chronic gastrointestinal diseases.
Collapse
Affiliation(s)
- Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Shubhankar Suman
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA; Center of Excellence In Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
40
|
Takemura S, Ichikawa H, Naito Y, Takagi T, Yoshikawa T, Minamiyama Y. S-allyl cysteine ameliorates the quality of sperm and provides protection from age-related sperm dysfunction and oxidative stress in rats. J Clin Biochem Nutr 2014; 55:155-61. [PMID: 25411519 PMCID: PMC4227827 DOI: 10.3164/jcbn.14-39] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 05/13/2014] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species play a central role in the pathophysiology of the age-related decrease in male fertility. It has been reported that the total protein of DJ-1 was decreased in a proteomic analysis of seminal plasma from asthenozoospermia patients and a DJ-1 protein acts as a sensor of cellular redox homeostasis. Therefore, we evaluated the age-related changes in the ratio of the oxidized/reduced forms of the DJ-1 protein in the epididymis. In addition, the protective effects of S-allyl cysteine (SAC), a potent antioxidant, were evaluated against sperm dysfunction. Male rats aged 15–75 weeks were used to assess age-associated sperm function and oxidative stress. Sperm count increased until 25 weeks, but then decreased at 50 and 75 weeks. The rate of sperm movement at 75 weeks was decreased to approximately 60% of the rate observed at 25 weeks. Expression of DJ-1 decreased, but oxidized-DJ-1 increased with age. In addition, 4-hydroxy-2-nonenal modified proteins in the epididymis increased until 50 weeks of age. The total number and DNA synthetic potential of the sperm increased until 25 weeks, and then decreased. In rats 75 weeks of age, SAC (0.45% diet) attenuated the decrease in the number, motility, and DNA synthesis of sperm and inhibited the oxidized proteins. These results suggest that SAC ameliorates the quality of sperm subjected to age-associated oxidative stress.
Collapse
Affiliation(s)
- Shigekazu Takemura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Hiroshi Ichikawa
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto 610-0321, Japan
| | - Yuji Naito
- Department of Gastroenterology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tomohisa Takagi
- Department of Gastroenterology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Yoshikawa
- Department of Gastroenterology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yukiko Minamiyama
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan ; Food Hygiene and Environmental Health Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho, Shimogamo, Sakyo-ku, Kyoto 606-8522, Japan
| |
Collapse
|
41
|
Trehalose improves human fibroblast deficits in a new CHIP-mutation related ataxia. PLoS One 2014; 9:e106931. [PMID: 25259530 PMCID: PMC4178022 DOI: 10.1371/journal.pone.0106931] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 08/10/2014] [Indexed: 11/19/2022] Open
Abstract
In this work we investigate the role of CHIP in a new CHIP-mutation related ataxia and the therapeutic potential of trehalose. The patient's fibroblasts with a new form of hereditary ataxia, related to STUB1 gene (CHIP) mutations, and three age and sex-matched controls were treated with epoxomicin and trehalose. The effects on cell death, protein misfolding and proteostasis were evaluated. Recent studies have revealed that mutations in STUB-1 gene lead to a growing list of molecular defects as deregulation of protein quality, inhibition of proteasome, cell death, decreased autophagy and alteration in CHIP and HSP70 levels. In this CHIP-mutant patient fibroblasts the inhibition of proteasome with epoxomicin induced severe pathophysiological age-associated changes, cell death and protein ubiquitination. Additionally, treatment with epoxomicin produced a dose-dependent increase in the number of cleaved caspase-3 positive cells. However, co-treatment with trehalose, a disaccharide of glucose present in a wide variety of organisms and known as a autophagy enhancer, reduced these pathological events. Trehalose application also increased CHIP and HSP70 expression and GSH free radical levels. Furthermore, trehalose augmented macro and chaperone mediated autophagy (CMA), rising the levels of LC3, LAMP2, CD63 and increasing the expression of Beclin-1 and Atg5-Atg12. Trehalose treatment in addition increased the percentage of immunoreactive cells to HSC70 and LAMP2 and reduced the autophagic substrate, p62. Although this is an individual case based on only one patient and the statistical comparisons are not valid between controls and patient, the low variability among controls and the obvious differences with this patient allow us to conclude that trehalose, through its autophagy activation capacity, anti-aggregation properties, anti-oxidative effects and lack of toxicity, could be very promising for the treatment of CHIP-mutation related ataxia, and possibly a wide spectrum of neurodegenerative disorders related to protein disconformation.
Collapse
|
42
|
Antioxidant supplement inhibits skeletal muscle constitutive autophagy rather than fasting-induced autophagy in mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:315896. [PMID: 25028602 PMCID: PMC4084590 DOI: 10.1155/2014/315896] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/28/2014] [Accepted: 05/16/2014] [Indexed: 12/13/2022]
Abstract
In this study, we tested the hypothesis that NAC administration leads to reduced oxidative stress and thus to decreased expression of autophagy markers in young mice. Our results reveal that NAC administration results in reduced muscle mRNA levels of several autophagy markers, including Beclin-1, Atg7, LC3, Atg9, and LAMP2. However, NAC supplement fails to block the activation of skeletal muscle autophagy in response to fasting, because fasting significantly increases the mRNA level of several autophagy markers and LC3 lipidation. We further examined the effects of NAC administration on mitochondrial antioxidant capacity in fed and 24-hour fasted mice. Our results clearly show that NAC administration depresses the expression of manganese superoxide dismutase (MnSOD) and TP53-induced glycolysis and apoptosis regulator (TIGAR), both of which play a predominant antioxidant role in mitochondria by reducing ROS level. In addition, we found no beneficial effect of NAC supplement on muscle mass but it can protect from muscle loss in response to fasting. Collectively, our findings indicate that ROS is required for skeletal muscle constitutive autophagy, rather than starvation-induced autophagy, and that antioxidant NAC inhibits constitutive autophagy by the regulation of mitochondrial ROS production and antioxidant capacity.
Collapse
|
43
|
Luo C, Li Y, Yang L, Wang X, Long J, Liu J. Superparamagnetic iron oxide nanoparticles exacerbate the risks of reactive oxygen species-mediated external stresses. Arch Toxicol 2014; 89:357-69. [PMID: 24847785 DOI: 10.1007/s00204-014-1267-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 05/13/2014] [Indexed: 01/13/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (IONPs) have been widely applied in numerous biomedical fields. The evaluation of the toxicity of IONPs to the environment and human beings is indispensable to guide their applications. IONPs are usually considered to have good biocompatibility; however, some literatures have reported the toxicity of IONPs in vitro and in vivo. The controversy surrounding the biocompatibility of IONPs prompted us to carefully consider the biological effects of IONPs, especially under stress conditions. However, the potential risks of IONPs under stress conditions have not yet been evaluated in depth. Acrolein is widespread in the environment and modulates stress-induced gene activation and cell death in many organs and tissues. In this study, we assessed the sensitivity of H9c2 cardiomyocyte cells embedded with IONPs to acrolein and investigated the possible molecular mechanisms involved in this sensitivity. IONPs, which alone exhibited no toxicity, sensitized the H9c2 cardiomyocytes to acrolein-induced dysfunction. The IONP/acrolein treatment induced a loss of viability, membrane disruption, reactive oxygen species (ROS) generation, Erk activation, mitochondrial and lysosomal dysfunction, and necrosis in H9c2 cells. Treatment with an ROS generation inhibitor (diphenyleneiodonium) or an iron chelator (deferoxamine) prevented the IONP/acrolein-induced loss of viability, suggesting that ROS and IONP degradation facilitated the toxicity of the IONP/acrolein treatment in H9c2 cells. Our data suggest that cells embedded in IONPs are more vulnerable to oxidative stress, which confirms the hypothesis that nanoparticles can sensitize cells to the adverse effects of external stimulation. The present work provides a new perspective from which to evaluate the interactions between nanoparticles and cells.
Collapse
Affiliation(s)
- Cheng Luo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Frontier Institute of Science and Technology, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China,
| | | | | | | | | | | |
Collapse
|
44
|
Xie J, Potter A, Xie W, Lynch C, Seefeldt T. Evaluation of a dithiocarbamate derivative as a model of thiol oxidative stress in H9c2 rat cardiomyocytes. Free Radic Biol Med 2014; 70:214-22. [PMID: 24607690 PMCID: PMC4011664 DOI: 10.1016/j.freeradbiomed.2014.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/22/2014] [Accepted: 02/26/2014] [Indexed: 12/16/2022]
Abstract
Thiol redox state (TRS) refers to the balance between reduced thiols and their corresponding disulfides and is mainly reflected by the ratio of reduced and oxidized glutathione (GSH/GSSG). A decrease in GSH/GSSG, which reflects a state of thiol oxidative stress, as well as thiol modifications such as S-glutathionylation, has been shown to have important implications in a variety of cardiovascular diseases. Therefore, research models for inducing thiol oxidative stress are important tools for studying the pathophysiology of these disease states as well as examining the impact of pharmacological interventions on thiol pathways. The purpose of this study was to evaluate the use of a dithiocarbamate derivative, 2-acetylamino-3-[4-(2-acetylamino-2-carboxyethylsulfanylthiocarbonylamino)phenylthiocarbamoylsulfanyl]propionic acid (2-AAPA), as a pharmacological model of thiol oxidative stress by examining the extent of thiol modifications induced in H9c2 rat cardiomyocytes and its impact on cellular functions. The extent of thiol oxidative stress produced by 2-AAPA was also compared to other models of oxidative stress including hydrogen peroxide (H2O2), diamide, buthionine sulfoximine, and N,N׳-bis(2-chloroethyl)-N-nitroso-urea. Results indicated that 2-AAPA effectively inhibited glutathione reductase and thioredoxin reductase activities and decreased the GSH/GSSG ratio by causing a significant accumulation of GSSG. 2-AAPA also increased the formation of protein disulfides as well as S-glutathionylation. The alteration in TRS led to a loss of mitochondrial membrane potential, release of cytochrome c, and increase in reactive oxygen species production. Compared to other models, 2-AAPA is more potent at creating a state of thiol oxidative stress with lower cytotoxicity, higher specificity, and more pharmacological relevance, and could be utilized as a research tool to study TRS-related normal and abnormal biochemical processes in cardiovascular diseases.
Collapse
Affiliation(s)
- Jiashu Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Ashley Potter
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Wei Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Christophina Lynch
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Teresa Seefeldt
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA.
| |
Collapse
|
45
|
Pre-endurance training prevents acute alcoholic liver injury in rats through the regulation of damaged mitochondria accumulation and mitophagy balance. Hepatol Int 2014. [PMID: 26202644 DOI: 10.1007/s12072-014-9529-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM The aim of this study is to investigate the effect of pre-endurance training on the prevention of alcohol-induced acute hepatic injury and on hepatic mitophagy. METHODS Forty-eight male Sprague-Dawley rats were randomly divided into four groups: (1) control group, (2) 12-week exercise training group, (3) 5-day alcohol intake group, and (4) 12-week exercise training plus 5-day alcohol intake group. The rats were examined to determine the following: BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), hypoxia-inducible factor-1α (HIF-1α), cytochrome P450 2E1 (CYP2E1), alcohol dehydrogenase (ADH), microtubule-associated protein 1 light chain 3 (LC3II), Beclin1 mRNA and protein expressions, mitochondrial reactive oxygen species (ROS) production, mitochondrial thiobarbituric acid-reactive substances (TBARS) level, aconitase and ATP synthase activities, mitochondrial inner membrane potential, NADH/NAD(+) ratio, triglyceride (TG), the number of mtDNA and mitochondrial respiration functions in liver tissue, and serum ALT and AST. RESULTS Pre-endurance training attenuated acute alcohol treatment-induced increase in mitochondrial TBARS, ROS production, NADH/NAD(+) ratio, state 4 respiration rate, TG, serum ALT and AST, as well as BNIP3, HIF-1α, LC3II, and Beclin 1 mRNA and protein levels, however, CYP2E1 and ADH mRNA and protein levels unchanged. Meanwhile, it attenuated the acute alcohol intake-induced decrease in aconitase activity, inner mitochondrial membrane potential (Δψ), ATP synthase activity, state 3 respiration rate, respiratory control ratio, and the number of mtDNA. CONCLUSION Pre-endurance training can decrease acute alcohol intake-induced damaged mitochondria accumulation and reduced acute alcohol intake-induced mitophagy, which built a new balance between mitophagy and damaged mitochondria accumulation.
Collapse
|
46
|
Sun W, Zheng Y, Lu Z, Cui Y, Tian Q, Xiao S, Liu F, Liu J. Overexpression of S100A7 protects LPS-induced mitochondrial dysfunction and stimulates IL-6 and IL-8 in HaCaT cells. PLoS One 2014; 9:e92927. [PMID: 24671027 PMCID: PMC3966836 DOI: 10.1371/journal.pone.0092927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/26/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND S100A7 (or psoriasin) is distributed in the cytoplasm of keratinocytes of normal human epidermis, and it is overexpressed in many epidermal inflammatory diseases. Lipopolysaccharide (LPS) induces mitochondrial function changes, which play important roles in multiple cellular mechanisms including inflammation. Although S100A7 expression is regulated by various factors in the human epidermis during inflammation, whether S100A7 interacts with mitochondria in keratinocytes is not clear. OBJECTIVES Our study was designed to investigate whether S100A7 could prohibit mitochondrial dysfunction and stimulate cytokines in cultured normal HaCaT cells treated with LPS. RESULTS We generated HaCaT cells that constitutively express enhanced green fluorescence protein (EGFP)-S100A7 (S100A7-EGFP) or EGFP alone, as a control. Here, we show that S100A7-EGFP HaCaT cells exhibit an increase in mitochondrial DNA (mtDNA) copy number and mitochondrial membrane potential (MMP). qRT-PCR revealed that expression of three main mitochondrial biogenesis-associated genes was significantly increased: PPAR-coactivator-1alpha (PGC-1α), the mitochondrial transcription factor A (Tfam) and nuclear respiratory factor-1 (NRF1). S100A7 overexpression increased mtDNA content and effectively increased intracellular adenosine 5'-triphosphate (ATP) production, while decreasing reactive oxygen species (ROS) generation. S100A7 overexpression also significantly decreased the expression of Mfn2 and increased DRP1 expression compared with control EGFP cells. S100A7 down-regulated the expression of the autophagy-related proteins Beclin-1 and LC3B. S100A7 also increased expression of IL-6 and IL-8 cytokines. Knockdown of S100A7 decreased MMP and disrupted mitochondrial homeostasis. CONCLUSIONS These findings demonstrate that S100A7 stimulates mitochondrial biogenesis and increases mitochondrial function in HaCaT cells treated with LPS; and S100A7 also promotes secretion of IL-6 and IL-8.
Collapse
Affiliation(s)
- Wenyan Sun
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Yan Zheng
- Department of Dermatology, the 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P. R. China
- * E-mail:
| | - Zhuoyang Lu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Yang Cui
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Qiong Tian
- Department of Dermatology, the 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P. R. China
| | - Shengxiang Xiao
- Department of Dermatology, the 2nd Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P. R. China
| | - Feng Liu
- Department of Medicine, University of California Irvine Medical School, Irvine, United States of America
- Chao Family Comprehensive Cancer Center, University of California Irvine Medical School, Irvine, United States of America
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, United States of America
| |
Collapse
|
47
|
Liu K, Zhao H, Ji K, Yan C. MERRF/MELAS overlap syndrome due to the m.3291T>C mutation. Metab Brain Dis 2014; 29:139-44. [PMID: 24338029 DOI: 10.1007/s11011-013-9464-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
We report the case of a 19-year-old Chinese female harboring the m.3291T>C mutation in the MT-TL1 gene encoding the mitochondrial transfer RNA for leucine. She presented with a complex phenotype characterized by progressive cerebellar ataxia, frequent myoclonus seizures, recurrent stroke-like episodes, migraine-like headaches with nausea and vomiting, and elevated resting lactate blood level. It is known that the myoclonus epilepsy with ragged-red fibers (MERRF) is characterized by cerebellar ataxia and myoclonus epilepsy, while that the mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) is characterized by recurrent stroke-like episodes, migraine-like headaches, and elevated resting lactate blood level. So the patient's clinical manifestations suggest the presence of a MERRF/MELAS overlap syndrome. Muscle biopsy of the patient showed the presence of numerous scattered ragged-red fibers, some cytochrome c oxidase-deficient fibers, and several strongly succinate dehygrogenase-reactive vessels, suggestive of a mitochondrial disorder. Direct sequencing of the complete mitochondrial genome of the proband revealed no mutations other than the T-to-C transition at nucleotide position 3291. Restriction fragment length polymorphism analysis of the proband and her family revealed maternal inheritance of the mutation in a heteroplasmic manner. The analysis of aerobic respiration and glycolysis demonstrated that the fibroblasts from the patient had mitochondrial dysfunction. Our results suggest that the m.3291T>C is pathogenic. This study is the first to describe the m.3291T>C mutation in association with the MERRF/MELAS overlap syndrome.
Collapse
Affiliation(s)
- Kaiming Liu
- Laboratory of Neuromuscular Disorders and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | | | | | | |
Collapse
|
48
|
Morales CR, Pedrozo Z, Lavandero S, Hill JA. Oxidative stress and autophagy in cardiovascular homeostasis. Antioxid Redox Signal 2014; 20:507-18. [PMID: 23641894 PMCID: PMC3894700 DOI: 10.1089/ars.2013.5359] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Autophagy is an evolutionarily ancient process of intracellular protein and organelle recycling required to maintain cellular homeostasis in the face of a wide variety of stresses. Dysregulation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) leads to oxidative damage. Both autophagy and ROS/RNS serve pathological or adaptive roles within cardiomyocytes, depending on the context. RECENT ADVANCES ROS/RNS and autophagy communicate with each other via both transcriptional and post-translational events. This cross talk, in turn, regulates the structural integrity of cardiomyocytes, promotes proteostasis, and reduces inflammation, events critical to disease pathogenesis. CRITICAL ISSUES Dysregulation of either autophagy or redox state has been implicated in many cardiovascular diseases. Cardiomyocytes are rich in mitochondria, which make them particularly sensitive to oxidative damage. Maintenance of mitochondrial homeostasis and elimination of defective mitochondria are each critical to the maintenance of redox homeostasis. FUTURE DIRECTIONS The complex interplay between autophagy and oxidative stress underlies a wide range of physiological and pathological events and its elucidation holds promise of potential clinical applicability.
Collapse
Affiliation(s)
- Cyndi R Morales
- 1 Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center , Dallas, Texas
| | | | | | | |
Collapse
|
49
|
Malaud E, Merle D, Piquer D, Molina L, Salvetat N, Rubrecht L, Dupaty E, Galea P, Cobo S, Blanc A, Saussine M, Marty-Ané C, Albat B, Meilhac O, Rieunier F, Pouzet A, Molina F, Laune D, Fareh J. Local carotid atherosclerotic plaque proteins for the identification of circulating biomarkers in coronary patients. Atherosclerosis 2014; 233:551-558. [PMID: 24530963 DOI: 10.1016/j.atherosclerosis.2013.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/18/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To identify circulating biomarkers that originate from atherosclerotic vulnerable plaques and that could predict future cardiovascular events. METHODS After a protein enrichment step (combinatorial peptide ligand library approach), we performed a two-dimensional electrophoresis comparative analysis on human carotid plaque protein extracts (fibrotic and hemorrhagic atherosclerotic plaques). In silico analysis of the biological processes was applied on proteomic data. Luminex xMAP assays were used to quantify inflammatory components in carotid plaques. The systemic quantification of proteins originating from vulnerable plaques in blood samples from patients with stable and unstable coronary disease was evaluated. RESULTS A total of 118 proteins are differentially expressed in fibrotic and hemorrhagic plaques, and allowed the identification of three biological processes related to atherosclerosis (platelet degranulation, vascular autophagy and negative regulation of fibrinolysis). The multiplex assays revealed an increasing expression of VEGF, IL-6, IL-8, IP-10 and RANTES in hemorrhagic as compared to fibrotic plaques (p<0.05). Measurement of protein expressions in plasmas from patients with stable and unstable coronary disease identified a combination of biomarkers, including proteins of the smooth muscle cell integrity (Calponin-1), oxidative stress (DJ-1) and inflammation (IL-8), that allows the accurate classification of patients at risk (p=0.0006). CONCLUSION Using tissue protein enrichment technology, we validated proteins that are differentially expressed in hemorrhagic plaques as potential circulating biomarkers of coronary patients. Combinations of such circulating biomarkers could be used to stratify coronary patients.
Collapse
Affiliation(s)
- Eric Malaud
- UMR3145 CNRS Bio-Rad, SysDiag, Montpellier, France
| | | | | | | | | | | | | | | | - Sandra Cobo
- UMR3145 CNRS Bio-Rad, SysDiag, Montpellier, France
| | | | - Max Saussine
- Vascular Surgery Department, Arnaud de Villeneuve Hospital, CHU Montpellier, France
| | - Charles Marty-Ané
- Vascular Surgery Department, Arnaud de Villeneuve Hospital, CHU Montpellier, France
| | - Bernard Albat
- Vascular Surgery Department, Arnaud de Villeneuve Hospital, CHU Montpellier, France
| | | | | | - Agnes Pouzet
- Bio-Rad Laboratories, Marnes la Coquette, France
| | | | - Daniel Laune
- UMR3145 CNRS Bio-Rad, SysDiag, Montpellier, France
| | | |
Collapse
|
50
|
Luo C, Wang H, Chen X, Cui Y, Li H, Long J, Mo X, Liu J. Protection of H9c2 rat cardiomyoblasts against oxidative insults by total paeony glucosides from Radix Paeoniae Rubrae. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 21:20-24. [PMID: 24035226 DOI: 10.1016/j.phymed.2013.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/02/2013] [Accepted: 08/02/2013] [Indexed: 06/02/2023]
Abstract
Total paeony glucosides (TPG) extracted from the roots of Radix Paeoniae Rubrae, have been approved for the therapy of rheumatoid arthritis by the State Food and Drug Administration. We previously demonstrated the myocardial protective effects of TPG in both isoprenaline-induced myocardial ischemia rat and acute myocardial infarction rat. However, the underlying mechanism of TPG effect in cardiomyocytes remains to be investigated. The aims of this study were to elucidate the effect of TPG on the activities of antioxidant defense targets and the bioenergetic system in rat cardiomyocytes. The changes of viability, antioxidant defense system activities, protein contents, and mitochondrial functions in tert-butyl hydroperoxide challenged H9c2 rat cardiomyoblasts were evaluated. The results suggest that TPG ameliorated cardiomyoblast dysfunction by preserving antioxidant defense and bioenergetic system.
Collapse
Affiliation(s)
- Cheng Luo
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an 710049, China
| | | | | | | | | | | | | | | |
Collapse
|