101
|
Abstract
Sestrins are a family of proteins that respond to a variety of environmental stresses, including genotoxic, oxidative, and nutritional stresses. Sestrins affect multiple signaling pathways: AMP-activated protein kinase, mammalian target of rapamycin complexes, insulin-AKT, and redox signaling pathways. By regulating these pathways, Sestrins are thought to help adapt to stressful environments and subsequently restore cell and tissue homeostasis. In this review, we describe how Sestrins mediate physiological stress responses in the context of nutritional and chemical stresses (liver), physical movement and exercise (skeletal muscle), and chemical, physical, and inflammatory injuries (heart). These findings also support the idea that Sestrins are a molecular mediator of hormesis, a paradoxical beneficial effect of low- or moderate-level stresses in living organisms.
Collapse
Affiliation(s)
- Myungjin Kim
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA; ,
| | - Allison H Kowalsky
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA; ,
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA; ,
| |
Collapse
|
102
|
Haidurov A, Budanov AV. Sestrin family - the stem controlling healthy ageing. Mech Ageing Dev 2020; 192:111379. [PMID: 33022334 DOI: 10.1016/j.mad.2020.111379] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 01/18/2023]
Abstract
Sestrins are a family of stress-responsive antioxidant proteins responsible for regulation of cell viability and metabolism. The best known Sestrin targets are mTORC1 and mTORC2 kinases that control different cellular processes including growth, viability, autophagy, and mitochondrial metabolism. Inactivation of the single Sestrin gene in invertebrates has an adverse impact on their healthspan and longevity, whereas each of the three Sestrin genes in mammals and other vertebrate organisms has a different impact on maintenance of a particular tissue, affecting its stress tolerance, function and regenerative capability. As a result, Sestrins attenuate ageing and suppress development of many age-related diseases including myocardial infarction, muscle atrophy, diabetes, and immune dysfunction, but exacerbate development of chronic obstructive pulmonary disease. Moreover, Sestrins play opposite roles in carcinogenesis in different tissues. Stem cells support tissue remodelling that influences ageing, and Sestrins might suppress ageing and age-related pathologies through control of stem cell biology. In this review, we will discuss the potential link between Sestrins, stem cells, and ageing.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrei V Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland; Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
103
|
Texada MJ, Koyama T, Rewitz K. Regulation of Body Size and Growth Control. Genetics 2020; 216:269-313. [PMID: 33023929 PMCID: PMC7536854 DOI: 10.1534/genetics.120.303095] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The control of body and organ growth is essential for the development of adults with proper size and proportions, which is important for survival and reproduction. In animals, adult body size is determined by the rate and duration of juvenile growth, which are influenced by the environment. In nutrient-scarce environments in which more time is needed for growth, the juvenile growth period can be extended by delaying maturation, whereas juvenile development is rapidly completed in nutrient-rich conditions. This flexibility requires the integration of environmental cues with developmental signals that govern internal checkpoints to ensure that maturation does not begin until sufficient tissue growth has occurred to reach a proper adult size. The Target of Rapamycin (TOR) pathway is the primary cell-autonomous nutrient sensor, while circulating hormones such as steroids and insulin-like growth factors are the main systemic regulators of growth and maturation in animals. We discuss recent findings in Drosophila melanogaster showing that cell-autonomous environment and growth-sensing mechanisms, involving TOR and other growth-regulatory pathways, that converge on insulin and steroid relay centers are responsible for adjusting systemic growth, and development, in response to external and internal conditions. In addition to this, proper organ growth is also monitored and coordinated with whole-body growth and the timing of maturation through modulation of steroid signaling. This coordination involves interorgan communication mediated by Drosophila insulin-like peptide 8 in response to tissue growth status. Together, these multiple nutritional and developmental cues feed into neuroendocrine hubs controlling insulin and steroid signaling, serving as checkpoints at which developmental progression toward maturation can be delayed. This review focuses on these mechanisms by which external and internal conditions can modulate developmental growth and ensure proper adult body size, and highlights the conserved architecture of this system, which has made Drosophila a prime model for understanding the coordination of growth and maturation in animals.
Collapse
Affiliation(s)
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Denmark
| |
Collapse
|
104
|
Zhu SY, Yao RQ, Li YX, Zhao PY, Ren C, Du XH, Yao YM. Lysosomal quality control of cell fate: a novel therapeutic target for human diseases. Cell Death Dis 2020; 11:817. [PMID: 32999282 PMCID: PMC7528093 DOI: 10.1038/s41419-020-03032-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023]
Abstract
In eukaryotic cells, lysosomes are digestive centers where biological macromolecules are degraded by phagocytosis and autophagy, thereby maintaining cellular self-renewal capacity and energy supply. Lysosomes also serve as signaling hubs to monitor the intracellular levels of nutrients and energy by acting as platforms for the assembly of multiple signaling pathways, such as mammalian target of rapamycin complex 1 (mTORC1) and adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK). The structural integrity and functional balance of lysosomes are essential for cell function and viability. In fact, lysosomal damage not only disrupts intracellular clearance but also results in the leakage of multiple contents, which pose great threats to the cell by triggering cell death pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis. The collapse of lysosomal homeostasis is reportedly critical for the pathogenesis and development of various diseases, such as tumors, neurodegenerative diseases, cardiovascular diseases, and inflammatory diseases. Lysosomal quality control (LQC), comprising lysosomal repair, lysophagy, and lysosomal regeneration, is rapidly initiated in response to lysosomal damage to maintain lysosomal structural integrity and functional homeostasis. LQC may be a novel but pivotal target for disease treatment because of its indispensable role in maintaining intracellular homeostasis and cell fate.
Collapse
Affiliation(s)
- Sheng-Yu Zhu
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.,Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China.,School of Medicine, Nankai University, 300071, Tianjin, People's Republic of China
| | - Ren-Qi Yao
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.,Department of Burn Surgery, Changhai Hospital, Naval Medical University, 200433, Shanghai, People's Republic of China
| | - Yu-Xuan Li
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China
| | - Peng-Yue Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China
| | - Chao Ren
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.
| | - Xiao-Hui Du
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China.
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.
| |
Collapse
|
105
|
Shin J, Bae J, Park S, Kang HG, Shin SM, Won G, Kim JS, Cho SG, Choi Y, Oh SM, Shin J, Kim JS, Park HW. mTOR-Dependent Role of Sestrin2 in Regulating Tumor Progression of Human Endometrial Cancer. Cancers (Basel) 2020; 12:cancers12092515. [PMID: 32899752 PMCID: PMC7565818 DOI: 10.3390/cancers12092515] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Mammalian target of rapamycin complex 1 (mTORC1), a key controller of growth and environmental stress signaling, is frequently activated in human cancers. Sestrin2 (SESN2), a highly conserved stress-inducible protein, is one of the negative feedback mechanisms for inhibiting chronic activation of mTORC1. This study aimed to investigate the expression and clinical implications of SESN2 in endometrial cancer using an in vitro and in vivo approach. The analysis indicated increased levels of SESN2 and mTORC1 pathway activity in cancer tissues than in normal tissues. High SESN2 expression correlated with shorter patient survival duration. However, lentiviral overexpression of SESN2 and mTOR inhibitors suppressed cancer cell proliferation, migration, and epithelial–mesenchymal transition. Our study provides strong evidence for prognostic significance of SESN2, and its association with mTORC1 pathway and endometrial cancer growth. Thus, the results identified SESN2 as a potential therapeutic target in endometrial cancer. Abstract Oncogenic activation of the mammalian target of rapamycin complex 1 (mTORC1) leads to endometrial cancer cell growth and proliferation. Sestrin2 (SESN2), a highly conserved stress-inducible protein, is involved in homeostatic regulation via inhibition of reactive oxygen species (ROS) and mTORC1. However, the role of SESN2 in human endometrial cancer remains to be investigated. Here, we investigated expression, clinical significance, and underlying mechanisms of SESN2 in endometrial cancer. SESN2 was upregulated more in endometrial cancer tissues than in normal endometrial tissues. Furthermore, upregulation of SESN2 statistically correlated with shorter overall survival and disease-free survival in patients with endometrial cancer. SESN2 expression strongly correlated with mTORC1 activity, suggesting its impact on prognosis in endometrial cancer. Additionally, knockdown of SESN2 promoted cell proliferation, migration, and ROS production in endometrial cancer cell lines HEC-1A and Ishikawa. Treatment of these cells with mTOR inhibitors reversed endometrial cancer cell proliferation, migration, and epithelial–mesenchymal transition (EMT) marker expression. Moreover, in a xenograft nude mice model, endometrial cancer growth increased by SESN2 knockdown. Thus, our study provides evidence for the prognostic significance of SESN2, and a relationship between SESN2, the mTORC1 pathway, and endometrial cancer growth, suggesting SESN2 as a potential therapeutic target in endometrial cancer.
Collapse
Affiliation(s)
- Jiha Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
| | - Jeongyun Bae
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
| | - Sumi Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
| | - Hyun-Goo Kang
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
| | - Seong Min Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
| | - Gunho Won
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
- Department Centers for Disease Control & Prevention, National Institute of Health, Cheongju 28159, Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (S.-G.C.); (Y.C.)
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (S.-G.C.); (Y.C.)
| | - Sang-Muk Oh
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
- Department of Biochemistry, Konyang University College of Medicine, Daejeon 35365, Korea
| | - Jongdae Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
| | - Jeong Sig Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea
- Correspondence: (J.S.K.); (H.-W.P.); Tel.: +82-42-600-8677 (H.-W.P.)
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea; (J.S.); (J.B.); (S.P.); (H.-G.K.); (S.M.S.); (J.S.)
- Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Korea; (G.W.); (J.-S.K.); (S.-M.O.)
- Correspondence: (J.S.K.); (H.-W.P.); Tel.: +82-42-600-8677 (H.-W.P.)
| |
Collapse
|
106
|
Lee W, Ahn JH, Park HH, Kim HN, Kim H, Yoo Y, Shin H, Hong KS, Jang JG, Park CG, Choi EY, Bae JS, Seo YK. COVID-19-activated SREBP2 disturbs cholesterol biosynthesis and leads to cytokine storm. Signal Transduct Target Ther 2020; 5:186. [PMID: 32883951 PMCID: PMC7471497 DOI: 10.1038/s41392-020-00292-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/15/2023] Open
Abstract
Sterol regulatory element binding protein-2 (SREBP-2) is activated by cytokines or pathogen, such as virus or bacteria, but its association with diminished cholesterol levels in COVID-19 patients is unknown. Here, we evaluated SREBP-2 activation in peripheral blood mononuclear cells of COVID-19 patients and verified the function of SREBP-2 in COVID-19. Intriguingly, we report the first observation of SREBP-2 C-terminal fragment in COVID-19 patients' blood and propose SREBP-2 C-terminal fragment as an indicator for determining severity. We confirmed that SREBP-2-induced cholesterol biosynthesis was suppressed by Sestrin-1 and PCSK9 expression, while the SREBP-2-induced inflammatory responses was upregulated in COVID-19 ICU patients. Using an infectious disease mouse model, inhibitors of SREBP-2 and NF-κB suppressed cytokine storms caused by viral infection and prevented pulmonary damages. These results collectively suggest that SREBP-2 can serve as an indicator for severity diagnosis and therapeutic target for preventing cytokine storm and lung damage in severe COVID-19 patients.
Collapse
Affiliation(s)
- Wonhwa Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - June Hong Ahn
- Division of Pulmonology and Allergy, Department of Internal Medicine, College of Medicine, Yeungnam University and Regional Center for Respiratory Diseases, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea
| | - Hee Ho Park
- Department of Biotechnology and Bioengineering, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Hong Nam Kim
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyelim Kim
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Youngbum Yoo
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Hyosoo Shin
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kyung Soo Hong
- Division of Pulmonology and Allergy, Department of Internal Medicine, College of Medicine, Yeungnam University and Regional Center for Respiratory Diseases, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea
| | - Jong Geol Jang
- Division of Pulmonology and Allergy, Department of Internal Medicine, College of Medicine, Yeungnam University and Regional Center for Respiratory Diseases, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
| | - Eun Young Choi
- Division of Pulmonology and Allergy, Department of Internal Medicine, College of Medicine, Yeungnam University and Regional Center for Respiratory Diseases, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea.
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Young-Kyo Seo
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
107
|
Chae HS, Gil M, Saha SK, Kwak HJ, Park HW, Vellingiri B, Cho SG. Sestrin2 Expression Has Regulatory Properties and Prognostic Value in Lung Cancer. J Pers Med 2020; 10:jpm10030109. [PMID: 32882793 PMCID: PMC7565522 DOI: 10.3390/jpm10030109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/23/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer remains the most dangerous type of cancer despite recent progress in therapeutic modalities. Development of prognostic markers and therapeutic targets is necessary to enhance lung cancer patient survival. Sestrin family genes (Sestrin1, Sestrin2, and Sestrin3) are involved in protecting cells from stress. In particular, Sestrin2, which mainly protects cells from oxidative stress and acts as a leucine sensor protein in mammalian target of rapamycin (mTOR) signaling, is thought to affect various cancers in different ways. To investigate the role of Sestrin2 expression in lung cancer cells, we knocked down Sestrin2 in A549, a non-small cell lung cancer cell line; this resulted in reduced cell proliferation, migration, sphere formation, and drug resistance, suggesting that Sestrin2 is closely related to lung cancer progression. We analyzed Sestrin2 expression in human tissue using various bioinformatic databases and confirmed higher expression of Sestrin2 in lung cancer cells than in normal lung cells using Oncomine and the Human Protein Atlas. Moreover, analyses using Prognoscan and KMplotter showed that Sestrin2 expression is negatively correlated with the survival of lung cancer patients in multiple datasets. Co-expressed gene analysis revealed Sestrin2-regulated genes and possible associated pathways. Overall, these data suggest that Sestrin2 expression has prognostic value and that it is a possible therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Hee Sung Chae
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (H.S.C.); (M.G.); (S.K.S.); (H.J.K.)
| | - Minchan Gil
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (H.S.C.); (M.G.); (S.K.S.); (H.J.K.)
| | - Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (H.S.C.); (M.G.); (S.K.S.); (H.J.K.)
| | - Hee Jeung Kwak
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (H.S.C.); (M.G.); (S.K.S.); (H.J.K.)
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea;
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India;
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (H.S.C.); (M.G.); (S.K.S.); (H.J.K.)
- Correspondence: ; Tel.: +82-2-450-4207; Fax: +82-2-444-4207
| |
Collapse
|
108
|
Liu Y, Du X, Huang Z, Zheng Y, Quan N. Sestrin 2 controls the cardiovascular aging process via an integrated network of signaling pathways. Ageing Res Rev 2020; 62:101096. [PMID: 32544433 DOI: 10.1016/j.arr.2020.101096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/03/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
As an inevitable biological process, cardiovascular aging is the greatest risk factor for cardiovascular diseases (CVDs). Sestrin 2 (Sesn2), a stress-inducible and age-related protein associated with various stress conditions, plays a pivotal role in slowing this process. It acts as an anti-aging agent, mainly through its antioxidant enzymatic activity and regulation of antioxidant signaling pathways, as well as by activating adenosine monophosphate-activated protein kinase and inhibiting mammalian target of rapamycin complex 1. In this review, we first introduce the biochemical functions of Sesn2 in the cardiovascular aging process, and describe how Sesn2 expression is regulated under various stress conditions. Next, we emphasize the role of Sesn2 signal transduction in a series of age-related CVDs, including hypertension, myocardial ischemia and reperfusion, atherosclerosis, and heart failure, as well as provide potential mechanisms for the association of Sesn2 with CVDs. Finally, we present the potential therapeutic applications of Sesn2-directed therapy and future prospects.
Collapse
Affiliation(s)
- Yunxia Liu
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xiaoyu Du
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhehao Huang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Yang Zheng
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Nanhu Quan
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
109
|
Kitada M, Xu J, Ogura Y, Monno I, Koya D. Mechanism of Activation of Mechanistic Target of Rapamycin Complex 1 by Methionine. Front Cell Dev Biol 2020; 8:715. [PMID: 32850834 PMCID: PMC7431653 DOI: 10.3389/fcell.2020.00715] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
Nutrients are closely involved in the regulation of lifespan and metabolic health. Cellular activities, such as the regulation of metabolism, growth, and aging, are mediated by a network of nutrients and nutrient-sensing pathways. Among the nutrient-sensing pathways, the mechanistic target of rapamycin complex 1 (mTORC1) acts as the central regulator of cellular functions, which include autophagy. Autophagy plays a significant role in the removal of protein aggregates and damaged or excess organelles, including mitochondria, to maintain intracellular homeostasis, which is involved in lifespan extension and cardiometabolic health. Moreover, dietary methionine restriction may have a beneficial effect on lifespan extension and metabolic health. In contrast, methionine may activate mTORC1 and suppress autophagy. As the mechanism of methionine sensing on mTORC1, SAMTOR was identified as a sensor of S-adenosyl methionine (SAM), a metabolite of methionine, in the cytoplasm. Conversely, methionine may activate the mTORC1 signaling pathway through the activation of phosphatase 2A (PP2A) because of increased methylation in response to intracellular SAM levels. In this review, we summarized the recent findings regarding the mechanism via which methionine activates mTORC1.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Jing Xu
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Itaru Monno
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
110
|
Pathological Consequences of Hepatic mTORC1 Dysregulation. Genes (Basel) 2020; 11:genes11080896. [PMID: 32764389 PMCID: PMC7465966 DOI: 10.3390/genes11080896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 12/28/2022] Open
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) is a central regulator of metabolism that integrates environmental inputs, including nutrients, growth factors, and stress signals. mTORC1 activation upregulates anabolism of diverse macromolecules, such as proteins, lipids, and nucleic acids, while downregulating autolysosomal catabolism. mTORC1 dysregulation is often found in various diseases, including cancer, cardiovascular and neurodegenerative diseases, as well as metabolic syndromes involving obesity and type II diabetes. As an essential metabolic organ, the liver requires proper regulation of mTORC1 for maintaining homeostasis and preventing pathologies. For instance, aberrant hyper- or hypoactivation of mTORC1 disrupts hepatocellular homeostasis and damages the structural and functional integrity of the tissue, leading to prominent liver injury and the development of hepatocellular carcinogenesis. Proper regulation of mTORC1 during liver diseases may be beneficial for restoring liver function and ameliorating the detrimental consequences of liver failure.
Collapse
|
111
|
Li W, Zhao L, Wang J. Searching for the Mechanisms of Mammalian Cellular Aging Through Underlying Gene Regulatory Networks. Front Genet 2020; 11:593. [PMID: 32714367 PMCID: PMC7340167 DOI: 10.3389/fgene.2020.00593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/15/2020] [Indexed: 01/16/2023] Open
Abstract
Aging attracts the attention throughout the history of humankind. However, it is still challenging to understand how the internal driving forces, for example, the fundamental building blocks of life, such as genes and proteins, as well as the environments work together to determine longevity in mammals. In this study, we built a gene regulatory network for mammalian cellular aging based on the experimental literature and quantify its underlying driving force for the dynamics as potential and flux landscape. We found three steady-state attractors: a fast-aging state attractor, slow-aging state attractor, and intermediate state attractor. The system can switch from one state attractor to another driven by the intrinsic or external forces through the genetics and the environment. We identified the dominant path from the slow-aging state directly to the fast-aging state. We also identified the dominant path from slow-aging to fast-aging through an intermediate state. We quantified the evolving landscape for revealing the dynamic characteristics of aging through certain regulation changes in time. We also predicted the key genes and regulations for fast-aging and slow-aging through the analysis of the stability for landscape basins. We also found the oscillation dynamics between fast-aging and slow-aging and showed that more energy is required to sustain such oscillations. We found that the flux is the dynamic cause and the entropy production rate the thermodynamic origin of the phase transitions or the bifurcations between the three-state phase and oscillation phase. The landscape quantification provides a global and physical approach to explore the underlying mechanisms of cellular aging in mammals.
Collapse
Affiliation(s)
- Wenbo Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Lei Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Jin Wang
- Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
112
|
Augert A, Mathsyaraja H, Ibrahim AH, Freie B, Geuenich MJ, Cheng PF, Alibeckoff SP, Wu N, Hiatt JB, Basom R, Gazdar A, Sullivan LB, Eisenman RN, MacPherson D. MAX Functions as a Tumor Suppressor and Rewires Metabolism in Small Cell Lung Cancer. Cancer Cell 2020; 38:97-114.e7. [PMID: 32470392 PMCID: PMC7363581 DOI: 10.1016/j.ccell.2020.04.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Small cell lung cancer (SCLC) is a highly aggressive and lethal neoplasm. To identify candidate tumor suppressors we applied CRISPR/Cas9 gene inactivation screens to a cellular model of early-stage SCLC. Among the top hits was MAX, the obligate heterodimerization partner for MYC family proteins that is mutated in human SCLC. Max deletion increases growth and transformation in cells and dramatically accelerates SCLC progression in an Rb1/Trp53-deleted mouse model. In contrast, deletion of Max abrogates tumorigenesis in MYCL-overexpressing SCLC. Max deletion in SCLC resulted in derepression of metabolic genes involved in serine and one-carbon metabolism. By increasing serine biosynthesis, Max-deleted cells exhibit resistance to serine depletion. Thus, Max loss results in metabolic rewiring and context-specific tumor suppression.
Collapse
Affiliation(s)
- Arnaud Augert
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Haritha Mathsyaraja
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ali H Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Brian Freie
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Michael J Geuenich
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Quest University Canada, 3200 University Boulevard, Squamish, BC V8B 0N8, Canada
| | - Pei-Feng Cheng
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sydney P Alibeckoff
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Nan Wu
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Joseph B Hiatt
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Ryan Basom
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Adi Gazdar
- University of Texas, Southwestern, USA, 6000 Harry Hines Boulevard, Dallas, TX 75235, USA
| | - Lucas B Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robert N Eisenman
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
113
|
Quan N, Li X, Zhang J, Han Y, Sun W, Ren D, Tong Q, Li J. Substrate metabolism regulated by Sestrin2-mTORC1 alleviates pressure overload-induced cardiac hypertrophy in aged heart. Redox Biol 2020; 36:101637. [PMID: 32863202 PMCID: PMC7363709 DOI: 10.1016/j.redox.2020.101637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 01/12/2023] Open
Abstract
Sestrin2 (Sesn2) is a stress sensor for the mammalian target of rapamycin complex 1 (mTORC1) pathway. Aging impairs cardiac mTORC1 activation, thereby sensitizing the heart to hypertrophy. C57BL/6 J young wild-type (young WT; 4-6 months), aged WT (24-26 months), and young Sestrin2 knockout mice (Y-Sesn2 KO; 4-6 months) underwent transverse aortic constriction (TAC) for pressure overload. Cardiac expression of Sesn2 decreased with age. At 4 weeks after TAC, aged WT and Y-Sesn2 KO exhibited larger hearts and impaired cardiac function, compared with young WT mice. Augmented phosphorylation of mTOR and downstream effectors; damaged mitochondria and elevated redox markers, as well as and impaired glucose and fatty acid oxidation were observed in aged WT and Y-Sesn2 KO hearts. A pressure overload-induced interaction between Sesn2 and GTPase-activating protein activity toward Rags 2 (GATOR2), which positively regulates mTORC1, was impaired in aged WT hearts. Adeno-associated virus 9-Sesn2 treatment rescued Sesn2 expression, attenuated mTORC1 activation, and increased pressure overload tolerance in aged WT and Y-Sesn2 KO hearts. These results indicated that cardiac Sesn2 acts as a pressure overload sensor for mTORC1. Furthermore, Sesn2 deficiency may cause increased sensitivity to hypertrophy in elderly individuals.
Collapse
Affiliation(s)
- Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China,Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA,Corresponding author. Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China. Tel.: +86 13844803504.
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jingwen Zhang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Ying Han
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Weiju Sun
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Qian Tong
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China,Corresponding author. Tel.: +86 15804300981.
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| |
Collapse
|
114
|
Ambrosio S, Majello B. Autophagy Roles in Genome Maintenance. Cancers (Basel) 2020; 12:E1793. [PMID: 32635505 PMCID: PMC7407194 DOI: 10.3390/cancers12071793] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, a considerable correlation has emerged between autophagy and genome integrity. A range of mechanisms appear to be involved where autophagy participates in preventing genomic instability, as well as in DNA damage response and cell fate decision. These initial findings have attracted particular attention in the context of malignancy; however, the crosstalk between autophagy and DNA damage response is just beginning to be explored and key questions remain that need to be addressed, to move this area of research forward and illuminate the overall consequence of targeting this process in human therapies. Here we present current knowledge on the complex crosstalk between autophagy and genome integrity and discuss its implications for cancer cell survival and response to therapy.
Collapse
Affiliation(s)
- Susanna Ambrosio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy;
| | - Barbara Majello
- Department of Biology, University of Naples ‘Federico II’, 80138 Naples, Italy
| |
Collapse
|
115
|
Qian J, Su S, Liu P. Experimental Approaches in Delineating mTOR Signaling. Genes (Basel) 2020; 11:E738. [PMID: 32630768 PMCID: PMC7397015 DOI: 10.3390/genes11070738] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 11/16/2022] Open
Abstract
The mTOR signaling controls essential biological functions including proliferation, growth, metabolism, autophagy, ageing, and others. Hyperactivation of mTOR signaling leads to a plethora of human disorders; thus, mTOR is an attractive drug target. The discovery of mTOR signaling started from isolation of rapamycin in 1975 and cloning of TOR genes in 1993. In the past 27 years, numerous research groups have contributed significantly to advancing our understanding of mTOR signaling and mTOR biology. Notably, a variety of experimental approaches have been employed in these studies to identify key mTOR pathway members that shape up the mTOR signaling we know today. Technique development drives mTOR research, while canonical biochemical and yeast genetics lay the foundation for mTOR studies. Here in this review, we summarize major experimental approaches used in the past in delineating mTOR signaling, including biochemical immunoprecipitation approaches, genetic approaches, immunofluorescence microscopic approaches, hypothesis-driven studies, protein sequence or motif search driven approaches, and bioinformatic approaches. We hope that revisiting these distinct types of experimental approaches will provide a blueprint for major techniques driving mTOR research. More importantly, we hope that thinking and reasonings behind these experimental designs will inspire future mTOR research as well as studies of other protein kinases beyond mTOR.
Collapse
Affiliation(s)
- Jiayi Qian
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.Q.); (S.S.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Siyuan Su
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.Q.); (S.S.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.Q.); (S.S.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
116
|
Shorning BY, Dass MS, Smalley MJ, Pearson HB. The PI3K-AKT-mTOR Pathway and Prostate Cancer: At the Crossroads of AR, MAPK, and WNT Signaling. Int J Mol Sci 2020; 21:E4507. [PMID: 32630372 PMCID: PMC7350257 DOI: 10.3390/ijms21124507] [Citation(s) in RCA: 362] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Oncogenic activation of the phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB/AKT), and mammalian target of rapamycin (mTOR) pathway is a frequent event in prostate cancer that facilitates tumor formation, disease progression and therapeutic resistance. Recent discoveries indicate that the complex crosstalk between the PI3K-AKT-mTOR pathway and multiple interacting cell signaling cascades can further promote prostate cancer progression and influence the sensitivity of prostate cancer cells to PI3K-AKT-mTOR-targeted therapies being explored in the clinic, as well as standard treatment approaches such as androgen-deprivation therapy (ADT). However, the full extent of the PI3K-AKT-mTOR signaling network during prostate tumorigenesis, invasive progression and disease recurrence remains to be determined. In this review, we outline the emerging diversity of the genetic alterations that lead to activated PI3K-AKT-mTOR signaling in prostate cancer, and discuss new mechanistic insights into the interplay between the PI3K-AKT-mTOR pathway and several key interacting oncogenic signaling cascades that can cooperate to facilitate prostate cancer growth and drug-resistance, specifically the androgen receptor (AR), mitogen-activated protein kinase (MAPK), and WNT signaling cascades. Ultimately, deepening our understanding of the broader PI3K-AKT-mTOR signaling network is crucial to aid patient stratification for PI3K-AKT-mTOR pathway-directed therapies, and to discover new therapeutic approaches for prostate cancer that improve patient outcome.
Collapse
Affiliation(s)
| | | | | | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, Wales, UK; (B.Y.S.); (M.S.D.); (M.J.S.)
| |
Collapse
|
117
|
Gao A, Li F, Zhou Q, Chen L. Sestrin2 as a potential therapeutic target for cardiovascular diseases. Pharmacol Res 2020; 159:104990. [PMID: 32505836 DOI: 10.1016/j.phrs.2020.104990] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/17/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022]
Abstract
Sestrin2 is a cysteine sulfinyl reductase that plays crucial roles in regulation of antioxidant actions. Sestrin2 provides cytoprotection against multiple stress conditions, including hypoxia, endoplasmic reticulum (ER) stress and oxidative stress. Recent research reveals that upregulation of Sestrin2 is induced by various transcription factors such as p53 and activator protein 1 (AP-1), which further promotes AMP-activated protein kinase (AMPK) activation and inhibits mammalian target of rapamycin protein kinase (mTOR) signaling. Sestrin2 triggers autophagy activity to reduce cellular reactive oxygen species (ROS) levels by promoting nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) activation and Kelch-like ECH-associated protein 1 (Keap1) degradation, which plays a pivotal role in homeostasis of metabolic regulation. Under hypoxia and ER stress conditions, elevated Sestrin2 expression maintains cellular homeostasis through regulation of antioxidant genes. Sestrin2 is responsible for diminishing cellular ROS accumulation through autophagy via AMPK activation, which displays cardioprotection effect in cardiovascular diseases. In this review, we summarize the recent understanding of molecular structure, biological roles and biochemical functions of Sestrin2, and discuss the roles and mechanisms of Sestrin2 in autophagy, hypoxia and ER stress. Understanding the precise functions and exact mechanism of Sestrin2 in cellular homeostasis will provide the evidence for future experimental research and aid in the development of novel therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Anbo Gao
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, Hunan, People's Republic of China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421002, Hunan, People's Republic of China
| | - Feng Li
- Medical Shcool, Hunan University of Chinese Medicine, Changsha 410000, Hunan, People's Republic of China
| | - Qun Zhou
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, Hunan, People's Republic of China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, Hunan, People's Republic of China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421002, Hunan, People's Republic of China; Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Provincial Science and Technology Department, 28 Western Changshen Road, Hengyang 421002, Hunan, People's Republic of China.
| |
Collapse
|
118
|
Sun Y, Wu Y, Tang S, Liu H, Jiang Y. Sestrin proteins in cardiovascular disease. Clin Chim Acta 2020; 508:43-46. [PMID: 32407780 DOI: 10.1016/j.cca.2020.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the main cause of mortality and morbidity worldwide. Cardiovascular diseases such as AMI, AS, cardiac hypertrophy and cardiac fibrosis can be alleviated by controlling the inflammatory response, cellular proliferation, production of ROS and secretion of cytokines. Sestrins are considered to be protective proteins which can prevent age-related diseases. A potential mechanism is an inhibitory effect on mTORC, which depends on the sestrin-AMPK-mTORC pathway. Sestrins regulate the M1/M2 macrophage balance, level of ROS and certain cytokines during stress and cardiovascular diseases through the above pathway. Sestrins exert different functions in diverse organs and tissue. According to existing studies, the main functions of sestrins are strongly associated with the pathological features of cardiovascular disease and exert protective roles in cardiovascular disease. Based on the current evidence, we present a mini-review on the physiological functions and mechanism of sestrins in cardiology. The purpose of this review is to summarize the functions and mechanism of sestrins in common cardiovascular diseases, to raise awareness in clinicians that sestrins may be an important potential target for controlling progression of disease.
Collapse
Affiliation(s)
- Yunfeng Sun
- Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Huanghe Road, Urumqi City, XinJiang Province, China
| | - Yawei Wu
- Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Huanghe Road, Urumqi City, XinJiang Province, China
| | - Shuting Tang
- Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Huanghe Road, Urumqi City, XinJiang Province, China
| | - Hao Liu
- Guangdong Second Provincial General Hospital. Add: Xingangzhong Road, Guangzhou City, Guangdong Province, China
| | - Yingping Jiang
- Guangdong Second Provincial General Hospital. Add: Xingangzhong Road, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
119
|
Kanno A, Asahara SI, Furubayashi A, Masuda K, Yoshitomi R, Suzuki E, Takai T, Kimura-Koyanagi M, Matsuda T, Bartolome A, Hirota Y, Yokoi N, Inaba Y, Inoue H, Matsumoto M, Inoue K, Abe T, Wei FY, Tomizawa K, Ogawa W, Seino S, Kasuga M, Kido Y. GCN2 regulates pancreatic β cell mass by sensing intracellular amino acid levels. JCI Insight 2020; 5:128820. [PMID: 32376799 DOI: 10.1172/jci.insight.128820] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/01/2020] [Indexed: 01/09/2023] Open
Abstract
EIF2AK4, which encodes the amino acid deficiency-sensing protein GCN2, has been implicated as a susceptibility gene for type 2 diabetes in the Japanese population. However, the mechanism by which GCN2 affects glucose homeostasis is unclear. Here, we show that insulin secretion is reduced in individuals harboring the risk allele of EIF2AK4 and that maintenance of GCN2-deficient mice on a high-fat diet results in a loss of pancreatic β cell mass. Our data suggest that GCN2 senses amino acid deficiency in β cells and limits signaling by mechanistic target of rapamycin complex 1 to prevent β cell failure during the consumption of a high-fat diet.
Collapse
Affiliation(s)
- Ayumi Kanno
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Shun-Ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Ayuko Furubayashi
- Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Katsuhisa Masuda
- Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Risa Yoshitomi
- Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Emi Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Tomoko Takai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | | | - Tomokazu Matsuda
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Alberto Bartolome
- Naomi Berrie Diabetes Center and Department of Medicine, Columbia University, New York, New York, USA
| | - Yushi Hirota
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Norihide Yokoi
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuka Inaba
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Takaya Abe
- Laboratory for Animal Resource Development and.,Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Fan-Yan Wei
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Kasuga
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and.,Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Science, Kobe, Japan
| |
Collapse
|
120
|
Xu D, Dai W, Kutzler L, Lacko HA, Jefferson LS, Dennis MD, Kimball SR. ATF4-Mediated Upregulation of REDD1 and Sestrin2 Suppresses mTORC1 Activity during Prolonged Leucine Deprivation. J Nutr 2020; 150:1022-1030. [PMID: 31875479 PMCID: PMC7198311 DOI: 10.1093/jn/nxz309] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The protein kinase target of rapamycin (mTOR) in complex 1 (mTORC1) is activated by amino acids and in turn upregulates anabolic processes. Under nutrient-deficient conditions, e.g., amino acid insufficiency, mTORC1 activity is suppressed and autophagy is activated. Intralysosomal amino acids generated by autophagy reactivate mTORC1. However, sustained mTORC1 activation during periods of nutrient insufficiency would likely be detrimental to cellular homeostasis. Thus, mechanisms must exist to prevent amino acids released by autophagy from reactivating the kinase. OBJECTIVE The objective of the present study was to test whether mTORC1 activity is inhibited during prolonged leucine deprivation through ATF4-dependent upregulation of the mTORC1 suppressors regulated in development and DNA damage response 1 (REDD1) and Sestrin2. METHODS Mice (8 wk old; C57Bl/6 × 129SvEV) were food deprived (FD) overnight and one-half were refed the next morning. Mouse embryo fibroblasts (MEFs) deficient in ATF4, REDD1, and/or Sestrin2 were deprived of leucine for 0-16 h. mTORC1 activity and ATF4, REDD1, and Sestrin2 expression were assessed in liver and cell lysates. RESULTS Refeeding FD mice resulted in activation of mTORC1 in association with suppressed expression of both REDD1 and Sestrin2 in the liver. In cells in culture, mTORC1 exhibited a triphasic response to leucine deprivation, with an initial suppression followed by a transient reactivation from 2 to 4 h and a subsequent resuppression after 8 h. Resuppression occurred concomitantly with upregulated expression of ATF4, REDD1, and Sestrin2. However, in cells lacking ATF4, neither REDD1 nor Sestrin2 expression was upregulated by leucine deprivation, and resuppression of mTORC1 was absent. Moreover, in cells lacking either REDD1 or Sestrin2, mTORC1 resuppression was attenuated, and in cells lacking both proteins resuppression was further blunted. CONCLUSIONS The results suggest that leucine deprivation upregulates expression of both REDD1 and Sestrin2 in an ATF4-dependent manner, and that upregulated expression of both proteins is involved in resuppression of mTORC1 during prolonged leucine deprivation.
Collapse
Affiliation(s)
- Dandan Xu
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Weiwei Dai
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Lydia Kutzler
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Holly A Lacko
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Leonard S Jefferson
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA,Address correspondence to SRK (e-mail: )
| |
Collapse
|
121
|
Kovaleva IE, Tokarchuk AV, Zheltukhin AO, Dalina AA, Safronov GG, Evstafieva AG, Lyamzaev KG, Chumakov PM, Budanov AV. Mitochondrial localization of SESN2. PLoS One 2020; 15:e0226862. [PMID: 32287270 PMCID: PMC7156099 DOI: 10.1371/journal.pone.0226862] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
SESN2 is a member of the evolutionarily conserved sestrin protein family found in most of the Metazoa species. The SESN2 gene is transcriptionally activated by many stress factors, including metabolic derangements, reactive oxygen species (ROS), and DNA-damage. As a result, SESN2 controls ROS accumulation, metabolism, and cell viability. The best-known function of SESN2 is the inhibition of the mechanistic target of rapamycin complex 1 kinase (mTORC1) that plays a central role in support of cell growth and suppression of autophagy. SESN2 inhibits mTORC1 activity through interaction with the GATOR2 protein complex preventing an inhibitory effect of GATOR2 on the GATOR1 protein complex. GATOR1 stimulates GTPase activity of the RagA/B small GTPase, the component of RagA/B:RagC/D complex, preventing mTORC1 translocation to the lysosomes and its activation by the small GTPase Rheb. Despite the well-established role of SESN2 in mTORC1 inhibition, other SESN2 activities are not well-characterized. We recently showed that SESN2 could control mitochondrial function and cell death via mTORC1-independent mechanisms, and these activities might be explained by direct effects of SESN2 on mitochondria. In this work, we examined mitochondrial localization of SESN2 and demonstrated that SESN2 is located on mitochondria and can be directly involved in the regulation of mitochondrial functions.
Collapse
Affiliation(s)
| | | | - Andrei O. Zheltukhin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra A. Dalina
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Grigoriy G. Safronov
- Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra G. Evstafieva
- Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrei V. Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- * E-mail:
| |
Collapse
|
122
|
Integrating Mouse and Human Genetic Data to Move beyond GWAS and Identify Causal Genes in Cholesterol Metabolism. Cell Metab 2020; 31:741-754.e5. [PMID: 32197071 PMCID: PMC7184639 DOI: 10.1016/j.cmet.2020.02.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 01/14/2020] [Accepted: 02/20/2020] [Indexed: 12/27/2022]
Abstract
Identifying the causal gene(s) that connects genetic variation to a phenotype is a challenging problem in genome-wide association studies (GWASs). Here, we develop a systematic approach that integrates mouse liver co-expression networks with human lipid GWAS data to identify regulators of cholesterol and lipid metabolism. Through our approach, we identified 48 genes showing replication in mice and associated with plasma lipid traits in humans and six genes on the X chromosome. Among these 54 genes, 25 have no previously identified role in lipid metabolism. Based on functional studies and integration with additional human lipid GWAS datasets, we pinpoint Sestrin1 as a causal gene associated with plasma cholesterol levels in humans. Our validation studies demonstrate that Sestrin1 influences plasma cholesterol in multiple mouse models and regulates cholesterol biosynthesis. Our results highlight the power of combining mouse and human datasets for prioritization of human lipid GWAS loci and discovery of lipid genes.
Collapse
|
123
|
Lee S, Byun JK, Park M, Woo Kim S, Lee S, Kim JG, Lee IK, Choi YK, Park KG. Melatonin inhibits vascular smooth muscle cell proliferation and apoptosis through upregulation of Sestrin2. Exp Ther Med 2020; 19:3454-3460. [PMID: 32373191 PMCID: PMC7197251 DOI: 10.3892/etm.2020.8638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Excessive vascular smooth muscle cell (VSMC) proliferation contributes to the development of atherosclerosis and restenosis. Furthermore, apoptosis of VSMCs accelerates plaque rupture in the atherosclerotic vessels. Therefore, a strategy that regulates both VSMC proliferation and apoptosis is essential for the development of novel pharmacological tools for the treatment of atherosclerosis. Despite mounting evidence supporting the benefits of melatonin in diverse metabolic diseases, the role of melatonin in VSMC growth remains largely unknown. The present study revealed that melatonin inhibited both proliferation and apoptosis of primary cultured rat VSMCs. Melatonin induced mitochondrial energetic stress in VSMCs and subsequent induction of Sestrin2 via C/EBPβ. Melatonin-induced Sestrin2 suppressed mTORC1 activity in VSMCs, contributing to suppression of VSMC proliferation. Additionally, melatonin-induced upregulation of Sestrin2 blocked apoptosis by preventing excessive ROS generation. The results demonstrated that melatonin controlled VSMC proliferation and apoptosis via Sestrin2-mediated inhibition of mTORC1 and ROS scavenging. Therefore, melatonin should be considered as a lead compound for therapies aimed at preventing vessel lumen constriction during the course of atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Seunghyeong Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jun-Kyu Byun
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Republic of Korea.,Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Mihyang Park
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sung Woo Kim
- Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Sungwoo Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jung-Guk Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Yeon-Kyung Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Keun-Gyu Park
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea.,Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Republic of Korea.,Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
124
|
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 2020; 21:183-203. [PMID: 31937935 PMCID: PMC7102936 DOI: 10.1038/s41580-019-0199-y] [Citation(s) in RCA: 1650] [Impact Index Per Article: 330.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/21/2022]
Abstract
The mTOR pathway integrates a diverse set of environmental cues, such as growth factor signals and nutritional status, to direct eukaryotic cell growth. Over the past two and a half decades, mapping of the mTOR signalling landscape has revealed that mTOR controls biomass accumulation and metabolism by modulating key cellular processes, including protein synthesis and autophagy. Given the pathway's central role in maintaining cellular and physiological homeostasis, dysregulation of mTOR signalling has been implicated in metabolic disorders, neurodegeneration, cancer and ageing. In this Review, we highlight recent advances in our understanding of the complex regulation of the mTOR pathway and discuss its function in the context of physiology, human disease and pharmacological intervention.
Collapse
Affiliation(s)
- Grace Y Liu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA.
| |
Collapse
|
125
|
González A, Hall MN, Lin SC, Hardie DG. AMPK and TOR: The Yin and Yang of Cellular Nutrient Sensing and Growth Control. Cell Metab 2020; 31:472-492. [PMID: 32130880 DOI: 10.1016/j.cmet.2020.01.015] [Citation(s) in RCA: 491] [Impact Index Per Article: 98.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The AMPK (AMP-activated protein kinase) and TOR (target-of-rapamycin) pathways are interlinked, opposing signaling pathways involved in sensing availability of nutrients and energy and regulation of cell growth. AMPK (Yin, or the "dark side") is switched on by lack of energy or nutrients and inhibits cell growth, while TOR (Yang, or the "bright side") is switched on by nutrient availability and promotes cell growth. Genes encoding the AMPK and TOR complexes are found in almost all eukaryotes, suggesting that these pathways arose very early during eukaryotic evolution. During the development of multicellularity, an additional tier of cell-extrinsic growth control arose that is mediated by growth factors, but these often act by modulating nutrient uptake so that AMPK and TOR remain the underlying regulators of cellular growth control. In this review, we discuss the evolution, structure, and regulation of the AMPK and TOR pathways and the complex mechanisms by which they interact.
Collapse
Affiliation(s)
- Asier González
- Biozentrum, University of Basel, CH4056 Basel, Switzerland
| | - Michael N Hall
- Biozentrum, University of Basel, CH4056 Basel, Switzerland
| | - Sheng-Cai Lin
- School of Life Sciences, Xiamen University, Xiamen, 361102 Fujian, China
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, UK.
| |
Collapse
|
126
|
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) controls cell growth and metabolism in response to various environmental inputs, especially amino acids. In fact, the activity of mTORC1 is highly sensitive to changes in amino acid levels. Over past decades, a variety of proteins have been identified as participating in the mTORC1 pathway regulated by amino acids. Classically, the Rag guanosine triphosphatases (GTPases), which reside on the lysosome, transmit amino acid availability to the mTORC1 pathway and recruit mTORC1 to the lysosome upon amino acid sufficiency. Recently, several sensors of leucine, arginine, and S-adenosylmethionine for the amino acid-stimulated mTORC1 pathway have been coming to light. Characterization of these sensors is requisite for understanding how cells adjust amino acid sensing pathways to their different needs. In this review, we summarize recent advances in amino acid sensing mechanisms that regulate mTORC1 activity and highlight these identified sensors that accurately transmit specific amino acid signals to the mTORC1 pathway.
Collapse
Affiliation(s)
- Xiu-Zhi Li
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan 430070, China
| | - Xiang-Hua Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan 430070, China
| |
Collapse
|
127
|
Lee S, Shin J, Hong Y, Shin SM, Shin HW, Shin J, Lee SK, Park HW. Sestrin2 alleviates palmitate-induced endoplasmic reticulum stress, apoptosis, and defective invasion of human trophoblast cells. Am J Reprod Immunol 2020; 83:e13222. [PMID: 31958198 DOI: 10.1111/aji.13222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
PROBLEM Maternal obesity induces elevated saturated fatty acid palmitate levels in the blood and causes pregnancy complications such as gestational diabetes, preeclampsia, fetal growth abnormalities, and stillbirth. Sestrin2, a highly conserved stress-inducible protein, is involved in the cellular responses of various stress conditions and homeostatic regulation. However, the effects of Sestrin2 on trophoblast cells have not yet been investigated. Here, we investigated the role of Sestrin2 in palmitate-induced lipotoxicity and its underlying mechanisms in human first-trimester trophoblast cells (Sw.71). METHOD OF STUDY Mouse placental tissues were obtained from low-fat diet-fed mice (n = 14) and high-fat diet-fed mice (n = 14) at gestation day 17.5. Sw.71 cells were treated with palmitate or bovine serum albumin as vehicle controls. The role of Sestrin2 in palmitate-induced lipotoxicity was examined by immunocytochemistry, immunoblot analysis, quantitative real-time PCR, and invasion assay. RESULTS Expression of placental Sestrin2 was elevated in high-fat diet-fed dams compared to that of low-fat diet-fed dams. Prolonged treatment of Sw.71 cells with palmitate-induced endoplasmic reticulum (ER) stress-dependent expressions of Sestrin2 protein and mRNA, and the treatment also triggered apoptosis. Knockdown of Sestrin2 increased palmitate-mediated ER stress, inflammatory signaling, and apoptosis. Furthermore, Sestrin2 suppressed impaired trophoblast invasion caused by palmitate and attenuated palmitate-induced ER stress and inflammation via AMPK/mTORC1 pathways. CONCLUSION Our study provides the relationship between Sestrin2, AMPK/mTORC1 pathway, and trophoblast function, suggesting that Sestrin2 may be a novel potential therapeutic target for the prevention of pregnancy complications.
Collapse
Affiliation(s)
- Solji Lee
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Jiha Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Yeji Hong
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Korea
| | - Seong Min Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Hye Won Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea
| | - Jongdae Shin
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon, Korea.,Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
128
|
Autophagy as a Cellular Stress Response Mechanism in the Nervous System. J Mol Biol 2020; 432:2560-2588. [PMID: 31962122 DOI: 10.1016/j.jmb.2020.01.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/11/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Cells of an organism face with various types of insults during their lifetime. Exposure to toxins, metabolic problems, ischaemia/reperfusion, physical trauma, genetic diseases, neurodegenerative diseases are among the conditions that trigger cellular stress responses. In this context, autophagy is one of the mechanisms that supports cell survival under stressful conditions. Autophagic vesicle engulfs the cargo and transports it to lysosome for degradation and turnover. As such, autophagy eliminates abnormal proteins, clears damaged organelles, limits oxidative stress and helps to improve metabolic balance. Nervous system cells and particularly postmitotic neurons are highly sensitive to a spectrum of insults, and autophagy emerges as one of the key stress response mechanism, ensuring health and survival of these vulnerable cell types. In this review, we will overview mechanisms through which cells cope with stress, and how these stress responses regulate autophagy, with a special focus on the nervous system.
Collapse
|
129
|
Kim M, Sujkowski A, Namkoong S, Gu B, Cobb T, Kim B, Kowalsky AH, Cho CS, Semple I, Ro SH, Davis C, Brooks SV, Karin M, Wessells RJ, Lee JH. Sestrins are evolutionarily conserved mediators of exercise benefits. Nat Commun 2020; 11:190. [PMID: 31929512 PMCID: PMC6955242 DOI: 10.1038/s41467-019-13442-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 11/06/2019] [Indexed: 01/04/2023] Open
Abstract
Exercise is among the most effective interventions for age-associated mobility decline and metabolic dysregulation. Although long-term endurance exercise promotes insulin sensitivity and expands respiratory capacity, genetic components and pathways mediating the metabolic benefits of exercise have remained elusive. Here, we show that Sestrins, a family of evolutionarily conserved exercise-inducible proteins, are critical mediators of exercise benefits. In both fly and mouse models, genetic ablation of Sestrins prevents organisms from acquiring metabolic benefits of exercise and improving their endurance through training. Conversely, Sestrin upregulation mimics both molecular and physiological effects of exercise, suggesting that it could be a major effector of exercise metabolism. Among the various targets modulated by Sestrin in response to exercise, AKT and PGC1α are critical for the Sestrin effects in extending endurance. These results indicate that Sestrin is a key integrating factor that drives the benefits of chronic exercise to metabolism and physical endurance.
Collapse
Affiliation(s)
- Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alyson Sujkowski
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Sim Namkoong
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Bondong Gu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tyler Cobb
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Boyoung Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Allison H Kowalsky
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ian Semple
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Seung-Hyun Ro
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, University of Nebraska, Lincoln, NE, 68588, USA
| | - Carol Davis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Susan V Brooks
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Karin
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Robert J Wessells
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
130
|
Cordover E, Wei J, Patel C, Shan NL, Gionco J, Sargsyan D, Wu R, Cai L, Kong AN, Jacinto E, Minden A. KPT-9274, an Inhibitor of PAK4 and NAMPT, Leads to Downregulation of mTORC2 in Triple Negative Breast Cancer Cells. Chem Res Toxicol 2020; 33:482-491. [PMID: 31876149 DOI: 10.1021/acs.chemrestox.9b00376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Triple negative breast cancer (TNBC) is difficult to treat due to lack of druggable targets. We have found that treatment with the small molecule inhibitor KPT-9274 inhibits growth of TNBC cells and eventually leads to cell death. KPT-9274 is a dual specific inhibitor of PAK4 and Nicotinamide Phosphoribosyltransferase (NAMPT). The PAK4 protein kinase is often highly expressed in TNBC cells and has important roles in cell growth, survival, and migration. Previously we have found that inhibition of PAK4 leads to growth inhibition of TNBC cells both in vitro and in vivo. Likewise, NAMPT has been shown to be dysregulated in cancer due to its role in cell metabolism. In order to understand better how treating cells with KPT-9274 abrogates TNBC cell growth, we carried out an RNA sequencing of TNBC cells treated with KPT-9274. As a result, we identified Rictor as an important target that is inhibited in the KPT-9274 treated cells. Conversely, we found that Rictor is predicted to be activated when PAK4 is overexpressed in cells, which suggests a role for PAK4 in the regulation of Rictor. Rictor is a component of mTORC2, one of the complexes formed by the serine/threonine kinase mTOR. mTOR is important for the control of cell growth and metabolism. Our results suggest a new mechanism by which the KPT-9274 compound may block the growth of breast cancer cells, which is via inhibition of mTORC2 signaling. Consistent with this, sequencing analysis of PAK4 overexpressing cells indicates that PAK4 has a role in activation of the mTOR pathway.
Collapse
Affiliation(s)
- Emma Cordover
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Janet Wei
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Chadni Patel
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Naing Lin Shan
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - John Gionco
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Li Cai
- Department of Biomedical Engineering , Rutgers, The State University of New Jersey , 599 Taylor Road , Piscataway , New Jersey 08854 , United States
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology , Rutgers-Robert Wood Johnson Medical School , 683 Hoes Lane , Piscataway , New Jersey 08854 , United States
| | - Audrey Minden
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy , Rutgers, The State University of New Jersey , 164 Frelinghuysen Road , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
131
|
Kowalsky AH, Namkoong S, Mettetal E, Park HW, Kazyken D, Fingar DC, Lee JH. The GATOR2-mTORC2 axis mediates Sestrin2-induced AKT Ser/Thr kinase activation. J Biol Chem 2020; 295:1769-1780. [PMID: 31915252 DOI: 10.1074/jbc.ra119.010857] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Sestrins represent a family of stress-inducible proteins that prevent the progression of many age- and obesity-associated disorders. Endogenous Sestrins maintain insulin-dependent AKT Ser/Thr kinase (AKT) activation during high-fat diet-induced obesity, and overexpressed Sestrins activate AKT in various cell types, including liver and skeletal muscle cells. Although Sestrin-mediated AKT activation improves metabolic parameters, the mechanistic details underlying such improvement remain elusive. Here, we investigated how Sestrin2, the Sestrin homolog highly expressed in liver, induces strong AKT activation. We found that two known targets of Sestrin2, mTOR complex (mTORC) 1 and AMP-activated protein kinase, are not required for Sestrin2-induced AKT activation. Rather, phosphoinositol 3-kinase and mTORC2, kinases upstream of AKT, were essential for Sestrin2-induced AKT activation. Among these kinases, mTORC2 catalytic activity was strongly up-regulated upon Sestrin2 overexpression in an in vitro kinase assay, indicating that mTORC2 may represent the major link between Sestrin2 and AKT. As reported previously, Sestrin2 interacted with mTORC2; however, we found here that this interaction occurs indirectly through GATOR2, a pentameric protein complex that directly interacts with Sestrin2. Deleting or silencing WDR24 (WD repeat domain 24), the GATOR2 component essential for the Sestrin2-GATOR2 interaction, or WDR59, the GATOR2 component essential for the GATOR2-mTORC2 interaction, completely ablated Sestrin2-induced AKT activation. We also noted that Sestrin2 also directly binds to the pleckstrin homology domain of AKT and induces AKT translocation to the plasma membrane. These results uncover a signaling mechanism whereby Sestrin2 activates AKT through GATOR2 and mTORC2.
Collapse
Affiliation(s)
- Allison Ho Kowalsky
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Sim Namkoong
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109; Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Eric Mettetal
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Hwan-Woo Park
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109; Department of Cell Biology, Myunggok Medical Research Institute, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Dubek Kazyken
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Diane C Fingar
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109.
| |
Collapse
|
132
|
Huang M, Geug Kim H, Zhong X, Dong C, Zhang B, Fang Z, Zhang Y, Lu X, Saxena R, Liu Y, Zhang C, Liangpunsakul S, Dong XC. Sestrin 3 Protects Against Diet-Induced Nonalcoholic Steatohepatitis in Mice Through Suppression of Transforming Growth Factor β Signal Transduction. Hepatology 2020; 71:76-92. [PMID: 31215672 PMCID: PMC6920605 DOI: 10.1002/hep.30820] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022]
Abstract
Sestrin 3 (Sesn3) belongs to the three-member sestrin protein family. Sestrins have been implicated in antioxidative stress, adenosine monophosphate-activated protein kinase and mammalian target of rapamycin signal transduction, and metabolic homeostasis. However, the role of Sesn3 in the development of nonalcoholic steatohepatitis (NASH) has not been previously studied. In this work, we generated Sesn3 whole-body knockout and liver-specific transgenic mice to investigate the hepatic function of Sesn3 in diet-induced NASH. With only 4 weeks of dietary treatment, Sesn3 knockout mice developed severe NASH phenotype as characterized by hepatic steatosis, inflammation, and fibrosis. Strikingly, after 8-week feeding with a NASH-inducing diet, Sesn3 transgenic mice were largely protected against NASH development. Transcriptomic analysis revealed that multiple extracellular matrix-related processes were up-regulated, including transforming growth factor β (TGF-β) signaling and collagen production. Further biochemical and cell biological analyses have illustrated a critical control of the TGF-β-mothers against decapentaplegic homolog (Smad) pathway by Sesn3 at the TGF-β receptor and Smad3 levels. First, Sesn3 inhibits the TGF-β receptor through an interaction with Smad7; second, Sesn3 directly inhibits the Smad3 function through protein-protein interaction and cytosolic retention. Conclusion: Sesn3 is a critical regulator of the extracellular matrix and hepatic fibrosis by suppression of TGF-β-Smad3 signaling.
Collapse
Affiliation(s)
- Menghao Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Hyeong Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Xiaolin Zhong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN,Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Chuanpeng Dong
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN
| | | | - Zhigang Fang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Yang Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Xiaoyu Lu
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Yunlong Liu
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Chi Zhang
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN,Roudebush Veterans Administration Medical Center, Indianapolis, IN.,Corresponding authors: Suthat Liangpunsakul, MD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA, ; X. Charlie Dong, PhD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA, Phone: 317-278-1097, Fax: 317-274-4686,
| | - X. Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN,Corresponding authors: Suthat Liangpunsakul, MD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA, ; X. Charlie Dong, PhD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA, Phone: 317-278-1097, Fax: 317-274-4686,
| |
Collapse
|
133
|
ER stress contributes to autophagy induction by adiponectin in macrophages: Implication in cell survival and suppression of inflammatory response. Cytokine 2019; 127:154959. [PMID: 31877413 DOI: 10.1016/j.cyto.2019.154959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022]
Abstract
Adiponectin, the most abundant adipokine, exhibits various physiological functions. In addition to its critical role in lipid metabolism, recent studies have demonstrated its potent anti-inflammatory and cytoprotective properties. Accumulating evidence suggests that autophagy plays a critical role in various biological responses by adiponectin. However, the underlying mechanisms remain elusive. Herein, we investigated the role of ER stress in adiponectin-induced autophagy and its functional roles in biological responses by adiponectin in macrophages. In this study, globular adiponectin (gAcrp) significantly increased the expression of various ER stress markers in both RAW 264.7 and primary peritoneal macrophages. In addition, inhibition of ER stress by treatment with tauroursodeoxycholic acid (TUDCA) or gene silencing of CHOP prominently suppressed gAcrp-induced autophagy. Treatment with gAcrp also induced significant increase in sestrin2 expression. Interestingly, knockdown of sestrin2 prevented autophagy induction and inhibition of ER stress abrogated sestrin2 induction by gAcrp, collectively implying that ER stress critically contributes to gAcrp-induced autophagy activation via sestrin2 induction. Moreover, pretreatment with TUDCA restored suppression of TNF-α and IL-1β expression and attenuated the enhanced viability of macrophages induced by gAcrp. Taken together, these findings indicate the potential role of ER stress in autophagy activation, modulation of inflammatory responses, and cell survival by gAcrp in macrophages.
Collapse
|
134
|
Harvey RF, Pöyry TAA, Stoneley M, Willis AE. Signaling from mTOR to eIF2α mediates cell migration in response to the chemotherapeutic doxorubicin. Sci Signal 2019; 12:12/612/eaaw6763. [PMID: 31848319 DOI: 10.1126/scisignal.aaw6763] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After exposure to cytotoxic chemotherapeutics, tumor cells alter their translatome to promote cell survival programs through the regulation of eukaryotic initiation factor 4F (eIF4F) and ternary complex. Compounds that block mTOR signaling and eIF4F complex formation, such as rapamycin and its analogs, have been used in combination therapies to enhance cell killing, although their success has been limited. This is likely because the cross-talk between signaling pathways that coordinate eIF4F regulation with ternary complex formation after treatment with genotoxic therapeutics has not been fully explored. Here, we described a regulatory pathway downstream of p53 in which inhibition of mTOR after DNA damage promoted cross-talk signaling and led to eIF2α phosphorylation. We showed that eIF2α phosphorylation did not inhibit protein synthesis but was instead required for cell migration and that pharmacologically blocking this pathway with either ISRIB or trazodone limited cell migration. These results support the notion that therapeutic targeting of eIF2α signaling could restrict tumor cell metastasis and invasion and could be beneficial to subsets of patients with cancer.
Collapse
Affiliation(s)
- Robert F Harvey
- Medical Research Council Toxicology Unit, University of Cambridge, Lancaster Rd., Leicester LE1 9HN, UK
| | - Tuija A A Pöyry
- Medical Research Council Toxicology Unit, University of Cambridge, Lancaster Rd., Leicester LE1 9HN, UK
| | - Mark Stoneley
- Medical Research Council Toxicology Unit, University of Cambridge, Lancaster Rd., Leicester LE1 9HN, UK
| | - Anne E Willis
- Medical Research Council Toxicology Unit, University of Cambridge, Lancaster Rd., Leicester LE1 9HN, UK.
| |
Collapse
|
135
|
Ding B, Haidurov A, Chawla A, Parmigiani A, van de Kamp G, Dalina A, Yuan F, Lee JH, Chumakov PM, Grossman SR, Budanov AV. p53-inducible SESTRINs might play opposite roles in the regulation of early and late stages of lung carcinogenesis. Oncotarget 2019; 10:6997-7009. [PMID: 31857853 PMCID: PMC6916756 DOI: 10.18632/oncotarget.27367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/17/2019] [Indexed: 01/13/2023] Open
Abstract
SESTRINs (SESN1-3) are proteins encoded by an evolutionarily conserved gene family that plays an important role in the regulation of cell viability and metabolism in response to stress. Many of the effects of SESTRINs are mediated by negative and positive regulation of mechanistic target of rapamycin kinase complexes 1 and 2 (mTORC1 and mTORC2), respectively, that are often deregulated in human cancers where they support cell growth, proliferation, and cell viability. Besides their effects on regulation of mTORC1/2, SESTRINs also control the accumulation of reactive oxygen species, cell death, and mitophagy. SESN1 and SESN2 are transcriptional targets of tumor suppressor protein p53 and may mediate tumor suppressor activities of p53. Therefore, we conducted studies based on a mouse lung cancer model and human lung adenocarcinoma A549 cells to evaluate the potential impact of SESN1 and SESN2 on lung carcinogenesis. While we observed that expression of SESN1 and SESN2 is often decreased in human tumors, inactivation of Sesn2 in mice positively regulates tumor growth through a mechanism associated with activation of AKT, while knockout of Sesn1 has no additional impact on carcinogenesis in Sesn2-deficient mice. However, inactivation of SESN1 and/or SESN2 in A549 cells accelerates cell proliferation and imparts resistance to cell death in response to glucose starvation. We propose that despite their contribution to early tumor growth, SESTRINs might suppress late stages of carcinogenesis through inhibition of cell proliferation or activation of cell death in conditions of nutrient deficiency.
Collapse
Affiliation(s)
- Boxiao Ding
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.,These authors contributed equally to this work
| | - Alexander Haidurov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,These authors contributed equally to this work
| | - Ayesha Chawla
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Anita Parmigiani
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Gerarda van de Kamp
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Alexandra Dalina
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Fang Yuan
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia
| | - Steven R Grossman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.,VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrei V Budanov
- Engelhardt Institute of Molecular Biology, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russia.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
136
|
Iffland PH, Carson V, Bordey A, Crino PB. GATORopathies: The role of amino acid regulatory gene mutations in epilepsy and cortical malformations. Epilepsia 2019; 60:2163-2173. [PMID: 31625153 PMCID: PMC7155771 DOI: 10.1111/epi.16370] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) pathway has been implicated in a growing number of malformations of cortical development (MCD) associated with intractable epilepsy. Mutations in single genes encoding mTOR pathway regulatory proteins have been linked to MCD such as focal cortical dysplasia (FCD) types IIa and IIb, hemimegalencephaly (HME), and megalencephaly. Recent studies have demonstrated that the GATOR1 protein complex, comprised of DEPDC5, NPRL3, and NPRL2, plays a pivotal role in regulating mTOR signaling in response to cellular amino acid levels and that mutations in DEPDC5, NPRL3, or NPRL2 are linked to FCD, HME, and seizures. Histopathological analysis of FCD and HME tissue specimens resected from individuals harboring DEPDC5, NPRL3, or NPRL2 gene mutations reveals hyperactivation of mTOR pathway signaling. Family pedigrees carrying mutations in either DEPDC5 or NPRL3 share clinical phenotypes of epilepsy and MCD, as well as intellectual and neuropsychiatric disabilities. Interestingly, some individuals with seizures associated with DEPDC5, NPRL3, or NPRL2 variants exhibit normal brain imaging suggesting either occult MCD or a role for these genes in non-lesional neocortical epilepsy. Mouse models resulting from knockdown or knockout of either Depdc5 or Nprl3 exhibit altered cortical lamination, neuronal dysmorphogenesis, and enhanced neuronal excitability as reported in models resulting from direct mTOR activation through expression of its canonical activator RHEB. The role of the GATOR1 proteins in regulating mTOR signaling suggest plausible options for mTOR inhibition in the treatment of epilepsy associated with mutations in DEPDC5, NPRL3, or NPRL2.
Collapse
Affiliation(s)
- Philip H. Iffland
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vincent Carson
- The Clinic for Special Children, Strasburg, Pennsylvania
| | - Angelique Bordey
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Peter B. Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
137
|
Wei Y, Bettedi L, Ting CY, Kim K, Zhang Y, Cai J, Lilly MA. The GATOR complex regulates an essential response to meiotic double-stranded breaks in Drosophila. eLife 2019; 8:e42149. [PMID: 31650955 PMCID: PMC6834368 DOI: 10.7554/elife.42149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/13/2019] [Indexed: 01/18/2023] Open
Abstract
The TORC1 regulator GATOR1/SEACIT controls meiotic entry and early meiotic events in yeast. However, how metabolic pathways influence meiotic progression in metazoans remains poorly understood. Here we examine the role of the TORC1 regulators GATOR1 and GATOR2 in the response to meiotic double-stranded breaks (DSB) during Drosophila oogenesis. We find that in mutants of the GATOR2 component mio, meiotic DSBs trigger the constitutive downregulation of TORC1 activity and a permanent arrest in oocyte growth. Conversely, in GATOR1 mutants, high TORC1 activity results in the delayed repair of meiotic DSBs and the hyperactivation of p53. Unexpectedly, we found that GATOR1 inhibits retrotransposon expression in the presence of meiotic DSBs in a pathway that functions in parallel to p53. Thus, our studies have revealed a link between oocyte metabolism, the repair of meiotic DSBs and retrotransposon expression.
Collapse
Affiliation(s)
- Youheng Wei
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouChina
| | - Lucia Bettedi
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Chun-Yuan Ting
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Kuikwon Kim
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Yingbiao Zhang
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Jiadong Cai
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouChina
| | - Mary A Lilly
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
138
|
Beaumatin F, O'Prey J, Barthet VJA, Zunino B, Parvy JP, Bachmann AM, O'Prey M, Kania E, Gonzalez PS, Macintosh R, Lao LY, Nixon C, Lopez J, Long JS, Tait SWG, Ryan KM. mTORC1 Activation Requires DRAM-1 by Facilitating Lysosomal Amino Acid Efflux. Mol Cell 2019; 76:163-176.e8. [PMID: 31492633 PMCID: PMC6892261 DOI: 10.1016/j.molcel.2019.07.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/15/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023]
Abstract
Sensing nutrient availability is essential for appropriate cellular growth, and mTORC1 is a major regulator of this process. Mechanisms causing mTORC1 activation are, however, complex and diverse. We report here an additional important step in the activation of mTORC1, which regulates the efflux of amino acids from lysosomes into the cytoplasm. This process requires DRAM-1, which binds the membrane carrier protein SCAMP3 and the amino acid transporters SLC1A5 and LAT1, directing them to lysosomes and permitting efficient mTORC1 activation. Consequently, we show that loss of DRAM-1 also impacts pathways regulated by mTORC1, including insulin signaling, glycemic balance, and adipocyte differentiation. Interestingly, although DRAM-1 can promote autophagy, this effect on mTORC1 is autophagy independent, and autophagy only becomes important for mTORC1 activation when DRAM-1 is deleted. These findings provide important insights into mTORC1 activation and highlight the importance of DRAM-1 in growth control, metabolic homeostasis, and differentiation.
Collapse
Affiliation(s)
- Florian Beaumatin
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jim O'Prey
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Valentin J A Barthet
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Barbara Zunino
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jean-Philippe Parvy
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | | | - Margaret O'Prey
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Elżbieta Kania
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Pablo Sierra Gonzalez
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Robin Macintosh
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Laurence Y Lao
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Colin Nixon
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jonathan Lopez
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jaclyn S Long
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
139
|
Aiello A, Farzaneh F, Candore G, Caruso C, Davinelli S, Gambino CM, Ligotti ME, Zareian N, Accardi G. Immunosenescence and Its Hallmarks: How to Oppose Aging Strategically? A Review of Potential Options for Therapeutic Intervention. Front Immunol 2019; 10:2247. [PMID: 31608061 PMCID: PMC6773825 DOI: 10.3389/fimmu.2019.02247] [Citation(s) in RCA: 465] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Aging is accompanied by remodeling of the immune system. With time, this leads to a decline in immune efficacy, resulting in increased vulnerability to infectious diseases, diminished responses to vaccination, and a susceptibility to age-related inflammatory diseases. An age-associated immune alteration, extensively reported in previous studies, is the reduction in the number of peripheral blood naïve cells, with a relative increase in the frequency of memory cells. These two alterations, together with inflamm-aging, are considered the hallmarks of immunosenescence. Because aging is a plastic process, it is influenced by both nutritional and pharmacological interventions. Therefore, the role of nutrition and of immunomodulation in immunosenescence is discussed, due to the multifactorial influence on these hallmarks. The close connection between nutrition, intake of bioactive nutrients and supplements, immune function, and inflammation demonstrate the key role of dietary strategies as regulators of immune response and inflammatory status, hence as possible modulators of the rate of immunosenescence. In addition, potential options for therapeutic intervention are clarified. In particular, the use of interleukin-7 as growth factor for naïve T cells, the function of checkpoint inhibitors in improving T cell responses during aging and, the potential of drugs that inhibit mitogen-activated protein kinases and their interaction with nutrient signaling pathways are discussed. Finally, it is suggested that the inclusion of appropriate combinations of toll-like receptor agonists may enhance the efficacy of vaccination in older adults.
Collapse
Affiliation(s)
- Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Farzin Farzaneh
- Molecular Medicine Group, Department of Hematological Medicine, School of Cancer & Pharmaceutical Sciences, The Rayne Institute, King's College London, London, United Kingdom
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Caterina Maria Gambino
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Nahid Zareian
- Molecular Medicine Group, Department of Hematological Medicine, School of Cancer & Pharmaceutical Sciences, The Rayne Institute, King's College London, London, United Kingdom
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
140
|
Sánchez-Álvarez M, Strippoli R, Donadelli M, Bazhin AV, Cordani M. Sestrins as a Therapeutic Bridge between ROS and Autophagy in Cancer. Cancers (Basel) 2019; 11:cancers11101415. [PMID: 31546746 PMCID: PMC6827145 DOI: 10.3390/cancers11101415] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/15/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023] Open
Abstract
The regulation of Reactive Oxygen Species (ROS) levels and the contribution therein from networks regulating cell metabolism, such as autophagy and the mTOR-dependent nutrient-sensing pathway, constitute major targets for selective therapeutic intervention against several types of tumors, due to their extensive rewiring in cancer cells as compared to healthy cells. Here, we discuss the sestrin family of proteins—homeostatic transducers of oxidative stress, and drivers of antioxidant and metabolic adaptation—as emerging targets for pharmacological intervention. These adaptive regulators lie at the intersection of those two priority nodes of interest in antitumor intervention—ROS control and the regulation of cell metabolism and autophagy—therefore, they hold the potential not only for the development of completely novel compounds, but also for leveraging on synergistic strategies with current options for tumor therapy and classification/stadiation to achieve personalized medicine.
Collapse
Affiliation(s)
- Miguel Sánchez-Álvarez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC). Madrid 28029, Spain.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome 00161, Italy.
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona 37134, Italy.
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplantation Surgery, Ludwig-Maximilians University, Munich 81377, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich 80366, Germany.
| | - Marco Cordani
- IMDEA Nanociencia, C/Faraday 9, Ciudad Universitaria de Cantoblanco, Madrid 28049, Spain..
| |
Collapse
|
141
|
Abstract
The proteasome degrades most cellular proteins in a controlled and tightly regulated manner and thereby controls many processes, including cell cycle, transcription, signalling, trafficking and protein quality control. Proteasomal degradation is vital in all cells and organisms, and dysfunction or failure of proteasomal degradation is associated with diverse human diseases, including cancer and neurodegeneration. Target selection is an important and well-established way to control protein degradation. In addition, mounting evidence indicates that cells adjust proteasome-mediated degradation to their needs by regulating proteasome abundance through the coordinated expression of proteasome subunits and assembly chaperones. Central to the regulation of proteasome assembly is TOR complex 1 (TORC1), which is the master regulator of cell growth and stress. This Review discusses how proteasome assembly and the regulation of proteasomal degradation are integrated with cellular physiology, including the interplay between the proteasome and autophagy pathways. Understanding these mechanisms has potential implications for disease therapy, as the misregulation of proteasome function contributes to human diseases such as cancer and neurodegeneration.
Collapse
|
142
|
Liu Y, Kim HG, Dong E, Dong C, Huang M, Liu Y, Liangpunsakul S, Dong XC. Sesn3 deficiency promotes carcinogen-induced hepatocellular carcinoma via regulation of the hedgehog pathway. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2685-2693. [PMID: 31351129 DOI: 10.1016/j.bbadis.2019.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023]
Abstract
Sestrin 3 (Sesn3) belongs to a small protein family that has been implicated in multiple biological processes including anti-oxidative stress, anti-aging, cell signaling, and metabolic homeostasis. However, the role of Sesn3 in hepatocellular carcinoma (HCC) remains unclear. Here we generated a Sesn3 knockout mouse model and induced HCC development by a combination of a single dose of diethylnitrosamine and chronic feeding of a choline deficient-high fat diet. After 6 months of the dietary treatment, Sesn3 knockout mice developed more severe HCC with higher levels of alpha-fetoprotein, arginase 1, and cytokeratin 19, but also higher metastatic rates than wild-type mice. Histological analysis revealed elevated extracellular matrix and cancer stem cell markers including Acta2, Cd44, and Cd133. Signaling analysis showed activated IL6-Stat3 and Akt pathways. Biochemical and microscopic analyses uncovered a novel inhibitory regulation of Gli2, a downstream transcription factor of the hedgehog signaling, by Sesn3. Two of the Gli2-regulated genes - Pdgfrb and Cd44 were upregulated in the Sesn3-deficient liver tissue. In conclusion, our data suggest that Sesn3 plays a critical tumor suppressor role in the liver partly through the inhibition of the hedgehog signaling.
Collapse
Affiliation(s)
- Yunjian Liu
- Department of Hepatobiliary Surgery, the Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province 332000, China; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hyeong Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Chuanpeng Dong
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Suthat Liangpunsakul
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaocheng Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
143
|
Hua H, Kong Q, Zhang H, Wang J, Luo T, Jiang Y. Targeting mTOR for cancer therapy. J Hematol Oncol 2019; 12:71. [PMID: 31277692 PMCID: PMC6612215 DOI: 10.1186/s13045-019-0754-1] [Citation(s) in RCA: 614] [Impact Index Per Article: 102.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. mTOR is usually assembled into several complexes such as mTOR complex 1/2 (mTORC1/2). In cooperation with raptor, rictor, LST8, and mSin1, key components in mTORC1 or mTORC2, mTOR catalyzes the phosphorylation of multiple targets such as ribosomal protein S6 kinase β-1 (S6K1), eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), Akt, protein kinase C (PKC), and type-I insulin-like growth factor receptor (IGF-IR), thereby regulating protein synthesis, nutrients metabolism, growth factor signaling, cell growth, and migration. Activation of mTOR promotes tumor growth and metastasis. Many mTOR inhibitors have been developed to treat cancer. While some of the mTOR inhibitors have been approved to treat human cancer, more mTOR inhibitors are being evaluated in clinical trials. Here, we update recent advances in exploring mTOR signaling and the development of mTOR inhibitors for cancer therapy. In addition, we discuss the mechanisms underlying the resistance to mTOR inhibitors in cancer cells.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingbin Kong
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
144
|
Sciarretta S, Forte M, Frati G, Sadoshima J. New Insights Into the Role of mTOR Signaling in the Cardiovascular System. Circ Res 2019; 122:489-505. [PMID: 29420210 DOI: 10.1161/circresaha.117.311147] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR (mechanistic target of rapamycin) is a master regulator of several crucial cellular processes, including protein synthesis, cellular growth, proliferation, autophagy, lysosomal function, and cell metabolism. mTOR interacts with specific adaptor proteins to form 2 multiprotein complexes, called mTORC1 (mTOR complex 1) and mTORC2 (mTOR complex 2). In the cardiovascular system, the mTOR pathway regulates both physiological and pathological processes in the heart. It is needed for embryonic cardiovascular development and for maintaining cardiac homeostasis in postnatal life. Studies involving mTOR loss-of-function models revealed that mTORC1 activation is indispensable for the development of adaptive cardiac hypertrophy in response to mechanical overload. mTORC2 is also required for normal cardiac physiology and ensures cardiomyocyte survival in response to pressure overload. However, partial genetic or pharmacological inhibition of mTORC1 reduces cardiac remodeling and heart failure in response to pressure overload and chronic myocardial infarction. In addition, mTORC1 blockade reduces cardiac derangements induced by genetic and metabolic disorders and has been reported to extend life span in mice. These studies suggest that pharmacological targeting of mTOR may represent a therapeutic strategy to confer cardioprotection, although clinical evidence in support of this notion is still scarce. This review summarizes and discusses the new evidence on the pathophysiological role of mTOR signaling in the cardiovascular system.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Maurizio Forte
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Giacomo Frati
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Junichi Sadoshima
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.).
| |
Collapse
|
145
|
Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette FA, Topisirovic I, Hulea L. mTOR as a central regulator of lifespan and aging. F1000Res 2019; 8:F1000 Faculty Rev-998. [PMID: 31316753 PMCID: PMC6611156 DOI: 10.12688/f1000research.17196.1] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
The mammalian/mechanistic target of rapamycin (mTOR) is a key component of cellular metabolism that integrates nutrient sensing with cellular processes that fuel cell growth and proliferation. Although the involvement of the mTOR pathway in regulating life span and aging has been studied extensively in the last decade, the underpinning mechanisms remain elusive. In this review, we highlight the emerging insights that link mTOR to various processes related to aging, such as nutrient sensing, maintenance of proteostasis, autophagy, mitochondrial dysfunction, cellular senescence, and decline in stem cell function.
Collapse
Affiliation(s)
- David Papadopoli
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Karine Boulay
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Lawrence Kazak
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
| | - Michael Pollak
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Frédérick A. Mallette
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Ivan Topisirovic
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Laura Hulea
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| |
Collapse
|
146
|
Li R, Huang Y, Semple I, Kim M, Zhang Z, Lee JH. Cardioprotective roles of sestrin 1 and sestrin 2 against doxorubicin cardiotoxicity. Am J Physiol Heart Circ Physiol 2019; 317:H39-H48. [PMID: 31026186 PMCID: PMC6692737 DOI: 10.1152/ajpheart.00008.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/29/2019] [Accepted: 04/16/2019] [Indexed: 01/31/2023]
Abstract
Doxorubicin is a chemotherapy medication widely used to treat a variety of cancers. Even though it offers one of the most effective anti-cancer treatments, its clinical use is limited because of its strong cardiotoxicity that can lead to fatal conditions. Here, we show that sestrin 1 and sestrin 2, members of the sestrin family of proteins that are stress-inducible regulators of metabolism, are critical for suppressing doxorubicin cardiotoxicity and coordinating the AMPK-mammalian target of rapamycin complex 1 (mTORC1) autophagy signaling network for cardioprotection. Expression of both sestrin 1 and sestrin 2 was highly increased in the mouse heart after doxorubicin injection. Genetic ablation of sestrin 1 and sestrin 2 rendered mice more vulnerable to doxorubicin and exacerbated doxorubicin-induced cardiac pathologies including cardiomyocyte apoptosis and cardiac dysfunction. These pathologies were associated with strong dysregulation of the cardiac signaling network, including suppression of the AMPK pathway and activation of the mTORC1 pathway. Consistent with AMPK downregulation and mTORC1 upregulation, autophagic activity of heart tissue was diminished, leading to prominent accumulation of autophagy substrate, p62/SQSTM1. Taken together, our results indicate that sestrin 1 and sestrin 2 are important cardioprotective proteins that coordinate metabolic signaling pathways and autophagy to minimize cardiac damage in response to doxorubicin insult. Augmenting this protective mechanism could provide a novel therapeutic rationale for prevention and treatment of doxorubicin cardiotoxicity. NEW & NOTEWORTHY Doxorubicin is a highly efficient chemotherapeutic medicine; however, its use is limited because of its strong cardiotoxicity. Here, we show that sestrin 1 and sestrin 2 are critical protectors of cardiomyocytes from doxorubicin damage. By upregulating AMPK and autophagic activities and suppressing mammalian target of rapamycin complex 1 and oxidative stress, sestrins counteract detrimental effects of doxorubicin on cardiomyocytes. Correspondingly, loss of sestrin 1 and sestrin 2 produced remarkable dysregulation of these pathways, leading to prominent cardiac cell death and deterioration of heart function.
Collapse
Affiliation(s)
- Ruiting Li
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education , Nanjing , China
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education , Nanjing , China
| | - Ian Semple
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Myungjin Kim
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education , Nanjing , China
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
147
|
Inpanathan S, Botelho RJ. The Lysosome Signaling Platform: Adapting With the Times. Front Cell Dev Biol 2019; 7:113. [PMID: 31281815 PMCID: PMC6595708 DOI: 10.3389/fcell.2019.00113] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are the terminal degradative compartment of autophagy, endocytosis and phagocytosis. What once was viewed as a simple acidic organelle in charge of macromolecular digestion has emerged as a dynamic organelle capable of integrating cellular signals and producing signal outputs. In this review, we focus on the concept that the lysosome surface serves as a platform to assemble major signaling hubs like mTORC1, AMPK, GSK3 and the inflammasome. These molecular assemblies integrate and facilitate cross-talk between signals such as amino acid and energy levels, membrane damage and infection, and ultimately enable responses such as autophagy, cell growth, membrane repair and microbe clearance. In particular, we review how molecular machinery like the vacuolar-ATPase proton pump, sestrins, the GATOR complexes, and the Ragulator, modulate mTORC1, AMPK, GSK3 and inflammation. We then elaborate how these signals control autophagy initiation and resolution, TFEB-mediated lysosome adaptation, lysosome remodeling, antigen presentation, inflammation, membrane damage repair and clearance. Overall, by being at the cross-roads for several membrane pathways, lysosomes have emerged as the ideal surveillance compartment to sense, integrate and elicit cellular behavior and adaptation in response to changing environmental and cellular conditions.
Collapse
Affiliation(s)
- Subothan Inpanathan
- Department of Chemistry and Biology, Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology, Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| |
Collapse
|
148
|
Cordani M, Sánchez-Álvarez M, Strippoli R, Bazhin AV, Donadelli M. Sestrins at the Interface of ROS Control and Autophagy Regulation in Health and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1283075. [PMID: 31205582 PMCID: PMC6530209 DOI: 10.1155/2019/1283075] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/14/2019] [Indexed: 12/30/2022]
Abstract
Reactive oxygen species (ROS) and autophagy are two highly complex and interrelated components of cell physiopathology, but our understanding of their integration and their contribution to cell homeostasis and disease is still limited. Sestrins (SESNs) belong to a family of highly conserved stress-inducible proteins that orchestrate antioxidant and autophagy-regulating functions protecting cells from various noxious stimuli, including DNA damage, oxidative stress, hypoxia, and metabolic stress. They are also relevant modulators of metabolism as positive regulators of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibitors of mammalian target of rapamycin complex 1 (mTORC1). Since perturbations in these pathways are central to multiple disorders, SESNs might constitute potential novel therapeutic targets of broad interest. In this review, we discuss the current understanding of regulatory and effector networks of SESNs, highlighting their significance as potential biomarkers and therapeutic targets for different diseases, such as aging-related diseases, metabolic disorders, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), CNB-CSIC-IMDEA Nanociencia Associated Unit “Unidad de Nanobiotecnología”, Madrid 28049, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Raffaele Strippoli
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.C.S., Rome, Italy
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplantation Surgery, Ludwig Maximilian University, Munich, Germany
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
149
|
Luo C, Zheng N, Zhao S, Wang J. Sestrin2 Negatively Regulates Casein Synthesis through the SH3BP4-mTORC1 Pathway in Response to AA Depletion or Supplementation in Cow Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:4849-4859. [PMID: 30969118 DOI: 10.1021/acs.jafc.9b00716] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sestrin2 (SESN2) negatively regulates the mammalian target of rapamycin complex 1 (mTORC1) pathway and casein synthesis in response to amino acid (AA) depletion in cow mammary epithelial cells (CMECs); however, the underlying mechanism is unclear. In the current study, the regulation of SESN2 on AA-mediated β-casein (CSN2) synthesis in CMECs and its mechanism were investigated. Overexpression and silencing of SESN2 demonstrated that SESN2 negatively regulated AA-mediated expression of CSN2 and mTORC1 pathway. Co-immunoprecipitation analysis showed that SESN2 interacted with SH3 domain-binding protein 4 (SH3BP4). Overexpression and silencing of SH3BP4 demonstrated that SH3BP4 negatively regulated AA-mediated expression of CSN2 and mTORC1 pathway and that SESN2 negatively regulated expression of CSN2 and mTORC1 pathway through the SH3BP4 in the presence and absence of AA. The absence or presence of AA demonstrated that AA negatively regulated expression and nuclear localization of activating transcription factor 4 (ATF4). Overexpression and silencing of ATF4 demonstrated that AA negatively regulated SESN2 expression through ATF4. Together, these results indicate that SESN2 negatively regulates the mTORC1 pathway and subsequent CSN2 synthesis through the SH3BP4 in response to AA absence or presence in CMECs.
Collapse
Affiliation(s)
- Chaochao Luo
- State Key Laboratory of Animal Nutrition , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs , Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing 100193 , P. R. China
| |
Collapse
|
150
|
Gan L, Seki A, Shen K, Iyer H, Han K, Hayer A, Wollman R, Ge X, Lin JR, Dey G, Talbot WS, Meyer T. The lysosomal GPCR-like protein GPR137B regulates Rag and mTORC1 localization and activity. Nat Cell Biol 2019; 21:614-626. [PMID: 31036939 PMCID: PMC6649673 DOI: 10.1038/s41556-019-0321-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/27/2019] [Indexed: 12/13/2022]
Abstract
Cell growth is controlled by a lysosomal signaling complex containing Rag small GTPases and mTORC1 kinase. Here we carried out a microscopy-based genome-wide human siRNA screen and discovered a lysosome-localized G-protein coupled receptor (GPCR)-like protein, GPR137B, that interacts with Rag GTPases, increases Rag localization and activity, and thereby regulates mTORC1 translocation and activity. High GPR137B expression can recruit and activate mTORC1 in the absence of amino acids. Furthermore, GPR137B also regulates the dissociation of activated Rag from lysosomes, suggesting that GPR137B controls a cycle of Rag activation and dissociation from lysosomes. GPR137B knockout cells exhibited defective autophagy and an expanded lysosome compartment, similar to Rag knockout cells. Like zebrafish RagA mutants, GPR137B mutant zebrafish had upregulated TFEB target gene expression and an expanded lysosome compartment in microglia. Thus, GPR137B is a GPCR-like lysosomal regulatory protein that controls dynamic Rag and mTORC1 localization and activity as well as lysosome morphology.
Collapse
Affiliation(s)
- Lin Gan
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Akiko Seki
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Kimberle Shen
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Harini Iyer
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Kyuho Han
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Roy Wollman
- Department of Integrative Biology and Physiology and Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Xuecai Ge
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Jerry R Lin
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Gautam Dey
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|