1
|
Sun H, Meng Y, Yao L, Du S, Li Y, Zhou Q, Liu Y, Dian Y, Sun Y, Wang X, Liang X, Deng G, Chen X, Zeng F. Ubiquitin-specific protease 22 controls melanoma metastasis and vulnerability to ferroptosis through targeting SIRT1/PTEN/PI3K signaling. MedComm (Beijing) 2024; 5:e684. [PMID: 39135915 PMCID: PMC11318338 DOI: 10.1002/mco2.684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/15/2024] Open
Abstract
Metastasis is a major contributing factor that affects the prognosis of melanoma patients. Nevertheless, the underlying molecular mechanisms involved in melanoma metastasis are not yet entirely understood. Here, we identified ubiquitin-specific protease 22 (USP22) as a pro-oncogenic protein in melanoma through screening the survival profiles of 52 ubiquitin-specific proteases (USPs). USP22 demonstrates a strong association with poor clinical outcomes and is significantly overexpressed in melanoma. Ablation of USP22 expression remarkably attenuates melanoma migration, invasion, and epithelial-mesenchymal transition in vitro and suppresses melanoma metastasis in vivo. Mechanistically, USP22 controls melanoma metastasis through the SIRT1/PTEN/PI3K pathway. In addition, we conducted an United States Food and Drug Administration-approved drug library screening and identified topotecan as a clinically applicable USP22-targeting molecule by promoting proteasomal degradation of USP22. Finally, we found that both pharmacological and genetic silence of USP22 sensitize RSL3-induced ferroptosis through suppressing the PI3K/Akt/mTOR pathway and its downstream SCD, and ferroptosis inhibitor could partly rescued the decreased lung metastasis by topotecan in vivo. Overall, our findings reveal a prometastatic role of USP22 and identify topotecan as a potent USP22-targeting drug to limit melanoma metastasis.
Collapse
Affiliation(s)
- Huiyan Sun
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
- Department of Breast ReconstructionTianjin Medical UniversityCancer Institute and HospitalTianjinChina
| | - Yu Meng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Lei Yao
- Department of Liver SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Songtao Du
- Department of Colorectal Surgical OncologyThe Tumor Hospital of Harbin Medical UniversityHarbinChina
| | - Yayun Li
- Department of DermatologyThe Third Xiangya Hospital Central South UniversityChangshaChina
| | - Qian Zhou
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yihuang Liu
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yating Dian
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yuming Sun
- Department of Plastic and Cosmetic SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiaomin Wang
- Department of Breast SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiao‐wei Liang
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Guangtong Deng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Xiang Chen
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Furong Zeng
- Department of OncologyXiangya Hospital Central South UniversityChangshaChina
| |
Collapse
|
2
|
Sharma S, Gali S, Kundu A, Park JH, Kim JS, Kim HS. Tenovin-1, a Selective SIRT1/2 Inhibitor, Attenuates High-fat Diet-induced Hepatic Fibrosis via Inhibition of HSC Activation in ZDF Rats. Int J Biol Sci 2024; 20:3334-3352. [PMID: 38993557 PMCID: PMC11234213 DOI: 10.7150/ijbs.97304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 07/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of non-alcoholic fatty liver disease (NAFLD) progression to advanced stages, especially upon high-fat diet (HFD). HFD-induced hepatic fibrosis can be marked by oxidative stress, inflammation, and activation of hepatic stellate cells. Sirtuin 1/2 (SIRT1/2), NAD-dependent class III histone deacetylases, are involved in attenuation of fibrosis. In our conducted research, TGF-β1-activated LX-2 cells, free fatty acid (FFA)-treated simultaneous co-culture (SCC) cells, and HFD-induced hepatic fibrosis in Zucker diabetic fatty (ZDF) rats, a widely used animal model in the study of metabolic syndromes, were used to evaluate the protective effect of Tenovin-1, a SIRT1/2 inhibitor. ZDF rats were divided into chow diet, HFD, and HFD + Tenovin-1 groups. Tenovin-1 reduced hepatic damage, inhibited inflammatory cell infiltration, micro/ macro-vesicular steatosis and prevented collagen deposition HFD-fed rats. Tenovin-1 reduced serum biochemical parameters, triglyceride (TG) and malondialdehyde (MDA) levels but increased glutathione, catalase, and superoxide dismutase levels. Tenovin-1 mitigated proinflammatory cytokines IL-6, IL-1β, TNFα and fibrosis biomarkers in HFD rats, TGF-β1-activated LX-2 and FFA treated SCC cells. Additionally, Tenovin-1 suppressed SIRT1/2 expression and inhibited JNK-1 and STAT3 phosphorylation in HFD rats and FFA-treated SCC cells. In conclusion, Tenovin-1 attenuates hepatic fibrosis by stimulating antioxidants and inhibiting inflammatory cytokines under HFD conditions in diabetic rats.
Collapse
Affiliation(s)
- Swati Sharma
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Amit Kundu
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam-530045, Andhra Pradesh, India
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Jae-Sung Kim
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| |
Collapse
|
3
|
Noh M, Sim JY, Kim J, Ahn JH, Min HY, Lee JU, Park JS, Jeong JY, Lee JY, Lee SY, Lee HJ, Park CS, Lee HY. Particulate matter-induced metabolic recoding of epigenetics in macrophages drives pathogenesis of chronic obstructive pulmonary disease. JOURNAL OF HAZARDOUS MATERIALS 2024; 464:132932. [PMID: 37988864 DOI: 10.1016/j.jhazmat.2023.132932] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/19/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a group of illnesses associated with unresolved inflammation in response to toxic environmental stimuli. Persistent exposure to PM is a major risk factor for COPD, but the underlying mechanism remains unclear. Using our established mouse model of PM-induced COPD, we find that repeated PM exposure provokes macrophage-centered chronic inflammation and COPD development. Mechanistically, chronic PM exposure induces transcriptional downregulation of HAAO, KMO, KYNU, and QPRT in macrophages, which are the enzymes of de novo NAD+ synthesis pathway (kynurenine pathway; KP), via elevated chromatin binding of the CCCTC-binding factor (CTCF) near the transcriptional regulatory regions of the enzymes. Subsequent reduction of NAD+ and SIRT1 function increases histone acetylation, resulting in elevated expression of pro-inflammatory genes in PM-exposed macrophages. Activation of SIRT1 by nutraceutical resveratrol mitigated PM-induced chronic inflammation and COPD development. In agreement, increased levels of histone acetylation and decreased expression of KP enzymes were observed in pulmonary macrophages of COPD patients. We newly provide an evidence that dysregulated NAD+ metabolism and consecutive SIRT1 deficiency significantly contribute to the pathological activation of macrophages during PM-mediated COPD pathogenesis. Additionally, targeting PM-induced intertwined metabolic and epigenetic reprogramming in macrophages is an effective strategy for COPD treatment.
Collapse
Affiliation(s)
- Myungkyung Noh
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jeong Yeon Sim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jisung Kim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Natural Products Research Institute, Seoul National University, Seoul 08826, South Korea
| | - Jong-Uk Lee
- Department of Medical Bioscience, Graduate School, Soonchunhyang University, 22, Soonchunhyang-ro, Asan 31538, South Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Jae Young Lee
- Department of Environmental and Safety Engineering, Ajou University, Suwon 16499, South Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Gyeonggi do, South Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
4
|
Li X, Chang Y, Shen W, Huang G, Hu N, Lv H, Jin M. miR-138 from ADSC Exo accelerates wound healing by targeting SIRT1/PTEN pathway to promote angiogenesis and fibrosis. Cell Signal 2023; 111:110843. [PMID: 37544635 DOI: 10.1016/j.cellsig.2023.110843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Affiliation(s)
- Xue Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuzhen Chang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Weijun Shen
- Department of Anesthesiology, Tenth People's Hospital of Tongji University, No 301 Middle Yan Chang Road, Shanghai 200072, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Nan Hu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, 21000, Jiangsu, China.
| | - Haihong Lv
- Department of endocrinology, The First Hospital of Lanzhou University, #1 Donggang West Road Road, Lanzhou, 730000, Gansu, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
5
|
Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, Chen N. The regulation of PTEN: Novel insights into functions as cancer biomarkers and therapeutic targets. J Cell Physiol 2023; 238:1693-1715. [PMID: 37334436 DOI: 10.1002/jcp.31053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
This review summarizes the implications of the primary tumor suppressor protein phosphatase and tensin homolog (PTEN) in aggressive cancer development. PTEN interacts with other cellular proteins or factors suggesting the existence of an intricate molecular network that regulates their oncogenic function. Accumulating evidence has shown that PTEN exists and plays a role in the cytoplasmic organelles and in the nucleus. PTEN blocks phosphoinositide 3-kinases (PI3K)-protein kinase B-mammalian target of rapamycin signaling pathway by dephosphorylating phosphatidylinositol (PI)-3,4,5-triphosphate to PI-4,5-bisphosphate thus counteracting PI3K function. Studies have shown that PTEN expression is tightly regulated at transcriptional, posttranscriptional, and posttranslational levels (including protein-protein interactions and posttranslational modifications). Despite recent advances in PTEN research, the regulation and function of the PTEN gene remain largely unknown. How mutation or loss of specific exons in the PTEN gene occurs and involves in cancer development is not clear. This review illustrates the regulatory mechanisms of PTEN expression and discusses how PTEN participates in tumor development and/or suppression. Future prospects for the clinical applications are also highlighted.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yongli Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yuheng Liu
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoping Hu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Nan Chen
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
6
|
Langdon CG. Nuclear PTEN's Functions in Suppressing Tumorigenesis: Implications for Rare Cancers. Biomolecules 2023; 13:biom13020259. [PMID: 36830628 PMCID: PMC9953540 DOI: 10.3390/biom13020259] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) encodes a tumor-suppressive phosphatase with both lipid and protein phosphatase activity. The tumor-suppressive functions of PTEN are lost through a variety of mechanisms across a wide spectrum of human malignancies, including several rare cancers that affect pediatric and adult populations. Originally discovered and characterized as a negative regulator of the cytoplasmic, pro-oncogenic phosphoinositide-3-kinase (PI3K) pathway, PTEN is also localized to the nucleus where it can exert tumor-suppressive functions in a PI3K pathway-independent manner. Cancers can usurp the tumor-suppressive functions of PTEN to promote oncogenesis by disrupting homeostatic subcellular PTEN localization. The objective of this review is to describe the changes seen in PTEN subcellular localization during tumorigenesis, how PTEN enters the nucleus, and the spectrum of impacts and consequences arising from disrupted PTEN nuclear localization on tumor promotion. This review will highlight the immediate need in understanding not only the cytoplasmic but also the nuclear functions of PTEN to gain more complete insights into how important PTEN is in preventing human cancers.
Collapse
Affiliation(s)
- Casey G. Langdon
- Department of Pediatrics, Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; ; Tel.: +1-(843)-792-9289
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
7
|
Barzegari A, Omidi Y, Gueguen V, Meddahi-Pellé A, Letourneur D, Pavon-Djavid G. Nesting and fate of transplanted stem cells in hypoxic/ischemic injured tissues: The role of HIF1α/sirtuins and downstream molecular interactions. Biofactors 2023; 49:6-20. [PMID: 32939878 DOI: 10.1002/biof.1674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022]
Abstract
The nesting mechanisms and programming for the fate of implanted stem cells in the damaged tissue have been critical issues in designing and achieving cell therapies. The fracture site can induce senescence or apoptosis based on the surrounding harsh conditions, hypoxia, and oxidative stress (OS). Respiration deficiency, disruption in energy metabolism, and consequently OS induction change the biophysical, biochemical, and cellular components of the native tissue. Additionally, the homeostatic molecular players and cell signaling might be changed. Despite all aforementioned issues, in the native stem cell niche, physiological hypoxia is not toxic; rather, it is vitally required for homing, self-renewal, and differentiation. Hence, the key macromolecular players involved in the support of stem cell survival and re-adaptation to a new dysfunctional niche must be understood for managing the cell therapy outcome. Hypoxia-inducible factor 1-alpha is the master transcriptional regulator, involved in the cell response to hypoxia and the adaptation of stem cells to a new niche. This protein is regulated by interaction with sirtuins. Sirtuins are highly conserved NAD+-dependent enzymes that monitor the cellular energy status and modulate gene transcription, genome stability, and energy metabolism in response to environmental signals to modulate the homing and fate of stem cells. Herein, new insights into the nesting of stem cells in hypoxic-ischemic injured tissues were provided and their programming in a new dysfunctional niche along with the involved complex macromolecular players were critically discussed.
Collapse
Affiliation(s)
- Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
| | - Virginie Gueguen
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| | - Anne Meddahi-Pellé
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| | - Didier Letourneur
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| | - Graciela Pavon-Djavid
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| |
Collapse
|
8
|
Perevalova AM, Kobelev VS, Sisakyan VG, Gulyaeva LF, Pustylnyak VO. Role of Tumor Suppressor PTEN and Its Regulation in Malignant Transformation of Endometrium. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1310-1326. [PMID: 36509719 DOI: 10.1134/s0006297922110104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tumor-suppressive effects of PTEN are well-known, but modern evidence suggest that they are not limited to its ability to inhibit pro-oncogenic PI3K/AKT signaling pathway. Features of PTEN structure facilitate its interaction with substrates of different nature and display its activity in various ways both in the cytoplasm and in cell nuclei, which makes it possible to take a broader look at its ability to suppress tumor growth. The possible mechanisms of the loss of PTEN effects are also diverse - PTEN can be regulated at many levels, leading to change in the protein activity or its amount in the cell, while their significance for the development of malignant tumors has yet to be studied. Here we summarize the current data on the PTEN structure, its functions and changes in its regulatory mechanisms during malignant transformation of the cells, focusing on one of the most sensitive to the loss of PTEN types of malignant tumors - endometrial cancer.
Collapse
Affiliation(s)
| | - Vyacheslav S Kobelev
- Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| | - Virab G Sisakyan
- Novosibirsk Regional Oncology Center, Novosibirsk, 630108, Russia
| | - Lyudmila F Gulyaeva
- Novosibirsk State University, Novosibirsk, 630090, Russia.,Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| | - Vladimir O Pustylnyak
- Novosibirsk State University, Novosibirsk, 630090, Russia.,Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| |
Collapse
|
9
|
Moshfegh C, Rambow SG, Domenig SA, Pieńkowska-Schelling A, Bleul U, Vogel V. Differentiation of mouse embryonic stem cells into cells with spermatogonia-like morphology with chemical intervention-dependent increased gene expression of LIM homeobox 1 (Lhx1). Stem Cell Res 2022; 61:102780. [PMID: 35395624 DOI: 10.1016/j.scr.2022.102780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022] Open
Abstract
Spermatogonial stem cells (SSCs) originate from gonocytes that differentiate from primordial germ cells (PGCs). In the developing mouse testis, expression of the gene LIM homeobox 1 (Lhx1) marks the most undifferentiated SSCs, which has not yet been reported for spermatogonia-like cells generated in vitro. Previously, it was shown that a chemical intervention in male mouse embryonic stem (ES) cells in serum culture, including Sirtuin 1 (SIRT1) inhibitor Ex-527, DNA methyltransferase (DNMT) inhibitor RG-108 and electrophilic redox cycling compound tert-butylhydroquinone (tBHQ), was associated with molecular markers of PGC to gonocyte differentiation. Here, we report the in vitro differentiation of male mouse ES cells, cultured under dual chemical inhibition of GSK3β and MEK (2i) with leukemia inhibitory factor (LIF) (2iL) and serum, into cells with spermatogonia-like morphology (CSMs) and population-averaged expression of spermatogonia-specific genes by removal of 2iL and a specific schedule of twice daily partial medium replacement. Combination of this new protocol with the previously reported chemical intervention increased population-averaged gene expression of Lhx1 in the resulting CSMs. Furthermore, we detected single CSMs with strong nuclear LHX1/5 protein signal only in the chemical intervention group. We propose that further investigation of CSMs may provide new insights into male germline development.
Collapse
Affiliation(s)
- Cameron Moshfegh
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Switzerland.
| | - Sebastian G Rambow
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Seraina A Domenig
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Aldona Pieńkowska-Schelling
- Clinic of Reproductive Medicine, Department of Farm Animals, Vetsuisse Faculty, University of Zurich, Switzerland; Institute of Genetics, Vetsuisse Faculty, University of Bern, Switzerland
| | - Ulrich Bleul
- Clinic of Reproductive Medicine, Department of Farm Animals, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| |
Collapse
|
10
|
Cross-Talking Pathways of Forkhead Box O1 (FOXO1) Are Involved in the Pathogenesis of Alzheimer’s Disease and Huntington’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7619255. [PMID: 35154571 PMCID: PMC8831070 DOI: 10.1155/2022/7619255] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 12/18/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) and Huntington's disease (HD) are destructive worldwide diseases. Efforts have been made to elucidate the process of these two diseases, yet the pathogenesis remains elusive as it involves a combination of multiple factors, including genetic and environmental ones. To explore the potential role of forkhead box O1 (FOXO1) in the development of AD and HD, we identified 1,853 differentially expressed genes (DEGs) from 19,414 background genes in both the AD&HD/control and FOXO1-low/high groups. Four coexpression modules were predicted by the weighted gene coexpression network analysis (WGCNA), among which blue and turquoise modules had the strongest correlation with AD&HD and high expression of FOXO1. Functional enrichment analysis showed that DEGs in these modules were enriched in phagosome, cytokine-cytokine receptor interaction, cellular senescence, FOXO signaling pathway, pathways of neurodegeneration, GABAergic synapse, and AGE-RAGE signaling pathway in diabetic complications. Furthermore, the cross-talking pathways of FOXO1 in AD and HD were jointly determined in a global regulatory network, such as the FOXO signaling pathway, cellular senescence, and AGE-RAGE signaling pathway in diabetic complications. Based on the performance evaluation of the area under the curve of 85.6%, FOXO1 could accurately predict the onset of AD and HD. We then identified the cross-talking pathways of FOXO1 in AD and HD, respectively. More specifically, FOXO1 was involved in the FOXO signaling pathway and cellular senescence in AD; correspondingly, FOXO1 participated in insulin resistance, insulin, and the FOXO signaling pathways in HD. Next, we use GSEA to validate the biological processes in AD&HD and FOXO1 expression. In GSEA analysis, regulation of protein maturation and regulation of protein processing were both enriched in the AD&HD and FOXO1-high groups, suggesting that FOXO1 may have implications in onset and progression of these two diseases through protein synthesis. Consequently, a high expression of FOXO1 is a potential pathogenic factor in both AD and HD involving mechanisms of the FOXO signaling pathway, AGE-RAGE signaling pathway in diabetic complications, and cellular senescence. Our findings provide a comprehensive perspective on the molecular function of FOXO1 in the pathogenesis of AD and HD.
Collapse
|
11
|
Lou YY, Zheng X, Huang YP, Mu L, Zhang XG, Zhao ZW, Song Z, Zhang J, Yin ZQ, Pan K. New dammarane-type triterpenoid saponins from Gynostemma pentaphyllum and their Sirt1 agonist activity. Bioorg Chem 2021; 116:105357. [PMID: 34562675 DOI: 10.1016/j.bioorg.2021.105357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/12/2021] [Indexed: 12/12/2022]
Abstract
Gynostemma pentaphyllum (Thunb.) Makino (Cucurbitaceae family) is a perennial creeping plant with a common Chinese name of "south ginseng". To date, more than 250 individual saponins with dammarane-type skeleton have been isolated from G. pentaphyllum. The purpose of this study was the isolation and structural characterization of novel, minor gypenosides from G. pentaphyllum and evaluation of their Sirt1 agonist activity. Individual saponins from G. pentaphyllum were isolated and purified by a variety of chromatography techniques, and their structures were elucidated by means of various spectroscopic analysis and comparision with the reported data. Sirt1 enzyme activity detection kit was used to preliminarily evaluate the Sirt1 agonist activity of thirty three individual saponins purified from G. pentaphyllum. Fourteen new triterpenoid saponins named gypenoside CII-CXV (1-14) along with twenty six known compounds (15-40) were isolated from G. pentaphyllum. Thirty three of all the isolates were screened for Sirt1 agonist activity, and the results showed that three dammarane-type saponins (2, 18, 37) and one cucurbitane-type saponin 33 exhibited satisfactory Sirt1 agonist activity. These findings suggested that G. pentaphyllum was worthy of further investigation to find small molecule Sirt1 agonist and facilitate their utilization as "south ginseng".
Collapse
Affiliation(s)
- Yun-Yun Lou
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China; Laboratory of Translational Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Xian Zheng
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China; Laboratory of Translational Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Ya-Ping Huang
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China
| | - Long Mu
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiao-Gai Zhang
- Department of Endocrinology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211200, PR China
| | - Zhi-Wei Zhao
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China; Laboratory of Translational Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Zhe Song
- Instrumental Analysis Center of CPU, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China
| | - Jian Zhang
- Laboratory of Translational Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China.
| | - Zhi-Qi Yin
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Ke Pan
- Department of TCMs Pharmaceuticals & Department of Natural Medicinal Chemistry, School of TCM, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
12
|
Fangma Y, Zhou H, Shao C, Yu L, Yang J, Wan H, He Y. Hydroxysafflor Yellow A and Anhydrosafflor Yellow B Protect Against Cerebral Ischemia/Reperfusion Injury by Attenuating Oxidative Stress and Apoptosis via the Silent Information Regulator 1 Signaling Pathway. Front Pharmacol 2021; 12:739864. [PMID: 34658877 PMCID: PMC8514692 DOI: 10.3389/fphar.2021.739864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/11/2023] Open
Abstract
Hydroxysafflor yellow A (HSYA) and anhydrosafflor yellow B (AHSYB) are the main water-soluble compounds in Carthamus tinctorius L. However, studies on the effect of AHSYB on cerebral ischemia/reperfusion (I/R) injury and the therapeutic effect of HSYA by regulating silent information regulator 1 (SIRT1) pathway remain obscure. In this study, we investigated whether the neuroprotective effects of HSYA and AHSYB on oxygen-glucose deprivation/reoxygenation in primary-cultured hippocampal neuronal cells and the middle cerebral artery occlusion and reperfusion model in rats are associated with the regulation of the SIRT1 pathway. In vitro, HSYA and AHSYB increased cell viability, depressed oxidation properties, and reduced neuronal cell apoptosis. In vivo results showed that HSYA and AHSYB effectively reduced infarct volume, improved neurological function, suppressed apoptosis, and decreased the oxidative stress reaction. Besides, RT-PCR and Western blot analysis showed that HSYA and AHSYB increased the mRNA and protein expressions of the main factors in the SIRT1 pathway, including SIRT1, forkhead box O (FOXO) 1, and peroxisome proliferator–activated receptor coactivator 1α (PGC1α), decreased the expression of Bax, and increased the expression of Bcl-2. The results from immunohistochemistry also showed that the expressions of SIRT1, FOXO1, and PGC1α were increased after treatment with HSYA and AHSYB. Furthermore, the neuroprotective effects of HSYA and AHSYB were abolished by EX527 (SIRT1–specific inhibitor). These results indicated that HSYA and AHSYB should be developed into potential drugs for treating cerebral I/R injury via the SIRT1 pathway. Although HSYA and AHSYB have different chemical structures, both of them exert similar neuroprotective properties against I/R injury in vitro and in vivo, which means that AHSYB is also a non-negligible component in safflower.
Collapse
Affiliation(s)
- Yijia Fangma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huifen Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chongyu Shao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
13
|
Fu L, Han BK, Meng FF, Wang JW, Wang TY, Li HJ, Sun YY, Zou GN, Li XR, Li W, Bi YF, Ke Y, Liu HM. Jaridon 6, a new diterpene from Rabdosia rubescens (Hemsl.) Hara, can display anti-gastric cancer resistance by inhibiting SIRT1 and inducing autophagy. Phytother Res 2021; 35:5720-5733. [PMID: 34411362 DOI: 10.1002/ptr.7231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/20/2021] [Accepted: 06/29/2021] [Indexed: 11/10/2022]
Abstract
Tumor resistance is the main cause of treatment failure and is associated with many tumor factors. Jaridon 6, a new diterpene extracted from Rabdosia rubescens (Hemsl.) Hara, which has been previously extracted by our research team, has been tested having more obvious advantages in resistant tumor cells. However, its mechanism is unclear. In this study, we studied the effect and the specific mechanism of Jaridon 6 in resistant gastric cancer cells. Cytotoxicity test, colony test, western blotting, and nude test verified the anti-drug resistance ability of Jaridon 6 in the MGC803/PTX and MGC803/5-Fu cells. Jaridon 6 has shown obvious inhibitory effects in the sirtuin 1 (SIRT1) enzyme test. Transmission electron microscopy and immunofluorescence tests further proved the autophagic action of Jaridon 6. Jaridon 6 could inhibit the proliferation of the resistant gastric cancer cell in vivo and in vitro. Jaridon 6 inhibited SIRT1 enzyme and induced autophagy by inhibiting the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway. Thus, it may be considered for treating gastric cancer resistance by individual or combined administration, as an SIRT1 inhibitor and autophagy inducer.
Collapse
Affiliation(s)
- Ling Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Bing-Kai Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Fang-Feng Meng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Jun-Wei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Tian-Ye Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Hui-Ju Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Ying-Ying Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Guo-Na Zou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Xiao-Rui Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Wen Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Yue-Feng Bi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
14
|
Nguyen Huu T, Park J, Zhang Y, Park I, Yoon HJ, Woo HA, Lee SR. Redox Regulation of PTEN by Peroxiredoxins. Antioxidants (Basel) 2021; 10:antiox10020302. [PMID: 33669370 PMCID: PMC7920247 DOI: 10.3390/antiox10020302] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is known as a tumor suppressor gene that is frequently mutated in numerous human cancers and inherited syndromes. PTEN functions as a negative regulator of PI3K/Akt signaling pathway by dephosphorylating phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3) to phosphatidylinositol (4, 5)-bisphosphate (PIP2), which leads to the inhibition of cell growth, proliferation, cell survival, and protein synthesis. PTEN contains a cysteine residue in the active site that can be oxidized by peroxides, forming an intramolecular disulfide bond between Cys124 and Cys71. Redox regulation of PTEN by reactive oxygen species (ROS) plays a crucial role in cellular signaling. Peroxiredoxins (Prxs) are a superfamily of peroxidase that catalyzes reduction of peroxides and maintains redox homeostasis. Mammalian Prxs have 6 isoforms (I-VI) and can scavenge cellular peroxides. It has been demonstrated that Prx I can preserve and promote the tumor-suppressive function of PTEN by preventing oxidation of PTEN under benign oxidative stress via direct interaction. Also, Prx II-deficient cells increased PTEN oxidation and insulin sensitivity. Furthermore, Prx III has been shown to protect PTEN from oxidation induced by 15s-HpETE and 12s-HpETE, these are potent inflammatory and pro-oxidant mediators. Understanding the tight connection between PTEN and Prxs is important for providing novel therapies. Herein, we summarized recent studies focusing on the relationship of Prxs and the redox regulation of PTEN.
Collapse
Affiliation(s)
- Thang Nguyen Huu
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
| | - Jiyoung Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea;
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China;
| | - Iha Park
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
| | - Hyun Joong Yoon
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea;
- Correspondence: (H.A.W.); (S.-R.L.); Tel.: +82-2-3277-4654 (H.A.W.); +82-61-379-2775 (S.-R.L.); Fax: +82-2-3277-3760 (H.A.W.); +82-61-379-2782 (S.-R.L.)
| | - Seung-Rock Lee
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
- Correspondence: (H.A.W.); (S.-R.L.); Tel.: +82-2-3277-4654 (H.A.W.); +82-61-379-2775 (S.-R.L.); Fax: +82-2-3277-3760 (H.A.W.); +82-61-379-2782 (S.-R.L.)
| |
Collapse
|
15
|
Wang T, Wang Y, Liu L, Jiang Z, Li X, Tong R, He J, Shi J. Research progress on sirtuins family members and cell senescence. Eur J Med Chem 2020; 193:112207. [PMID: 32222662 DOI: 10.1016/j.ejmech.2020.112207] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/13/2020] [Accepted: 03/04/2020] [Indexed: 02/05/2023]
Abstract
Human aging is a phenomenon of gradual decline and loss of cell, tissue, organ and other functions under the action of external environment and internal factors. It is mainly related to genomic instability, telomere wear, mitochondrial dysfunction, protein balance disorder, antioxidant damage, microRNA expression disorder and so on. Sirtuins protein is a kind of deacetylase which can regulate cell metabolism and participate in a variety of cell physiological functions. It has been found that sirtuins family can prolong the lifespan of yeast. Sirtuins can inhibit human aging through many signaling pathways, including apoptosis signaling pathway, mTOR signaling pathway, sirtuins signaling pathway, AMPK signaling pathway, phosphatidylinositol 3 kinase (PI3K) signaling pathway and so on. Based on this, this paper reviews the action principle of anti-aging star members of sirtuins family Sirt1, Sirt3 and Sirt6 on anti-aging related signaling pathways and typical compounds, in order to provide ideas for the screening of anti-aging compounds of sirtuins family members.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yujue Wang
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Li Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Xingxing Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun He
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Jianyou Shi
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
16
|
Li H, Bai Z, Li C, Sheng C, Zhao X. EV71 infection induces cell apoptosis through ROS generation and SIRT1 activation. J Cell Biochem 2020; 121:4321-4331. [PMID: 31898369 DOI: 10.1002/jcb.29628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022]
Abstract
Several studies have substantiated the correlation between reactive oxygen species (ROS) and Sirtuin 1 (SIRT1). Normally, enterovirus 71 (EV71) is associated with severe clinical manifestations and death. However, the effect of EV71 on the induction of cellular death and the interplay between ROS/SIRT1 in cell death has not been confirmed yet. In the current study, an increase in the number of apoptotic cells was observed as soon as the EV71 infection was initiated in cells and mice. Furthermore, EV71 infection also promoted a rise in the levels of three commonly known proinflammatory cytokines, interleukin 1β (IL-1β), IL-6, and tumor necrosis factor-α. During EV71-induced apoptosis in the different cell lines, ROS generation and SIRT1 downregulation were observed. Further investigations showed that the administration of ROS inhibitor, N-acetyl- l-cysteine (NAC), reduced the level of apoptosis and inflammation, reduced EV71 propagation, and increased SIRT1 expression in EV71-infected cells. In addition, combined administration of NAC and EX527 (SIRT1 inhibitor) restored apoptosis in the EV71-infected cells, which was reduced due to NAC. This data demonstrated that ROS generation is positively associated with EV71-induced apoptosis and inflammation, while this effect could be reversed by SIRT1 inhibition. Collectively, we have shown that EV71 induces apoptosis and inflammation by promoting ROS generation and reducing SIRT1 expression.
Collapse
Affiliation(s)
- Hongyan Li
- Infectious Department, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zhenzi Bai
- Infectious Department, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chenghua Li
- Infectious Department, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chuanlun Sheng
- Infectious Department, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiaonan Zhao
- Infectious Department, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
17
|
Heshmati M, Soltani A, Sanaei MJ, Nahid-Samiei M, Shirzad H, Jami MS, GhatrehSamani M. Ghrelin induces autophagy and CXCR4 expression via the SIRT1/AMPK axis in lymphoblastic leukemia cell lines. Cell Signal 2019; 66:109492. [PMID: 31809874 DOI: 10.1016/j.cellsig.2019.109492] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/25/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is one of the most frequent malignancies in children, and the CXCR4 receptor plays an important role in the metastasis of this malignancy. Ghrelin is a hormone with various functions including stimulation of the release of growth hormone and autophagy in cancer cells. Moreover, SIRT1 and AMPK (AMP-activated protein kinase) stimulate expression of proteins involved in autophagy. On the other hand, autophagic cell death can be an alternative target for cancer therapy, in the absence of apoptosis. The relationship between ghrelin and the SIRT1/AMPK axis and the resulting effects on autophagy, apoptosis, proliferation, and expression of CXCR4 and the ghrelin receptor (GHS-R1a), in Jurkat and Molt-4 human lymphoblastic cell lines was not previously clear. Here we demonstrate that SIRT1 expression is upregulated during the induction of autophagy by ghrelin, an effect that is inhibited by inactivation of SIRT1/AMPK axis. In addition, ghrelin can affect CXCR4 and GHS-R1a expression. In conclusion, this work reveals that ghrelin induces autophagy, invasion, and downregulation of ghrelin receptor expression via the SIRT1/AMPK axis in lymphoblastic cell lines. However, in these cell lines ghrelin-induced autophagy does not lead to cell death due to weak induction of apoptosis.
Collapse
Affiliation(s)
- Masoud Heshmati
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Javad Sanaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahboobeh Nahid-Samiei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Saeid Jami
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Mahdi GhatrehSamani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
18
|
Calycosin-7- O- β- D-glucoside Attenuates OGD/R-Induced Damage by Preventing Oxidative Stress and Neuronal Apoptosis via the SIRT1/FOXO1/PGC-1 α Pathway in HT22 Cells. Neural Plast 2019; 2019:8798069. [PMID: 31885537 PMCID: PMC6915014 DOI: 10.1155/2019/8798069] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/02/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
Neuronal apoptosis induced by oxidative stress is a major pathological process that occurs after cerebral ischemia-reperfusion. Calycosin-7-O-β-D-glucoside (CG) is a representative component of isoflavones in Radix Astragali (RA). Previous studies have shown that CG has potential neuroprotective effects. However, whether CG alleviates neuronal apoptosis through antioxidant stress after ischemia-reperfusion remains unknown. To investigate the positive effects of CG on oxidative stress and apoptosis of neurons, we simulated the ischemia-reperfusion process in vitro using an immortalized hippocampal neuron cell line (HT22) and oxygen-glucose deprivation/reperfusion (OGD/R) model. CG significantly improved cell viability and reduced oxidative stress and neuronal apoptosis. In addition, CG treatment upregulated the expression of SIRT1, FOXO1, PGC-1α, and Bcl-2 and downregulated the expression of Bax. In summary, our findings indicate that CG alleviates OGD/R-induced damage via the SIRT1/FOXO1/PGC-1α signaling pathway. Thus, CG maybe a promising therapeutic candidate for brain injury associated with ischemic stroke.
Collapse
|
19
|
Shafieian Z, Bahari G, Hashemi M, Nakhaee A. SIRT1 Gene Polymorphisms Are Associated with Urinary Bladder Cancer in an Iranian Population. Rep Biochem Mol Biol 2019; 8:194-199. [PMID: 31832445 PMCID: PMC6844609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 01/21/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND The present study was undertaken to evaluate the possible association between silent information regulator of transcription 1 gene (SIRT 1) polymorphisms and risk of urinary bladder cancer (UBC) in an Iranian population. METHODS The SIRT1 polymorphisms rs3758391 T/C and rs369274325 G/A were evaluated in 120 Iranian bladder cancer patients and 118 healthy individuals as the control group. The SIRT1 rs369274325 G/A and rs3758391 T/C polymorphisms were genotyped using tetra-primer ARMS PCR and PCR-RFLP methods, respectively. RESULTS The SIRT1 rs3758391 TT genotype occurred significantly more frequently in the UBC patients than in the controls (13.3 vs. 1.7%) in both the additive and recessive models due to a significant difference in either of additive (TT vs. CC; OR= 9.529, P = 0.003) or recessive models (TT vs. CC + CT genotype; OR= 8.923, P = 0.002). Also, for rs369274325, the AG genotype was found in a significantly greater percentage of UBC patients than in controls (75.8 vs. 43.2%, respectively, P < 0.0001. CONCLUSION Our preliminary study suggests that SIRT1 rs3758391 T/C and rs369274325 G/A polymorphisms may confer an increased risk of bladder cancer in our patients.
Collapse
Affiliation(s)
- Zahra Shafieian
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Gholamreza Bahari
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Alireza Nakhaee
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
20
|
Qin R, Zhang L, Lin D, Xiao F, Guo L. Sirt1 inhibits HG-induced endothelial injury: Role of Mff-based mitochondrial fission and F‑actin homeostasis-mediated cellular migration. Int J Mol Med 2019; 44:89-102. [PMID: 31115479 PMCID: PMC6559301 DOI: 10.3892/ijmm.2019.4185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/18/2019] [Indexed: 12/11/2022] Open
Abstract
Although sirtuin 1 (Sirt1) has been found to be involved in diabetic vasculopathy and high glucose (HG)‑mediated endothelial injury, the underlying mechanisms remain to be fully elucidated. The aim of the present study was to investigate the role of Sirt1 in HG‑induced endothelial injury and its potential mechanism. In the present study, it was demonstrated that HG triggers the downregulation of Sirt1 by activating microRNA‑195 in human umbilical vein endothelial cells (HUVECs), as determined by western blot analysis in vivo and in vitro. Furthermore, a lower expression of Sirt1 was correlated with glucose metabolic abnormalities, aortic endothelial dysfunction and endothelial apoptosis as evidenced by western blot analysis and ELISA in mice. By contrast, the loss of Sirt1 evoked mitochondrial fission factor (Mff)‑mediated mitochondrial fission through the c‑Jun N‑terminal kinase (JNK) pathway, which contributes to the apoptosis of HUVECs. In addition, Sirt1 deficiency downregulated the migration of HUVECs through F‑actin dyshomeostasis. Collectively, the results identify Sirt1 as a protective factor, which inhibits the JNK/Mff/mitochondrial fission pathway and sustains F‑actin homeostasis, and has potential implications for novel approaches to diabetic vasculopathy.
Collapse
Affiliation(s)
- Ruijie Qin
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences
| | - Lina Zhang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology
| | - Dong Lin
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences
| | - Fei Xiao
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences
| |
Collapse
|
21
|
Ma J, Zhang Y, Ji H, Chen L, Chen T, Guo C, Zhang S, Jia J, Niu P. Overexpression of miR-138-5p suppresses MnCl 2 -induced autophagy by targeting SIRT1 in SH-SY5Y cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:539-547. [PMID: 30672645 DOI: 10.1002/tox.22708] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/24/2018] [Accepted: 12/26/2018] [Indexed: 06/09/2023]
Abstract
The mechanism of manganism caused by manganese (Mn), an important environmental risk factor for Parkinson's disease, is still unclear. Recent evidence suggested that autophagy participated in neurodegenerative diseases, in which microRNA played a crucial role. However, roles of microRNA in the aberrant autophagy that occurs in neurodegenerative diseases remains controversial. In nervous system, miRNA-138-5p is highly expressed and plays a key role in regulating memory and axon regeneration. Importantly, we also found that miR-138-5p expression decreased significantly after SH-SY5Y cells exposed to manganese chloride (MnCl2 ) in previous study. To explore the role of miR-138-5p in Mn-induced autophagy, autophagy associated indicators were detected. And we found that MnCl2 could induce autophagic dysregulation and inhibit expression of miR-138-5p. While the levels of LC3-II/LC3-I, Beclin1, and p62, the number of autophagosome formation significantly decreased after miR-138-5p over-expression, which demonstrated that miR-138-5p could clearly retard Mn-induced autophagy. In additional, we found there were classical and evolutionarily conserved miR-138-5p binding sites in 3'-UTR region of SIRT1, which was inhibited when overexpression of miR-138-5p. Therefore, it was speculated that elevated expression of SIRT1 may be resulted from inhibition of miR-138-5p after cells exposed to MnCl2 . Finally, we found that SIRT1 inhibitor EX-527 suppressed Mn-induced autophagy as well as miR-138-5p, while the suppression was reversed by SIRT1-specific activator SRT1720. These results indicated that overexpression of miR-138-5p suppressed Mn-induced autophagy by targeting SIRT1.
Collapse
Affiliation(s)
- Junxiang Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Yuanyuan Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Hongyun Ji
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Li Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Tian Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Shixuan Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Jiaxin Jia
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Tang BL. Why is NMNAT Protective against Neuronal Cell Death and Axon Degeneration, but Inhibitory of Axon Regeneration? Cells 2019; 8:cells8030267. [PMID: 30901919 PMCID: PMC6468476 DOI: 10.3390/cells8030267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide mononucleotide adenylyltransferase (NMNAT), a key enzyme for NAD+ synthesis, is well known for its activity in neuronal survival and attenuation of Wallerian degeneration. Recent investigations in invertebrate models have, however, revealed that NMNAT activity negatively impacts upon axon regeneration. Overexpression of Nmnat in laser-severed Drosophila sensory neurons reduced axon regeneration, while axon regeneration was enhanced in injured mechanosensory axons in C. elegansnmat-2 null mutants. These diametrically opposite effects of NMNAT orthologues on neuroprotection and axon regeneration appear counterintuitive as there are many examples of neuroprotective factors that also promote neurite outgrowth, and enhanced neuronal survival would logically facilitate regeneration. We suggest here that while NMNAT activity and NAD+ production activate neuroprotective mechanisms such as SIRT1-mediated deacetylation, the same mechanisms may also activate a key axonal regeneration inhibitor, namely phosphatase and tensin homolog (PTEN). SIRT1 is known to deacetylate and activate PTEN which could, in turn, suppress PI3 kinase–mTORC1-mediated induction of localized axonal protein translation, an important process that determines successful regeneration. Strategic tuning of Nmnat activity and NAD+ production in axotomized neurons may thus be necessary to promote initial survival without inhibiting subsequent regeneration.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
23
|
Álvarez-Garcia V, Tawil Y, Wise HM, Leslie NR. Mechanisms of PTEN loss in cancer: It's all about diversity. Semin Cancer Biol 2019; 59:66-79. [PMID: 30738865 DOI: 10.1016/j.semcancer.2019.02.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 01/04/2023]
Abstract
PTEN is a phosphatase which metabolises PIP3, the lipid product of PI 3-Kinase, directly opposing the activation of the oncogenic PI3K/AKT/mTOR signalling network. Accordingly, loss of function of the PTEN tumour suppressor is one of the most common events observed in many types of cancer. Although the mechanisms by which PTEN function is disrupted are diverse, the most frequently observed events are deletion of a single gene copy of PTEN and gene silencing, usually observed in tumours with little or no PTEN protein detectable by immunohistochemistry. Accordingly, with the exceptions of glioblastoma and endometrial cancer, mutations of the PTEN coding sequence are uncommon (<10%) in most types of cancer. Here we review the data relating to PTEN loss in seven common tumour types and discuss mechanisms of PTEN regulation, some of which appear to contribute to reduced PTEN protein levels in cancers.
Collapse
Affiliation(s)
- Virginia Álvarez-Garcia
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Yasmine Tawil
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Helen M Wise
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Nicholas R Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK.
| |
Collapse
|
24
|
Hsu YC, Wu YT, Tsai CL, Wei YH. Current understanding and future perspectives of the roles of sirtuins in the reprogramming and differentiation of pluripotent stem cells. Exp Biol Med (Maywood) 2019; 243:563-575. [PMID: 29557214 DOI: 10.1177/1535370218759636] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In mammalian cells, there are seven members of the sirtuin protein family (SIRT1-7). SIRT1, SIRT6, and SIRT7 catalyze posttranslational modification of proteins in the nucleus, SIRT3, SIRT4, and SIRT5 are in the mitochondria and SIRT2 is in the cytosol. SIRT1 can deacetylate the transcription factor SOX2 and regulate induced pluripotent stem cells (iPSCs) reprogramming through the miR-34a-SIRT1-p53 axis. SIRT2 can regulate the function of pluripotent stem cells through GSK3β. SIRT3 can positively regulate PPAR gamma coactivator 1-alpha (PGC-1α) expression during the differentiation of stem cells. SIRT4 has no direct role in regulating reprogramming but may have the potential to prevent senescence of somatic cells and to facilitate the reprogramming of iPSCs. SIRT5 can deacetylate STAT3, which is an important transcription factor in regulating pluripotency and differentiation of stem cells. SIRT6 can enhance the reprogramming efficiency of iPSCs from aged skin fibroblasts through miR-766 and increase the expression levels of the reprogramming genes including Sox2, Oct4, and Nanog through acetylation of histone H3 lysine 56. SIRT7 plays a regulatory role in the process of mesenchymal-to-epithelial transition (MET), which has been suggested to be a crucial process in the generation of iPSCs from fibroblasts. In this review, we summarize recent findings of the roles of sirtuins in the metabolic reprogramming and differentiation of stem cells and discuss the bidirectional changes in the gene expression and activities of sirtuins in the commitment of differentiation of mesenchymal stem cells (MSCs) and reprogramming of somatic cells to iPSCs, respectively. Thus, understanding the molecular basis of the interplay between different sirtuins and mitochondrial function will provide new insights into the regulation of differentiation of stem cells and iPSCs formation, respectively, and may help design effective stem cell therapies for regenerative medicine. Impact statement This is an extensive review of the recent advances in our understanding of the roles of some members of the sirtuins family, such as SIRT1, SIRT2, SIRT3, and SIRT6, in the regulation of intermediary metabolism during stem cell differentiation and in the reprogramming of somatic cells to form induced pluripotent stem cells (iPSCs). This article provides an updated integrated view on the mechanisms by which sirtuins-mediated posttranslational protein modifications regulate mitochondrial biogenesis, bioenergetics, and antioxidant defense in the maintenance and differentiation of stem cells and in iPSCs formation, respectively.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan.,*These two authors made equal contributions
| | - Yu-Ting Wu
- 2 Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan.,*These two authors made equal contributions
| | - Chia-Ling Tsai
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan
| | - Yau-Huei Wei
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan.,2 Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
25
|
Poly(ADP-Ribose) Polymerases in Host-Pathogen Interactions, Inflammation, and Immunity. Microbiol Mol Biol Rev 2018; 83:83/1/e00038-18. [PMID: 30567936 DOI: 10.1128/mmbr.00038-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The literature review presented here details recent research involving members of the poly(ADP-ribose) polymerase (PARP) family of proteins. Among the 17 recognized members of the family, the human enzyme PARP1 is the most extensively studied, resulting in a number of known biological and metabolic roles. This review is focused on the roles played by PARP enzymes in host-pathogen interactions and in diseases with an associated inflammatory response. In mammalian cells, several PARPs have specific roles in the antiviral response; this is perhaps best illustrated by PARP13, also termed the zinc finger antiviral protein (ZAP). Plant stress responses and immunity are also regulated by poly(ADP-ribosyl)ation. PARPs promote inflammatory responses by stimulating proinflammatory signal transduction pathways that lead to the expression of cytokines and cell adhesion molecules. Hence, PARP inhibitors show promise in the treatment of inflammatory disorders and conditions with an inflammatory component, such as diabetes, arthritis, and stroke. These functions are correlated with the biophysical characteristics of PARP family enzymes. This work is important in providing a comprehensive understanding of the molecular basis of pathogenesis and host responses, as well as in the identification of inhibitors. This is important because the identification of inhibitors has been shown to be effective in arresting the progression of disease.
Collapse
|
26
|
Naderali E, Khaki AA, Rad JS, Ali-Hemmati A, Rahmati M, Charoudeh HN. Regulation and modulation of PTEN activity. Mol Biol Rep 2018; 45:2869-2881. [PMID: 30145641 DOI: 10.1007/s11033-018-4321-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/20/2018] [Indexed: 01/04/2023]
Abstract
PTEN (Phosphatase and tensin homolog deleted on chromosome ten) is a tumor suppressor that is frequently mutated in most human cancers. PTEN is a lipid and protein phosphatase that antagonizes PI3K/AKT pathway through lipid phosphatase activity at the plasma membrane. More recent studies showed that, in addition to the putative role of PTEN as a PI(3,4,5)P3 3-phosphatase, it is a PI(3,4)P2 3-phosphatase during stimulation of class I PI3K signaling pathway by growth factor. Although PTEN tumor suppressor function via it's lipid phosphatase activity occurs primarily in the plasma membrane, it can also be found in the nucleus, in cytoplasmic organelles and extracellular space. PTEN has also shown phosphatase independent functions in the nucleus. PTEN can exit from the cell through exosomal export or secretion and has a tumor suppressor function in adjacent cells. PTEN has a critical role in growth, the cell cycle, protein synthesis, survival, DNA repair and migration. Understanding the regulation of PTEN function, activity, stability, localization and its dysregulation outcomes and also the intracellular and extracellular role of PTEN and paracrine role of PTEN-L in tumor cells as an exogenous therapeutic agent can help to improve clinical conceptualization and treatment of cancer.
Collapse
Affiliation(s)
- Elahe Naderali
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Afshin Khaki
- Department of Anatomical sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleymani Rad
- Department of Anatomical sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ali-Hemmati
- Department of Anatomical sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Rahmati
- Department of Clinical Biochemistry Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hojjatollah Nozad Charoudeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Cell Therapy Research Laboratory, Drug Applied Research Center, Tabriz University of Medical Sciences, P.O. Box: 51656-65811, Tabriz, Iran.
| |
Collapse
|
27
|
Lee YR, Chen M, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat Rev Mol Cell Biol 2018; 19:547-562. [DOI: 10.1038/s41580-018-0015-0] [Citation(s) in RCA: 399] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Yao H, Yao Z, Zhang S, Zhang W, Zhou W. Upregulation of SIRT1 inhibits H2O2‑induced osteoblast apoptosis via FoxO1/β‑catenin pathway. Mol Med Rep 2018; 17:6681-6690. [PMID: 29512706 DOI: 10.3892/mmr.2018.8657] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/18/2017] [Indexed: 11/06/2022] Open
Abstract
Osteoporosis is a disease that significantly influences life expectancy and quality in humans. Oxidative stress may stimulate bone marrow osteoclast differentiation and inhibit osteoblast (OB) differentiation. OB proliferation and differentiation are affected by the forkhead box O (FoxO)1/β‑catenin signaling pathway. The osteogenic differentiation of mesenchymal stem cells (MSCs) may be promoted by silent information regulator type‑1 [sirtuin (SIRT)1]. However, the molecular mechanism of SIRT1 regulation of osteogenic differentiation of MSCs remains unclear, and further elucidation is needed. The present study investigated the role of SIRT1 in the FoxO1/β‑catenin signaling pathway in oxidative stress and its mechanism in the osteoblastic progenitor cell line (MC3T3‑E1). The results demonstrated that OB apoptosis and elevated oxidative stress in cells were simulated by H2O2, which was inhibited by moderate SIRT1 overexpression through reducing the oxidative stress. Further studies revealed that FOXO1 and β‑catenin pathway activity was downregulated by SIRT1 and eventually resulted in inhibition of target genes, including the proapoptotic gene B cell lymphoma‑2 interacting mediator of cell death, DNA repair gene growth arrest and DNA damage inducible protein 45 and the OB differentiation suppressor gene peroxisome proliferator activated receptor (PPAR)‑γ. Furthermore, β‑catenin and PPAR‑γ were inhibited by SIRT1. Overall, the results of the present study suggest that moderate overexpression of SIRT1 (~3‑fold of normal level) may directly or indirectly inhibit apoptosis of OBs via the FOXO1 and β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Hanlin Yao
- Department of Orthopaedic, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Zhen Yao
- Department of Orthopaedic, Xinchang Hospital Affiliated to Wenzhou Medical University, Hangzhou, Zhejiang 312500, P.R. China
| | - Shaocheng Zhang
- Department of Orthopaedic, Changhai Hospital Affiliated to Second Military Medical University, Shanghai 200433, P.R. China
| | - Wenjun Zhang
- Department of Orthopaedic, Qingpu People's Hospital of Zhujiajue, Shanghai 201713, P.R. China
| | - Wen Zhou
- Department of Sports Science, Shanghai University of Sports, Shanghai 200433, P.R. China
| |
Collapse
|
29
|
Haddadi N, Lin Y, Travis G, Simpson AM, McGowan EM, Nassif NT. PTEN/PTENP1: 'Regulating the regulator of RTK-dependent PI3K/Akt signalling', new targets for cancer therapy. Mol Cancer 2018; 17:37. [PMID: 29455665 PMCID: PMC5817727 DOI: 10.1186/s12943-018-0803-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Regulation of the PI-3 kinase (PI3K)/Akt signalling pathway is essential for maintaining the integrity of fundamental cellular processes, cell growth, survival, death and metabolism, and dysregulation of this pathway is implicated in the development and progression of cancers. Receptor tyrosine kinases (RTKs) are major upstream regulators of PI3K/Akt signalling. The phosphatase and tensin homologue (PTEN), a well characterised tumour suppressor, is a prime antagonist of PI3K and therefore a negative regulator of this pathway. Loss or inactivation of PTEN, which occurs in many tumour types, leads to overactivation of RTK/PI3K/Akt signalling driving tumourigenesis. Cellular PTEN levels are tightly regulated by a number of transcriptional, post-transcriptional and post-translational regulatory mechanisms. Of particular interest, transcription of the PTEN pseudogene, PTENP1, produces sense and antisense transcripts that exhibit post-transcriptional and transcriptional modulation of PTEN expression respectively. These additional levels of regulatory complexity governing PTEN expression add to the overall intricacies of the regulation of RTK/PI-3 K/Akt signalling. This review will discuss the regulation of oncogenic PI3K signalling by PTEN (the regulator) with a focus on the modulatory effects of the sense and antisense transcripts of PTENP1 on PTEN expression, and will further explore the potential for new therapeutic opportunities in cancer treatment.
Collapse
Affiliation(s)
- Nahal Haddadi
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
| | - Yiguang Lin
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
| | - Glena Travis
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
| | - Ann M. Simpson
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
| | - Eileen M. McGowan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080 China
| | - Najah T. Nassif
- School of Life Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007 Australia
| |
Collapse
|
30
|
Shen Z, Ma Y, Ji Z, Hao Y, Yan X, Zhong Y, Tang X, Ren W. Arachidonic acid induces macrophage cell cycle arrest through the JNK signaling pathway. Lipids Health Dis 2018; 17:26. [PMID: 29426338 PMCID: PMC5807765 DOI: 10.1186/s12944-018-0673-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 02/05/2018] [Indexed: 01/14/2023] Open
Abstract
Background Arachidonic acid (AA) has potent pro-apoptotic effects on cancer cells at a low concentration and on macrophages at a very high concentration. However, the effects of AA on the macrophage cell cycle and related signaling pathways have not been fully investigated. Herein we aim to observe the effect of AA on macrophages cell cycle. Results AA exposure reduced the viability and number of macrophages in a dose- and time-dependent manner. The reduction in RAW264.7 cell viability was not caused by apoptosis, as indicated by caspase-3 and activated caspase-3 detection. Further research illustrated that AA exposure induced RAW264.7 cell cycle arrested at S phase, and some cell cycle-regulated proteins were altered accordingly. Moreover, JNK signaling was stimulated by AA, and the stimulation was partially reversed by a JNK signaling inhibitor in accordance with cell cycle-related factors. In addition, nuclear and total Foxo1/3a and phosphorylated Foxo1/3a were elevated by AA in a dose- and time-dependent manner, and this elevation was suppressed by the JNK signaling inhibitor. Conclusion Our study demonstrated that AA inhibits macrophage viability by inducing S phase cell cycle arrest. The JNK signaling pathway and the downstream FoxO transcription factors are involved in AA-induced RAW264.7 cell cycle arrest. Electronic supplementary material The online version of this article (10.1186/s12944-018-0673-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ziying Shen
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Yunqing Ma
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Zhonghao Ji
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Yang Hao
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Xuan Yan
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Yuan Zhong
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Xiaochun Tang
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China
| | - Wenzhi Ren
- Laboratory Animal Center, College of Animal Sciences, Jilin University, Xi'an Road, 5333#, Jilin, 130062, China.
| |
Collapse
|
31
|
Nakamura K, Zhang M, Kageyama S, Ke B, Fujii T, Sosa RA, Reed EF, Datta N, Zarrinpar A, Busuttil RW, Araujo JA, Kupiec-Weglinski JW. Macrophage heme oxygenase-1-SIRT1-p53 axis regulates sterile inflammation in liver ischemia-reperfusion injury. J Hepatol 2017; 67:1232-1242. [PMID: 28842295 PMCID: PMC5884687 DOI: 10.1016/j.jhep.2017.08.010] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Hepatic ischemia-reperfusion injury (IRI), characterized by exogenous antigen-independent local inflammation and hepatocellular death, represents a risk factor for acute and chronic rejection in liver transplantation. We aimed to investigate the molecular communication involved in the mechanism of liver IRI. METHODS We analyzed human liver transplants, primary murine macrophage cell cultures and IR-stressed livers in myeloid-specific heme oxygenase-1 (HO-1) gene mutant mice, for anti-inflammatory and cytoprotective functions of macrophage-specific HO-1/SIRT1 (sirtuin 1)/p53 (tumor suppressor protein) signaling. RESULTS Decreased HO-1 expression in human post-reperfusion liver transplant biopsies correlated with a deterioration in hepatocellular function (serum ALT; p<0.05) and inferior patient survival (p<0.05). In the low HO-1 liver transplant biopsy group, SIRT1/Arf (alternative reading frame)/p53/MDM2 (murine double minute 2) expression levels decreased (p<0.05) while cleaved caspase 3 and frequency of TUNEL+cells simultaneously increased (p<0.05). Immunofluorescence showed macrophages were the principal source of HO-1 in human and mouse IR-stressed livers. In vitro macrophage cultures revealed that HO-1 induction positively regulated SIRT1 signaling, whereas SIRT1-induced Arf inhibited ubiquitinating activity of MDM2 against p53, which in turn attenuated macrophage activation. In a murine model of hepatic warm IRI, myeloid-specific HO-1 deletion lacked SIRT1/p53, exacerbated liver inflammation and IR-hepatocellular death, whereas adjunctive SIRT1 activation restored p53 signaling and rescued livers from IR-damage. CONCLUSION This bench-to-bedside study identifies a new class of macrophages activated via the HO-1-SIRT1-p53 signaling axis in the mechanism of hepatic sterile inflammation. This mechanism could be a target for novel therapeutic strategies in liver transplant recipients. LAY SUMMARY Post-transplant low macrophage HO-1 expression in human liver transplants correlates with reduced hepatocellular function and survival. HO-1 regulates macrophage activation via the SIRT1-p53 signaling network and regulates hepatocellular death in liver ischemia-reperfusion injury. Thus targeting this pathway in liver transplant recipients could be of therapeutic benefit.
Collapse
Affiliation(s)
- Kojiro Nakamura
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Min Zhang
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Shoichi Kageyama
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Bibo Ke
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Takehiro Fujii
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Nakul Datta
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ali Zarrinpar
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Ronald W. Busuttil
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Jesus A. Araujo
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA,Corresponding authors. Addresses: Dumont-UCLA Transplant Center, 10833 Le Conte Ave, 77-120 CHS, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 4196; fax: +1 (310) 267 2358 (J.W. Kupiec-Weglinski) and UCLA Division of Cardiology, 10833 Le Conte Ave, CHS 43-264, Los Angeles, CA 90095, USA. Tel.: +1 (310) 825 3222; fax: +1 (310) 206 9133 (J.A. Araujo). (J.A. Araujo), (J.W. Kupiec-Weglinski)
| |
Collapse
|
32
|
Yu A, Dang W. Regulation of stem cell aging by SIRT1 - Linking metabolic signaling to epigenetic modifications. Mol Cell Endocrinol 2017; 455:75-82. [PMID: 28392411 PMCID: PMC7951659 DOI: 10.1016/j.mce.2017.03.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 01/09/2017] [Accepted: 03/29/2017] [Indexed: 12/22/2022]
Abstract
In mammals, profound changes in the population and functions of adult stem cells occur with age and these changes are thought to underlie functional decline and pathophysiology at the tissue and organismal levels associated with aging. SIRT1, a member of the conserved sirtuin family, functions as an anti-aging regulator for adult stem cells. Mediated through its regulatory roles in AMPK and mTORC1 pathways as well as gene expression, SIRT1 modulate the activities of genes maintaining stem cell functions and delays cellular senescence. Further investigation of the cross-talk between SIRT1 and other longevity target genes under different physiological conditions of stem cells may help us better design intervention strategies to antagonize stem cells aging.
Collapse
Affiliation(s)
- An Yu
- Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiwei Dang
- Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Chai R, Fu H, Zheng Z, Liu T, Ji S, Li G. Resveratrol inhibits proliferation and migration through SIRT1 mediated post‑translational modification of PI3K/AKT signaling in hepatocellular carcinoma cells. Mol Med Rep 2017; 16:8037-8044. [PMID: 28983625 PMCID: PMC5779887 DOI: 10.3892/mmr.2017.7612] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/12/2017] [Indexed: 01/29/2023] Open
Abstract
Resveratrol (RES), a polyphenolic compound present in grapes and red wine, has potential anticancer properties. The present study aimed to examine the effects of resveratrol and its underlying mechanism on hepatocellular carcinoma (HCC) cell lines HepG2, Bel‑7402 and SMMC‑7721. It was demonstrated that resveratrol inhibited the viability and proliferation of HCC cells assessed by MTT and EdU assays. TUNEL assay revealed that resveratrol induced cell apoptosis by increasing HCC apoptosis rate from 3±0.78% to 16±1.12% with upregulation of B‑cell lymphoma (Bcl)‑2 associated X, apoptosis regulator and cleaved‑poly (ADP‑Ribose) polymerase 1 (PARP), and downregulation of Bcl‑2, caspase‑3, caspase‑7 and PARP. As a sirtuin (SIRT) 1 activator, resveratrol elevated SIRT1 protein expression and its enzyme activity and decreased expression levels of phosphorylated (p)‑phosphoinositide‑3‑kinase (PI3K), p‑AKT Serine/Threonine Kinase 1 (AKT), and its downstream target p‑Forkhead Box O3a in HepG2 cells. Furthermore, inhibition of SIRT1 enzymatic activity by EX527 resulted in increased phosphorylation levels of PI3K and AKT. This demonstrated that resveratrol inhibited the PI3K/AKT pathway by SIRT1 activation. In addition to inhibition of cancer cell migration, tumor suppressor gene DLC1 Rho GTPase activating protein level was upregulated and its phosphorylation was enhanced by AKT with resveratrol treatment. These findings suggested that resveratrol inhibits proliferation and migration through SIRT1 mediated post‑translational modification of PI3K/AKT pathway in HCC cells.
Collapse
Affiliation(s)
- Rongfei Chai
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| | - Huiling Fu
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| | - Zhaodi Zheng
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| | - Tingting Liu
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| | - Shuhua Ji
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
34
|
Deota S, Shukla N, Kolthur-Seetharam U. Spatio-Temporal Control of Cellular and Organismal Physiology by Sirtuins. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Correia M, Perestrelo T, Rodrigues AS, Ribeiro MF, Pereira SL, Sousa MI, Ramalho-Santos J. Sirtuins in metabolism, stemness and differentiation. Biochim Biophys Acta Gen Subj 2017; 1861:3444-3455. [DOI: 10.1016/j.bbagen.2016.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/16/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
|
36
|
Weng Q, Liu Z, Li B, Liu K, Wu W, Liu H. Oxidative Stress Induces Mouse Follicular Granulosa Cells Apoptosis via JNK/FoxO1 Pathway. PLoS One 2016; 11:e0167869. [PMID: 27936150 PMCID: PMC5148000 DOI: 10.1371/journal.pone.0167869] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/21/2016] [Indexed: 01/03/2023] Open
Abstract
The c-Jun N-terminal protein kinase (JNK) plays an important role in the regulation of cell apoptosis. Forkhead box O (FoxO) transcription factors are involved in diverse biological processes, including cellular metabolism, cell apoptosis, and cell cycle. However, the JNK/FoxO1 pathway involved in the process of apoptosis induced by oxidative stress remains to be elucidated. Here, we demonstrated that the JNK activity significantly increased in response to oxidative stress in mouse follicular granulosa cells (MGCs). SP600125, a selective JNK inhibitor, attenuated the oxidative stress-induced MGCs apoptosis. Oxidative stress enhanced the FoxO1 nuclear translocation by activating the JNK activity. Moreover, JNK mediated the dissociation of FoxO1 from 14-3-3 proteins in MGCs after the treatment with H2O2. Finally, oxidative stress up-regulated the expression of FoxO1 via JNK mediation of FoxO1 self-regulation in MGCs. Taken together, our findings suggest that JNK/FoxO1 is involved in the regulation of oxidative stress-induced cell apoptosis in MGCs.
Collapse
Affiliation(s)
- Qiannan Weng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zequn Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Liaoning Province of Animal Product Safety Supervision, Shenyang, China
| | - Bojiang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kaiqing Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- * E-mail:
| |
Collapse
|
37
|
Bayarsaihan D. Deciphering the Epigenetic Code in Embryonic and Dental Pulp Stem Cells. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:539-563. [PMID: 28018144 PMCID: PMC5168831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A close cooperation between chromatin states, transcriptional modulation, and epigenetic modifications is required for establishing appropriate regulatory circuits underlying self-renewal and differentiation of adult and embryonic stem cells. A growing body of research has established that the epigenome topology provides a structural framework for engaging genes in the non-random chromosomal interactions to orchestrate complex processes such as cell-matrix interactions, cell adhesion and cell migration during lineage commitment. Over the past few years, the functional dissection of the epigenetic landscape has become increasingly important for understanding gene expression dynamics in stem cells naturally found in most tissues. Adult stem cells of the human dental pulp hold great promise for tissue engineering, particularly in the skeletal and tooth regenerative medicine. It is therefore likely that progress towards pulp regeneration will have a substantial impact on the clinical research. This review summarizes the current state of knowledge regarding epigenetic cues that have evolved to regulate the pluripotent differentiation potential of embryonic stem cells and the lineage determination of developing dental pulp progenitors.
Collapse
Affiliation(s)
- Dashzeveg Bayarsaihan
- Institute for System Genomics and Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
38
|
Yang Z, Xie C, Xu W, Liu G, Cao X, Li W, Chen J, Zhu Y, Luo S, Luo Z, Lu N. Phosphorylation and inactivation of PTEN at residues Ser380/Thr382/383 induced by Helicobacter pylori promotes gastric epithelial cell survival through PI3K/Akt pathway. Oncotarget 2016; 6:31916-26. [PMID: 26376616 PMCID: PMC4741650 DOI: 10.18632/oncotarget.5577] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023] Open
Abstract
Phosphorylation of PTEN at residues Ser380/Thr382/383 leads to loss of phosphatase activity and tumor suppressor function. Here, we found that phosphorylation of PTEN at residues Ser380/Thr382/383 was increased with gastric carcinogenesis, and more importantly, Helicobacter pylori was a trigger of this modification in chronic non-atrophic gastritis. H. pylori could phosphorylate and inactivate PTEN in vivo and in vitro, resulting in survival of gastric epithelial cells. Furthermore, stable expression of dominant-negative mutant PTEN or inhibition of Akt prevented the enhanced survival induced by H. pylori. These results indicate that PTEN phosphorylation at residues Ser380/Thr382/383 is a novel mechanism of PTEN inactivation in gastric carcinogenesis, and H. pylori triggers this modification, resulting in activation of the PI3K/Akt pathway and promotion of cell survival.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wenting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Gongmeizi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ximei Cao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhijun Luo
- The Medical College of Nanchang University, Nanchang, Jiangxi, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
39
|
Galangin Induces Autophagy via Deacetylation of LC3 by SIRT1 in HepG2 Cells. Sci Rep 2016; 6:30496. [PMID: 27460655 PMCID: PMC4962058 DOI: 10.1038/srep30496] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/06/2016] [Indexed: 12/31/2022] Open
Abstract
Galangin suppresses proliferation and induces apoptosis and autophagy in hepatocellular carcinoma (HCC) cells, but the precise mechanism is not clear. In this study, we demonstrated that galangin induced autophagy, enhanced the binding of SIRT1-LC3 and reduced the acetylation of endogenous LC3 in HepG2 cells. But this autophagy was inhibited by inactivation of SIRT1 meanwhile, galangin failed to reduce the acetylation of endogenous LC3 after SIRT1 was knocked-down. Collectively, these findings demonstrate a new mechanism by which galangin induces autophagy via the deacetylation of endogenous LC3 by SIRT1.
Collapse
|
40
|
Li BS, Guo WJ, Hong L, Liu YD, Liu C, Hong SS, Wu DB, Min J. Role of mechanical strain-activated PI3K/Akt signaling pathway in pelvic organ prolapse. Mol Med Rep 2016; 14:243-53. [PMID: 27176043 PMCID: PMC4918563 DOI: 10.3892/mmr.2016.5264] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Mechanical loading on pelvic supports contributes to pelvic organ prolapse (POP). However, the underlying mechanisms remain to be elucidated. Our previous study identified that mechanical strain induced oxidative stress (OS) and promoted apoptosis and senescence in pelvic support fibroblasts. The aim of the present study is to investigate the molecular signaling pathway linking mechanical force with POP. Using a four-point bending device, human uterosacral ligament fibroblasts (hUSLF) were exposed to mechanical tensile strain at a frequency of 0.3 Hz and intensity of 5333 µε, in the presence or absence of LY294002. The applied mechanical strain on hUSLF resulted in apoptosis and senescence, and decreased expression of procollagen type I α1. Mechanical strain activated phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt signaling and resulted in downregulated expression of glutathione peroxidase 1 and Mn-superoxide dismutase, and accumulation of intracellular reactive oxygen species. These effects were blocked by administration of LY294002. Furthermore, it was demonstrated that PI3K/Akt was activated in the uterosacral ligaments of POP patients, and that OS was increased and collagen type I production reduced. The results from the present study suggest that mechanical strain promotes apoptosis and senescence, and reduces collagen type I production via activation of PI3K/Akt-mediated OS signaling pathway in hUSLF. This process may be involved in the pathogenesis of POP as it results in relaxation and dysfunction of pelvic supports.
Collapse
Affiliation(s)
- Bing-Shu Li
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wen-Jun Guo
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Hong
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yao-Dan Liu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cheng Liu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sha-Sha Hong
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - De-Bin Wu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Min
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
41
|
A gonogenic stimulated transition of mouse embryonic stem cells with enhanced control of diverse differentiation pathways. Sci Rep 2016; 6:25104. [PMID: 27157261 PMCID: PMC4860597 DOI: 10.1038/srep25104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/08/2016] [Indexed: 11/17/2022] Open
Abstract
Embryonic stem (ES) cells share markers with undifferentiated primordial germ cells (PGCs). Here, we discovered that a cellular state with some molecular markers of male gonocyte induction, including a G1/S phase arrest and upregulation of specific genes such as Nanos2, Tdrd1, Ddx4, Zbtb16 and Plk1s1, can be chemically induced in male mouse ES cells in vitro, which we termed gonogenic stimulated transition (GoST). After longer culture of the resulting GoST cells without chemical stimulation, several molecular markers typical for early gonocytes were detected including the early gonocyte marker Tex101. Motivated by previous studies that found multipotency in cell lines derived from neonatal male germ cells in vitro, we then compared the differentiation potential of GoST cells to that of ES cells in vitro. Interestingly, GoST cells showed equal neurogenic, but enhanced cardiogenic and hepatogenic differentiation compared to ES cells in vitro. This work shows for the first time that some important molecular markers of the first developmental sexual differentiation program can be induced in male mouse ES cells in vitro and defines a novel concept to generate cells with enhanced multipotency.
Collapse
|
42
|
Yamamoto T, Tamaki K, Shirakawa K, Ito K, Yan X, Katsumata Y, Anzai A, Matsuhashi T, Endo J, Inaba T, Tsubota K, Sano M, Fukuda K, Shinmura K. Cardiac Sirt1 mediates the cardioprotective effect of caloric restriction by suppressing local complement system activation after ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2016; 310:H1003-14. [PMID: 26873964 DOI: 10.1152/ajpheart.00676.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/04/2016] [Indexed: 11/22/2022]
Abstract
Caloric restriction (CR) confers cardioprotection against ischemia-reperfusion (I/R) injury. We previously found the essential roles of endothelial nitric oxide synthase in the development of CR-induced cardioprotection and Sirt1 activation during CR (Shinmura K, Tamaki K, Ito K, Yan X, Yamamoto T, Katsumata Y, Matsuhashi T, Sano M, Fukuda K, Suematsu M, Ishii I. Indispensable role of endothelial nitric oxide synthase in caloric restriction-induced cardioprotection against ischemia-reperfusion injury.Am J Physiol Heart Circ Physiol 308: H894-H903, 2015). However, the exact mechanism by which Sirt1 in cardiomyocytes mediates the cardioprotective effect of CR remains undetermined. We subjected cardiomyocyte-specific Sirt1 knockout (CM-Sirt1(-/-)) mice and the corresponding control mice to either 3-mo ad libitum feeding or CR (-40%). Isolated perfused hearts were subjected to 25-min global ischemia, followed by 60-min reperfusion. The recovery of left ventricle function after I/R was improved, and total lactate dehydrogenase release into the perfusate during reperfusion was attenuated in the control mice treated with CR, but a similar cardioprotective effect of CR was not observed in the CM-Sirt1(-/-)mice. The expression levels of cardiac complement component 3 (C3) at baseline and the accumulation of C3 and its fragments in the ischemia-reperfused myocardium were attenuated by CR in the control mice, but not in the CM-Sirt1(-/-)mice. Resveratrol treatment also attenuated the expression levels of C3 protein in cultured neonatal rat ventricular cardiomyocytes. Moreover, the degree of myocardial I/R injury in conventional C3 knockout (C3(-/-)) mice treated with CR was similar to that in the ad libitum-fed C3(-/-)mice, although the expression levels of Sirt1 were enhanced by CR. These results demonstrate that cardiac Sirt1 plays an essential role in CR-induced cardioprotection against I/R injury by suppressing cardiac C3 expression. This is the first report suggesting that cardiac Sirt1 regulates the local complement system during CR.
Collapse
Affiliation(s)
- Tsunehisa Yamamoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kayoko Tamaki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of General Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kohsuke Shirakawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kentaro Ito
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaoxiang Yan
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Atsushi Anzai
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Takaaki Inaba
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan; and
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan; and
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Ken Shinmura
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of General Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
43
|
The expression of NLRX1 in C57BL/6 mice cochlear hair cells: Possible relation to aging- and neomycin-induced deafness. Neurosci Lett 2016; 616:138-46. [PMID: 26836140 DOI: 10.1016/j.neulet.2015.11.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 11/22/2015] [Accepted: 11/23/2015] [Indexed: 12/21/2022]
Abstract
Nucleotide-binding domain and leucine-rich-repeat-containing family member X1 (NLRX1) is a cytoplasmic pattern recognition receptor that is predominantly located in mitochondria, which is tightly related to mitochondrial damage, reactive oxygen species (ROS) production, inflammation and apoptosis. The present study was designed to explore whether NLRX1 expresses in C57BL/6 mice cochlear hair cells and, if so, to investigate the possible correlations between NLRX1 and hearing. The location and dynamic expression of NLRX1 were investigated by immunofluorescence, real-time PCR and Western blotting. Hearing thresholds of C57BL/6 mice were measured by auditory brainstem response (ABR). Moreover, the downstream inflammatory and apoptotic pathways regulated by NLRX1 were examined in age-related and neomycin-induced hair cell damage. Data showed that NLRX1 expressed in cytoplasm of C57BL/6 cochlear hair cells, especially in the cilia, which were essential for sound sensation. The expression of NLRX1 in hair cells increased as the mice grew up, and, decreased as they aged. Additionally, the activated apoptotic JNK pathway was detected in 9-month old mice with worse-hearing and 3-month old mice treated with neomycin. Overall, results indicate that NLRX1 may relate to hair cell maturity, hearing formation and maintenance, and promote hair cell apoptosis through JNK pathway induced by aging and neomycin.
Collapse
|
44
|
Wang WR, Liu EQ, Zhang JY, Li YX, Yang XF, He YH, Zhang W, Jing T, Lin R. Activation of PPAR alpha by fenofibrate inhibits apoptosis in vascular adventitial fibroblasts partly through SIRT1-mediated deacetylation of FoxO1. Exp Cell Res 2015; 338:54-63. [DOI: 10.1016/j.yexcr.2015.07.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/09/2015] [Accepted: 07/25/2015] [Indexed: 11/30/2022]
|
45
|
Bermúdez Brito M, Goulielmaki E, Papakonstanti EA. Focus on PTEN Regulation. Front Oncol 2015; 5:166. [PMID: 26284192 PMCID: PMC4515857 DOI: 10.3389/fonc.2015.00166] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/07/2015] [Indexed: 12/17/2022] Open
Abstract
The role of phosphatase and tensin homolog on chromosome 10 (PTEN) as a tumor suppressor has been for a long time attributed to its lipid phosphatase activity against PI(3,4,5)P3, the phospholipid product of the class I PI3Ks. Besides its traditional role as a lipid phosphatase at the plasma membrane, a wealth of data has shown that PTEN can function independently of its phosphatase activity and that PTEN also exists and plays a role in the nucleus, in cytoplasmic organelles, and extracellularly. Accumulating evidence has shed light on diverse physiological functions of PTEN, which are accompanied by a complex regulation of its expression and activity. PTEN levels and function are regulated transcriptionally, post-transcriptionally, and post-translationally. PTEN is also sensitive to regulation by its interacting proteins and its localization. Herein, we summarize the current knowledge on mechanisms that regulate the expression and enzymatic activity of PTEN and its role in human diseases.
Collapse
Affiliation(s)
- Miriam Bermúdez Brito
- Department of Biochemistry, School of Medicine, University of Crete , Heraklion , Greece
| | - Evangelia Goulielmaki
- Department of Biochemistry, School of Medicine, University of Crete , Heraklion , Greece
| | | |
Collapse
|
46
|
Time- and dose-dependent effects of ethanol on mouse embryonic stem cells. Reprod Toxicol 2015; 57:157-64. [PMID: 26073001 DOI: 10.1016/j.reprotox.2015.06.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 05/21/2015] [Accepted: 06/03/2015] [Indexed: 01/08/2023]
Abstract
Ethanol is a common solvent used with mouse embryonic stem (mES) cells in protocols to test chemicals for evidence of developmental toxicity. In this study, dose-response relationships for ethanol toxicity in mES cells were examined. For cells maintained in an undifferentiated state, ethanol significantly reduced viable cell numbers with estimated half maximal inhibitory concentrations of 1.5% and 0.8% ethanol after 24 and 48h, respectively, observations which correlated with significantly increased expression of apoptotic markers. For cells cultured to induce cardiomyocyte formation, up to 0.5% ethanol during the first two days failed to alter the outcome of differentiation, whereas 0.3% ethanol for 11 days significantly reduced the fraction of cultures containing contracting areas, an observation that correlated with significantly reduced cell numbers. These results suggest that ethanol is not an inert solvent at concentrations that might be used for developmental toxicity testing.
Collapse
|
47
|
Maiese K. FoxO proteins in the nervous system. Anal Cell Pathol (Amst) 2015; 2015:569392. [PMID: 26171319 PMCID: PMC4478359 DOI: 10.1155/2015/569392] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/31/2015] [Indexed: 02/07/2023] Open
Abstract
Acute as well as chronic disorders of the nervous system lead to significant morbidity and mortality for millions of individuals globally. Given the ability to govern stem cell proliferation and differentiated cell survival, mammalian forkhead transcription factors of the forkhead box class O (FoxO) are increasingly being identified as potential targets for disorders of the nervous system, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and auditory neuronal disease. FoxO proteins are present throughout the body, but they are selectively expressed in the nervous system and have diverse biological functions. The forkhead O class transcription factors interface with an array of signal transduction pathways that include protein kinase B (Akt), serum- and glucocorticoid-inducible protein kinase (SgK), IκB kinase (IKK), silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), growth factors, and Wnt signaling that can determine the activity and integrity of FoxO proteins. Ultimately, there exists a complex interplay between FoxO proteins and their signal transduction pathways that can significantly impact programmed cell death pathways of apoptosis and autophagy as well as the development of clinical strategies for the treatment of neurodegenerative disorders.
Collapse
|
48
|
LaBarge S, Migdal C, Schenk S. Is acetylation a metabolic rheostat that regulates skeletal muscle insulin action? Mol Cells 2015; 38:297-303. [PMID: 25824547 PMCID: PMC4400303 DOI: 10.14348/molcells.2015.0020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle insulin resistance, which increases the risk for developing various metabolic diseases, including type 2 diabetes, is a common metabolic disorder in obesity and aging. If potential treatments are to be developed to treat insulin resistance, then it is important to fully understand insulin signaling and glucose metabolism. While recent large-scale "omics" studies have revealed the acetylome to be comparable in size to the phosphorylome, the acetylation of insulin signaling proteins and its functional relevance to insulin-stimulated glucose transport and glucose metabolism is not fully understood. In this Mini Review we discuss the acetylation status of proteins involved in the insulin signaling pathway and review their potential effect on, and relevance to, insulin action in skeletal muscle.
Collapse
Affiliation(s)
- Samuel LaBarge
- Department of Orthopaedic Surgery, University of California, San Diego, CA, 92093,
USA
| | - Christopher Migdal
- Department of Orthopaedic Surgery, University of California, San Diego, CA, 92093,
USA
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, CA, 92093,
USA
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, 92093,
USA
| |
Collapse
|
49
|
Abstract
The controversy around sirtuins and their functions in aging has drawn in the past few years as much attention, if not more, from the scientific community and the public as they did when first proposed as the key conserved aging regulators in eukaryotes. With some of the basic observations on sirtuin longevity promoting functions being questioned in popular model systems, researchers are wondering if this family of conserved enzymes still holds strong potential as therapeutic targets. This review examines the several controversial issues around sirtuins and their functions in aging, calorie restriction, as well as age-related diseases in light of recent studies in mammalian systems and discusses whether modulators of sirtuins still hold the secret of life.:
Collapse
Affiliation(s)
- Weiwei Dang
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Maiese K. SIRT1 and stem cells: In the forefront with cardiovascular disease, neurodegeneration and cancer. World J Stem Cells 2015; 7:235-242. [PMID: 25815111 PMCID: PMC4369483 DOI: 10.4252/wjsc.v7.i2.235] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 12/10/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, nervous system disorders, and cancer in association with other diseases such as diabetes mellitus result in greater than sixty percent of the global annual deaths. These noncommunicable diseases also affect at least one-third of the population in low and middle-income countries and lead to hypertension, elevated cholesterol, malignancy, and neurodegenerative disorders such as Alzheimer’s disease and stroke. With the climbing lifespan of the world’s population, increased prevalence of these disorders is expected requiring the development of new therapeutic strategies against these disabling disease entities. Targeting stem cell proliferation for cardiac disease, vascular disorders, cancer, and neurodegenerative disorders is receiving great enthusiasm, especially those that focus upon SIRT1, a mammalian homologue of the yeast silent information regulator-2. Modulation of the cellular activity of SIRT1 can involve oversight by nicotinamide/nicotinic acid mononucleotide adenylyltransferase, mammalian forkhead transcription factors, mechanistic of rapamycin pathways, and cysteine-rich protein 61, connective tissue growth factor, and nephroblastoma over-expressed gene family members that can impact cytoprotective outcomes. Ultimately, the ability of SIRT1 to control the programmed cell death pathways of apoptosis and autophagy can determine not only cardiac, vascular, and neuronal stem cell development and longevity, but also the onset of tumorigenesis and the resistance against chemotherapy. SIRT1 therefore has a critical role and holds exciting prospects for new therapeutic strategies that can offer reparative processes for cardiac, vascular, and nervous system degenerative disorders as well as targeted control of aberrant cell growth during cancer.
Collapse
|