51
|
Rodríguez-Saavedra C, Morgado-Martínez LE, Burgos-Palacios A, King-Díaz B, López-Coria M, Sánchez-Nieto S. Moonlighting Proteins: The Case of the Hexokinases. Front Mol Biosci 2021; 8:701975. [PMID: 34235183 PMCID: PMC8256278 DOI: 10.3389/fmolb.2021.701975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Moonlighting proteins are defined as proteins with two or more functions that are unrelated and independent to each other, so that inactivation of one of them should not affect the second one and vice versa. Intriguingly, all the glycolytic enzymes are described as moonlighting proteins in some organisms. Hexokinase (HXK) is a critical enzyme in the glycolytic pathway and displays a wide range of functions in different organisms such as fungi, parasites, mammals, and plants. This review discusses HXKs moonlighting functions in depth since they have a profound impact on the responses to nutritional, environmental, and disease challenges. HXKs’ activities can be as diverse as performing metabolic activities, as a gene repressor complexing with other proteins, as protein kinase, as immune receptor and regulating processes like autophagy, programmed cell death or immune system responses. However, most of those functions are particular for some organisms while the most common moonlighting HXK function in several kingdoms is being a glucose sensor. In this review, we also analyze how different regulation mechanisms cause HXK to change its subcellular localization, oligomeric or conformational state, the response to substrate and product concentration, and its interactions with membrane, proteins, or RNA, all of which might impact the HXK moonlighting functions.
Collapse
Affiliation(s)
- Carolina Rodríguez-Saavedra
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Enrique Morgado-Martínez
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Andrés Burgos-Palacios
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Beatriz King-Díaz
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Montserrat López-Coria
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sobeida Sánchez-Nieto
- Laboratorio de Transporte y Percepción de Azúcares en Plantas, Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
52
|
Zhang R, Cheung CY, Seo SU, Liu H, Pardeshi L, Wong KH, Chow LMC, Chau MP, Wang Y, Lee AR, Kwon WY, Chen S, Chan BKW, Wong K, Choy RKW, Ko BCB. RUVBL1/2 Complex Regulates Pro-Inflammatory Responses in Macrophages via Regulating Histone H3K4 Trimethylation. Front Immunol 2021; 12:679184. [PMID: 34276666 PMCID: PMC8282052 DOI: 10.3389/fimmu.2021.679184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages play an important role in the host defense mechanism. In response to infection, macrophages activate a genetic program of pro-inflammatory response to kill any invading pathogen, and initiate an adaptive immune response. We have identified RUVBL2 - an ATP-binding protein belonging to the AAA+ (ATPase associated with diverse cellular activities) superfamily of ATPases - as a novel regulator in pro-inflammatory response of macrophages. Gene knockdown of Ruvbl2, or pharmacological inhibition of RUVBL1/2 activity, compromises type-2 nitric oxide synthase (Nos2) gene expression, nitric oxide production and anti-bacterial activity of mouse macrophages in response to lipopolysaccharides (LPS). RUVBL1/2 inhibitor similarly inhibits pro-inflammatory response in human monocytes, suggesting functional conservation of RUVBL1/2 in humans. Transcriptome analysis further revealed that major LPS-induced pro-inflammatory pathways in macrophages are regulated in a RUVBL1/2-dependent manner. Furthermore, RUVBL1/2 inhibition significantly reduced the level of histone H3K4me3 at the promoter region of Nos2 and Il6, two prototypical pro-inflammatory genes, and diminished the recruitment of NF-kappaB to the corresponding enhancers. Our study reveals RUVBL1/2 as an integral component of macrophage pro-inflammatory responses through epigenetic regulations, and the therapeutic potentials of RUVBL1/2 inhibitors in the treatment of diseases caused by aberrant activation of pro-inflammatory pathways.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| | - Chris Y Cheung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| | - Sang-Uk Seo
- Department of Microbiology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hang Liu
- The University Research Facility in Chemical and Environmental Analysis, The Hong Kong Polytechnic University, Hong Kong, China
| | - Lakhansing Pardeshi
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.,Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Koon Ho Wong
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.,Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Larry M C Chow
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| | - Mary P Chau
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yixiang Wang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ah Ra Lee
- Department of Microbiology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Woon Yong Kwon
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Sheng Chen
- Department of Infectious Diseases and Public Health, The City University of Hong Kong, Hong Kong, China
| | - Bill Kwan-Wai Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kenneth Wong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Richard K W Choy
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben C B Ko
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
53
|
Pacheco JM, Canal MV, Pereyra CM, Welchen E, Martínez-Noël GMA, Estevez JM. The tip of the iceberg: emerging roles of TORC1, and its regulatory functions in plant cells. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:4085-4101. [PMID: 33462577 DOI: 10.1093/jxb/eraa603] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Target of Rapamycin (TOR) is an evolutionarily conserved protein kinase that plays a central role in coordinating cell growth with light availability, the diurnal cycle, energy availability, and hormonal pathways. TOR Complex 1 (TORC1) controls cell proliferation, growth, metabolism, and defense in plants. Sugar availability is the main signal for activation of TOR in plants, as it also is in mammals and yeast. Specific regulators of the TOR kinase pathway in plants are inorganic compounds in the form of major nutrients in the soils, and light inputs via their impact on autotrophic metabolism. The lack of TOR is embryo-lethal in plants, whilst dysregulation of TOR signaling causes major alterations in growth and development. TOR exerts control as a regulator of protein translation via the action of proteins such as S6K, RPS6, and TAP46. Phytohormones are central players in the downstream systemic physiological TOR effects. TOR has recently been attributed to have roles in the control of DNA methylation, in the abundance of mRNA splicing variants, and in the variety of regulatory lncRNAs and miRNAs. In this review, we summarize recent discoveries in the plant TOR signaling pathway in the context of our current knowledge of mammalian and yeast cells, and highlight the most important gaps in our understanding of plants that need to be addressed in the future.
Collapse
Affiliation(s)
| | - María Victoria Canal
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas,, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Cintia M Pereyra
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC-CONICET) and Fundación para Investigaciones Biológicas Aplicadas (FIBA), Vieytes, Mar Del Plata, Argentina
| | - Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas,, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Giselle M A Martínez-Noël
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC-CONICET) and Fundación para Investigaciones Biológicas Aplicadas (FIBA), Vieytes, Mar Del Plata, Argentina
| | - José M Estevez
- Fundación Instituto Leloir and IIBBA-CONICET, Buenos Aires CP, Argentina
- Centro de Biotecnología Vegetal (CBV), Facultad de Ciencias de la Vida (FCsV), Universidad Andres Bello, Santiago, Chile and Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| |
Collapse
|
54
|
Romero-Pozuelo J, Figlia G, Kaya O, Martin-Villalba A, Teleman AA. Cdk4 and Cdk6 Couple the Cell-Cycle Machinery to Cell Growth via mTORC1. Cell Rep 2021; 31:107504. [PMID: 32294430 DOI: 10.1016/j.celrep.2020.03.068] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 12/20/2022] Open
Abstract
Cell growth is coupled to cell-cycle progression in mitotically proliferating mammalian cells, but the underlying molecular mechanisms are not well understood. CyclinD-Cdk4/6 is known to phosphorylate RB to promote S-phase entry, but recent work suggests they have additional functions. We show here that CyclinD-Cdk4/6 activates mTORC1 by binding and phosphorylating TSC2 on Ser1217 and Ser1452. Pharmacological inhibition of Cdk4/6 leads to a rapid, TSC2-dependent reduction of mTORC1 activity in multiple human and mouse cell lines, including breast cancer cells. By simultaneously driving mTORC1 and E2F, CyclinD-Cdk4/6 couples cell growth to cell-cycle progression. Consistent with this, we see that mTORC1 activity is cell cycle dependent in proliferating neural stem cells of the adult rodent brain. We find that Cdk4/6 inhibition reduces cell proliferation partly via TSC2 and mTORC1. This is of clinical relevance, because Cdk4/6 inhibitors are used for breast cancer therapy.
Collapse
Affiliation(s)
- Jesús Romero-Pozuelo
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Gianluca Figlia
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Oguzhan Kaya
- Heidelberg University, 69120 Heidelberg, Germany; Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana Martin-Villalba
- Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
55
|
Nakamura A, Kakihara Y, Funayama A, Haga K, Mikami T, Kobayashi D, Yoshida Y, Izumi K, Kobayashi T, Saeki M. HEATR1, a novel interactor of Pontin/Reptin, stabilizes Pontin/Reptin and promotes cell proliferation of oral squamous cell carcinoma. Biochem Biophys Res Commun 2021; 557:294-301. [PMID: 33894417 DOI: 10.1016/j.bbrc.2021.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022]
Abstract
Pontin and Reptin are closely related proteins belonging to the AAA+ (ATPases Associated with various cellular Activities) family. They form a hetero-oligomeric complex, Pontin/Reptin, which is involved in protein stability and assembly of the protein complexes as a molecular chaperone. Overexpression of Pontin and Reptin in tumor cells has been reported and is implicated in the development of various cancers. However, the molecular mechanism of Pontin/Reptin function in oral squamous cell carcinoma (OSCC) development remains unclear. Here, we identify HEAT repeat-containing protein 1 (HEATR1) as a novel binding factor of Pontin/Reptin. Functionally, HEATR1 stabilizes Pontin/Reptin and positively regulates OSCC cell proliferation by activating mTOR and pre-rRNA synthesis. We also find that HEATR1 expression is markedly upregulated in tumor region of OSCC tissue. Hence, we propose that HEATR1 is involved in the regulation of mTOR and ribosome biogenesis as a potential protein stabilizer of Pontin/Reptin in OSCC.
Collapse
Affiliation(s)
- Akihiko Nakamura
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan; Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Yoshito Kakihara
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan.
| | - Akinori Funayama
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Kenta Haga
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Toshihiko Mikami
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Daiki Kobayashi
- Omics Unit, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510, Japan
| | - Yutaka Yoshida
- Department of Structural Pathology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510, Japan
| | - Kenji Izumi
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Tadaharu Kobayashi
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Makio Saeki
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
56
|
Ogrodnik M. Cellular aging beyond cellular senescence: Markers of senescence prior to cell cycle arrest in vitro and in vivo. Aging Cell 2021; 20:e13338. [PMID: 33711211 PMCID: PMC8045927 DOI: 10.1111/acel.13338] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 12/14/2022] Open
Abstract
The field of research on cellular senescence experienced a rapid expansion from being primarily focused on in vitro aspects of aging to the vast territories of animal and clinical research. Cellular senescence is defined by a set of markers, many of which are present and accumulate in a gradual manner prior to senescence induction or are found outside of the context of cellular senescence. These markers are now used to measure the impact of cellular senescence on aging and disease as well as outcomes of anti-senescence interventions, many of which are at the stage of clinical trials. It is thus of primary importance to discuss their specificity as well as their role in the establishment of senescence. Here, the presence and role of senescence markers are described in cells prior to cell cycle arrest, especially in the context of replicative aging and in vivo conditions. Specifically, this review article seeks to describe the process of "cellular aging": the progression of internal changes occurring in primary cells leading to the induction of cellular senescence and culminating in cell death. Phenotypic changes associated with aging prior to senescence induction will be characterized, as well as their effect on the induction of cell senescence and the final fate of cells reviewed. Using published datasets on assessments of senescence markers in vivo, it will be described how disparities between quantifications can be explained by the concept of cellular aging. Finally, throughout the article the applicational value of broadening cellular senescence paradigm will be discussed.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds Vienna Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center Vienna Austria
- Austrian Cluster for Tissue Regeneration Vienna Austria
| |
Collapse
|
57
|
Abstract
Cells metabolize nutrients for biosynthetic and bioenergetic needs to fuel growth and proliferation. The uptake of nutrients from the environment and their intracellular metabolism is a highly controlled process that involves cross talk between growth signaling and metabolic pathways. Despite constant fluctuations in nutrient availability and environmental signals, normal cells restore metabolic homeostasis to maintain cellular functions and prevent disease. A central signaling molecule that integrates growth with metabolism is the mechanistic target of rapamycin (mTOR). mTOR is a protein kinase that responds to levels of nutrients and growth signals. mTOR forms two protein complexes, mTORC1, which is sensitive to rapamycin, and mTORC2, which is not directly inhibited by this drug. Rapamycin has facilitated the discovery of the various functions of mTORC1 in metabolism. Genetic models that disrupt either mTORC1 or mTORC2 have expanded our knowledge of their cellular, tissue, as well as systemic functions in metabolism. Nevertheless, our knowledge of the regulation and functions of mTORC2, particularly in metabolism, has lagged behind. Since mTOR is an important target for cancer, aging, and other metabolism-related pathologies, understanding the distinct and overlapping regulation and functions of the two mTOR complexes is vital for the development of more effective therapeutic strategies. This review discusses the key discoveries and recent findings on the regulation and metabolic functions of the mTOR complexes. We highlight findings from cancer models but also discuss other examples of the mTOR-mediated metabolic reprogramming occurring in stem and immune cells, type 2 diabetes/obesity, neurodegenerative disorders, and aging.
Collapse
Affiliation(s)
- Angelia Szwed
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Eugene Kim
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
58
|
Vuononvirta J, Marelli-Berg FM, Poobalasingam T. Metabolic regulation of T lymphocyte motility and migration. Mol Aspects Med 2021; 77:100888. [PMID: 32814624 DOI: 10.1016/j.mam.2020.100888] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/25/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
In order to fulfill their effector and patrolling functions, lymphocytes traffic through the body and need to adapt to different tissue microenvironments. First, mature lymphocytes egress the bone marrow and the thymus into the vascular system. Circulating lymphocytes can exit the vasculature and penetrate into the tissues, either for patrolling in search for pathogens or to eliminate infection and activate the adaptive immune response. The cytoskeletal reorganization necessary to sustain migration require high levels of energy thus presenting a substantial bioenergetic challenge to migrating cells. The metabolic regulation of lymphocyte motility and trafficking has only recently begun to be investigated. In this review we will summarize current knowledge of the crosstalk between cell metabolism and the cytoskeleton in T lymphocytes, and discuss the concept that lymphocyte metabolism may reprogram in response to migratory stimuli and adapt to the different environmental cues received during recirculation in tissues.
Collapse
Affiliation(s)
- Juho Vuononvirta
- William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | | |
Collapse
|
59
|
Yang G, Xia Y, Ren W. Glutamine metabolism in Th17/Treg cell fate: applications in Th17 cell-associated diseases. SCIENCE CHINA. LIFE SCIENCES 2021; 64:221-233. [PMID: 32671630 DOI: 10.1007/s11427-020-1703-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Alteration in the Th17/Treg cell balance is implicated in various autoimmune diseases and these disease-associated pathologies. Increasing investigations have shown that glutamine metabolism regulates the differentiation of Th17 and Treg cells. Here we summarize the mechanisms by which glutamine metabolism regulates Th17/Treg cell fate. Some examples of a glutamine metabolism-dependent modulation of the development and progression of several Th17 Treg cell-associated diseases are provided afterward. This review will provide a comprehensive understanding of the importance of glutamine metabolism in the fate of Th17 Treg cell differentiation.
Collapse
Affiliation(s)
- Guan Yang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
60
|
Condon KJ, Orozco JM, Adelmann CH, Spinelli JB, van der Helm PW, Roberts JM, Kunchok T, Sabatini DM. Genome-wide CRISPR screens reveal multitiered mechanisms through which mTORC1 senses mitochondrial dysfunction. Proc Natl Acad Sci U S A 2021; 118:e2022120118. [PMID: 33483422 PMCID: PMC7848693 DOI: 10.1073/pnas.2022120118] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In mammalian cells, nutrients and growth factors signal through an array of upstream proteins to regulate the mTORC1 growth control pathway. Because the full complement of these proteins has not been systematically identified, we developed a FACS-based CRISPR-Cas9 genetic screening strategy to pinpoint genes that regulate mTORC1 activity. Along with almost all known positive components of the mTORC1 pathway, we identified many genes that impact mTORC1 activity, including DCAF7, CSNK2B, SRSF2, IRS4, CCDC43, and HSD17B10 Using the genome-wide screening data, we generated a focused sublibrary containing single guide RNAs (sgRNAs) targeting hundreds of genes and carried out epistasis screens in cells lacking nutrient- and stress-responsive mTORC1 modulators, including GATOR1, AMPK, GCN2, and ATF4. From these data, we pinpointed mitochondrial function as a particularly important input into mTORC1 signaling. While it is well appreciated that mitochondria signal to mTORC1, the mechanisms are not completely clear. We find that the kinases AMPK and HRI signal, with varying kinetics, mitochondrial distress to mTORC1, and that HRI acts through the ATF4-dependent up-regulation of both Sestrin2 and Redd1. Loss of both AMPK and HRI is sufficient to render mTORC1 signaling largely resistant to mitochondrial dysfunction induced by the ATP synthase inhibitor oligomycin as well as the electron transport chain inhibitors piericidin and antimycin. Taken together, our data reveal a catalog of genes that impact the mTORC1 pathway and clarify the multifaceted ways in which mTORC1 senses mitochondrial dysfunction.
Collapse
Affiliation(s)
- Kendall J Condon
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Jose M Orozco
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Charles H Adelmann
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Jessica B Spinelli
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Pim W van der Helm
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Justin M Roberts
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142;
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
61
|
Azim MF, Burch-Smith TM. Organelles-nucleus-plasmodesmata signaling (ONPS): an update on its roles in plant physiology, metabolism and stress responses. CURRENT OPINION IN PLANT BIOLOGY 2020; 58:48-59. [PMID: 33197746 DOI: 10.1016/j.pbi.2020.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/26/2020] [Accepted: 09/27/2020] [Indexed: 05/03/2023]
Abstract
Plasmodesmata allow movement of metabolites and signaling molecules between plant cells and are, therefore, critical players in plant development and physiology, and in responding to environmental signals and stresses. There is emerging evidence that plasmodesmata are controlled by signaling originating from other organelles, primarily the chloroplasts and mitochondria. These signals act in the nucleus to alter expression of genetic pathways that control both trafficking via plasmodesmata and the plasmodesmatal pores themselves. This control circuit was dubbed organelle-nucleus-plasmodesmata signaling (ONPS). Here we discuss how ONPS arose during plant evolution and highlight the discovery of an ONPS-like module for regulating stomata. We also consider recent findings that illuminate details of the ONPS circuit and its roles in plant physiology, metabolism, and defense.
Collapse
Affiliation(s)
- Mohammad F Azim
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, United States
| | - Tessa M Burch-Smith
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, United States.
| |
Collapse
|
62
|
Unraveling the multifaceted nature of the nuclear function of mTOR. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118907. [PMID: 33189783 DOI: 10.1016/j.bbamcr.2020.118907] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/02/2020] [Accepted: 11/08/2020] [Indexed: 01/25/2023]
Abstract
Positioned at the axis between the cell and its environment, mTOR directs a wide range of cellular activity in response to nutrients, growth factors, and stress. Our understanding of the role of mTOR is evolving beyond the spatial confines of the cytosol, and its role in the nucleus becoming ever more apparent. In this review, we will address various studies that explore the role of nuclear mTOR (nmTOR) in specific cellular programs and how these pathways influence one another. To understand the emerging roles of nuclear mTOR, we discuss data and propose plausible mechanisms to offer novel ideas, hypotheses, and future research directions.
Collapse
|
63
|
Mugume Y, Kazibwe Z, Bassham DC. Target of Rapamycin in Control of Autophagy: Puppet Master and Signal Integrator. Int J Mol Sci 2020; 21:ijms21218259. [PMID: 33158137 PMCID: PMC7672647 DOI: 10.3390/ijms21218259] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
The target of rapamycin (TOR) is an evolutionarily-conserved serine/threonine kinase that senses and integrates signals from the environment to coordinate developmental and metabolic processes. TOR senses nutrients, hormones, metabolites, and stress signals to promote cell and organ growth when conditions are favorable. However, TOR is inhibited when conditions are unfavorable, promoting catabolic processes such as autophagy. Autophagy is a macromolecular degradation pathway by which cells degrade and recycle cytoplasmic materials. TOR negatively regulates autophagy through phosphorylation of ATG13, preventing activation of the autophagy-initiating ATG1-ATG13 kinase complex. Here we review TOR complex composition and function in photosynthetic and non-photosynthetic organisms. We also review recent developments in the identification of upstream TOR activators and downstream effectors of TOR. Finally, we discuss recent developments in our understanding of the regulation of autophagy by TOR in photosynthetic organisms.
Collapse
|
64
|
Ingargiola C, Turqueto Duarte G, Robaglia C, Leprince AS, Meyer C. The Plant Target of Rapamycin: A Conduc TOR of Nutrition and Metabolism in Photosynthetic Organisms. Genes (Basel) 2020; 11:genes11111285. [PMID: 33138108 PMCID: PMC7694126 DOI: 10.3390/genes11111285] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
Living organisms possess many mechanisms to sense nutrients and favorable conditions, which allow them to grow and develop. Photosynthetic organisms are very diverse, from green unicellular algae to multicellular flowering plants, but most of them are sessile and thus unable to escape from the biotic and abiotic stresses they experience. The Target of Rapamycin (TOR) signaling pathway is conserved in all eukaryotes and acts as a central regulatory hub between growth and extrinsic factors, such as nutrients or stress. However, relatively little is known about the regulations and roles of this pathway in plants and algae. Although some features of the TOR pathway seem to have been highly conserved throughout evolution, others clearly differ in plants, perhaps reflecting adaptations to different lifestyles and the rewiring of this primordial signaling module to adapt to specific requirements. Indeed, TOR is involved in plant responses to a vast array of signals including nutrients, hormones, light, stresses or pathogens. In this review, we will summarize recent studies that address the regulations of TOR by nutrients in photosynthetic organisms, and the roles of TOR in controlling important metabolic pathways, highlighting similarities and differences with the other eukaryotes.
Collapse
Affiliation(s)
- Camille Ingargiola
- Institut Jean-Pierre Bourgin (IJPB), INRAE, AgroParisTech, Université Paris-Saclay, 78000 Versailles, France; (C.I.); (G.T.D.); (A.-S.L.)
| | - Gustavo Turqueto Duarte
- Institut Jean-Pierre Bourgin (IJPB), INRAE, AgroParisTech, Université Paris-Saclay, 78000 Versailles, France; (C.I.); (G.T.D.); (A.-S.L.)
- Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam, Germany
| | - Christophe Robaglia
- Laboratoire de Génétique et Biophysique des Plantes, Faculté des Sciences de Luminy, UMR 7265, CEA, CNRS, BIAM, Aix Marseille Université, 13009 Marseille, France;
| | - Anne-Sophie Leprince
- Institut Jean-Pierre Bourgin (IJPB), INRAE, AgroParisTech, Université Paris-Saclay, 78000 Versailles, France; (C.I.); (G.T.D.); (A.-S.L.)
- Faculté des Sciences et d’Ingénierie, Sorbonne Université, UFR 927, 4 Place Jussieu, 75252 Paris, France
| | - Christian Meyer
- Institut Jean-Pierre Bourgin (IJPB), INRAE, AgroParisTech, Université Paris-Saclay, 78000 Versailles, France; (C.I.); (G.T.D.); (A.-S.L.)
- Correspondence:
| |
Collapse
|
65
|
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
|
66
|
Melick CH, Jewell JL. Regulation of mTORC1 by Upstream Stimuli. Genes (Basel) 2020; 11:genes11090989. [PMID: 32854217 PMCID: PMC7565831 DOI: 10.3390/genes11090989] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 01/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is an evolutionary conserved Ser/Thr protein kinase that senses multiple upstream stimuli to control cell growth, metabolism, and autophagy. mTOR is the catalytic subunit of mTOR complex 1 (mTORC1). A significant amount of research has uncovered the signaling pathways regulated by mTORC1, and the involvement of these signaling cascades in human diseases like cancer, diabetes, and ageing. Here, we review advances in mTORC1 regulation by upstream stimuli. We specifically focus on how growth factors, amino acids, G-protein coupled receptors (GPCRs), phosphorylation, and small GTPases regulate mTORC1 activity and signaling.
Collapse
Affiliation(s)
- Chase H. Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenna L. Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence:
| |
Collapse
|
67
|
TMBIM6/BI-1 contributes to cancer progression through assembly with mTORC2 and AKT activation. Nat Commun 2020; 11:4012. [PMID: 32782388 PMCID: PMC7419509 DOI: 10.1038/s41467-020-17802-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 07/16/2020] [Indexed: 01/09/2023] Open
Abstract
Transmembrane B cell lymphoma 2-associated X protein inhibitor motif-containing (TMBIM) 6, a Ca2+ channel-like protein, is highly up-regulated in several cancer types. Here, we show that TMBIM6 is closely associated with survival in patients with cervical, breast, lung, and prostate cancer. TMBIM6 deletion or knockdown suppresses primary tumor growth. Further, mTORC2 activation is up-regulated by TMBIM6 and stimulates glycolysis, protein synthesis, and the expression of lipid synthesis genes and glycosylated proteins. Moreover, ER-leaky Ca2+ from TMBIM6, a unique characteristic, is shown to affect mTORC2 assembly and its association with ribosomes. In addition, we identify that the BIA compound, a potentialTMBIM6 antagonist, prevents TMBIM6 binding to mTORC2, decreases mTORC2 activity, and also regulates TMBIM6-leaky Ca2+, further suppressing tumor formation and progression in cancer xenograft models. This previously unknown signaling cascade in which mTORC2 activity is enhanced via the interaction with TMBIM6 provides potential therapeutic targets for various malignancies. TMBIM6, a member of the transmembrane BI-1 motif-containing family of proteins, is overexpressed in many cancer types. Here, the authors show that TMBIM6 regulates AKT activation through mTORC2 assembly and ribosome association and identify an antagonist of TMBIM6 with anti-tumor properties.
Collapse
|
68
|
Tafur L, Kefauver J, Loewith R. Structural Insights into TOR Signaling. Genes (Basel) 2020; 11:E885. [PMID: 32759652 PMCID: PMC7464330 DOI: 10.3390/genes11080885] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022] Open
Abstract
The Target of Rapamycin (TOR) is a highly conserved serine/threonine protein kinase that performs essential roles in the control of cellular growth and metabolism. TOR acts in two distinct multiprotein complexes, TORC1 and TORC2 (mTORC1 and mTORC2 in humans), which maintain different aspects of cellular homeostasis and orchestrate the cellular responses to diverse environmental challenges. Interest in understanding TOR signaling is further motivated by observations that link aberrant TOR signaling to a variety of diseases, ranging from epilepsy to cancer. In the last few years, driven in large part by recent advances in cryo-electron microscopy, there has been an explosion of available structures of (m)TORC1 and its regulators, as well as several (m)TORC2 structures, derived from both yeast and mammals. In this review, we highlight and summarize the main findings from these reports and discuss both the fascinating and unexpected molecular biology revealed and how this knowledge will potentially contribute to new therapeutic strategies to manipulate signaling through these clinically relevant pathways.
Collapse
Affiliation(s)
- Lucas Tafur
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
| | - Jennifer Kefauver
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
| | - Robbie Loewith
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
- Swiss National Centre for Competence in Research (NCCR) in Chemical Biology, University of Geneva, Sciences II, Room 3-308, 30 Quai Ernest-Ansermet, CH1211 Geneva, Switzerland
| |
Collapse
|
69
|
Brunkard JO. Exaptive Evolution of Target of Rapamycin Signaling in Multicellular Eukaryotes. Dev Cell 2020; 54:142-155. [PMID: 32649861 PMCID: PMC7346820 DOI: 10.1016/j.devcel.2020.06.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022]
Abstract
Target of rapamycin (TOR) is a protein kinase that coordinates metabolism with nutrient and energy availability in eukaryotes. TOR and its primary interactors, RAPTOR and LST8, have been remarkably evolutionarily static since they arose in the unicellular last common ancestor of plants, fungi, and animals, but the upstream regulatory mechanisms and downstream effectors of TOR signaling have evolved considerable diversity in these separate lineages. Here, I focus on the roles of exaptation and adaptation in the evolution of novel signaling axes in the TOR network in multicellular eukaryotes, concentrating especially on amino acid sensing, cell-cell signaling, and cell differentiation.
Collapse
Affiliation(s)
- Jacob O Brunkard
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Plant Gene Expression Center, U.S. Department of Agriculture Agricultural Research Service, Albany, CA 94710, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
70
|
Shin SH, Lee JS, Zhang JM, Choi S, Boskovic ZV, Zhao R, Song M, Wang R, Tian J, Lee MH, Kim JH, Jeong M, Lee JH, Petukhov M, Lee SW, Kim SG, Zou L, Byun S. Synthetic lethality by targeting the RUVBL1/2-TTT complex in mTORC1-hyperactive cancer cells. SCIENCE ADVANCES 2020; 6:eaay9131. [PMID: 32789167 PMCID: PMC7399646 DOI: 10.1126/sciadv.aay9131] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 03/19/2020] [Indexed: 05/08/2023]
Abstract
Despite considerable efforts, mTOR inhibitors have produced limited success in the clinic. To define the vulnerabilities of mTORC1-addicted cancer cells and to find previously unknown therapeutic targets, we investigated the mechanism of piperlongumine, a small molecule identified in a chemical library screen to specifically target cancer cells with a hyperactive mTORC1 phenotype. Sensitivity to piperlongumine was dependent on its ability to suppress RUVBL1/2-TTT, a complex involved in chromatin remodeling and DNA repair. Cancer cells with high mTORC1 activity are subjected to higher levels of DNA damage stress via c-Myc and displayed an increased dependency on RUVBL1/2 for survival and counteracting genotoxic stress. Examination of clinical cancer tissues also demonstrated that high mTORC1 activity was accompanied by high RUVBL2 expression. Our findings reveal a previously unknown role for RUVBL1/2 in cell survival, where it acts as a functional chaperone to mitigate stress levels induced in the mTORC1-Myc-DNA damage axis.
Collapse
Affiliation(s)
- Seung Ho Shin
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Department of Food and Nutrition, Gyeongsang National University, Jinju 52828, Republic of Korea
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ji Su Lee
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jia-Min Zhang
- Massachusetts General Hospital Cancer Center, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Sungbin Choi
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Zarko V. Boskovic
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Ran Zhao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Mengqiu Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Rui Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Jie Tian
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Mee-Hyun Lee
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Jae Hwan Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Minju Jeong
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Michael Petukhov
- Petersburg Nuclear Physics Institute named after B.P. Konstantinov, NRC "Kurchatov Institute", Gatchina, Russia
- Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Sam W. Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
- Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sang Gyun Kim
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, USA
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Building 149 13th Street, Charlestown, MA 02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Sanguine Byun
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
71
|
Milanesi R, Coccetti P, Tripodi F. The Regulatory Role of Key Metabolites in the Control of Cell Signaling. Biomolecules 2020; 10:biom10060862. [PMID: 32516886 PMCID: PMC7356591 DOI: 10.3390/biom10060862] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
Robust biological systems are able to adapt to internal and environmental perturbations. This is ensured by a thick crosstalk between metabolism and signal transduction pathways, through which cell cycle progression, cell metabolism and growth are coordinated. Although several reports describe the control of cell signaling on metabolism (mainly through transcriptional regulation and post-translational modifications), much fewer information is available on the role of metabolism in the regulation of signal transduction. Protein-metabolite interactions (PMIs) result in the modification of the protein activity due to a conformational change associated with the binding of a small molecule. An increasing amount of evidences highlight the role of metabolites of the central metabolism in the control of the activity of key signaling proteins in different eukaryotic systems. Here we review the known PMIs between primary metabolites and proteins, through which metabolism affects signal transduction pathways controlled by the conserved kinases Snf1/AMPK, Ras/PKA and TORC1. Interestingly, PMIs influence also the mitochondrial retrograde response (RTG) and calcium signaling, clearly demonstrating that the range of this phenomenon is not limited to signaling pathways related to metabolism.
Collapse
|
72
|
Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nat Commun 2020; 11:1559. [PMID: 32214088 PMCID: PMC7096448 DOI: 10.1038/s41467-020-15267-z] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Microglia are highly motile cells that continuously monitor the brain environment and respond to damage-associated cues. While glucose is the main energy substrate used by neurons in the brain, the nutrients metabolized by microglia to support surveillance of the parenchyma remain unexplored. Here, we use fluorescence lifetime imaging of intracellular NAD(P)H and time-lapse two-photon imaging of microglial dynamics in vivo and in situ, to show unique aspects of the microglial metabolic signature in the brain. Microglia are metabolically flexible and can rapidly adapt to consume glutamine as an alternative metabolic fuel in the absence of glucose. During insulin-induced hypoglycemia in vivo or in aglycemia in acute brain slices, glutaminolysis supports the maintenance of microglial process motility and damage-sensing functions. This metabolic shift sustains mitochondrial metabolism and requires mTOR-dependent signaling. This remarkable plasticity allows microglia to maintain their critical surveillance and phagocytic roles, even after brain neuroenergetic homeostasis is compromised. Glucose is the main source of fuel in the brain. Here, the authors show that in the absence of glucose, glutamine is required for microglia to maintain their immune surveillance function.
Collapse
|
73
|
Zachari M, Ktistakis NT. Mammalian Mitophagosome Formation: A Focus on the Early Signals and Steps. Front Cell Dev Biol 2020; 8:171. [PMID: 32258042 PMCID: PMC7093328 DOI: 10.3389/fcell.2020.00171] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/02/2020] [Indexed: 11/15/2022] Open
Abstract
Mitophagy, a conserved intracellular process by which mitochondria are eliminated via the autophagic machinery, is a quality control mechanism which facilitates maintenance of a functional mitochondrial network and cell homeostasis, making it a key process in development and longevity. Mitophagy has been linked to multiple human disorders, especially neurodegenerative diseases where the long-lived neurons are relying on clearance of old/damaged mitochondria to survive. During the past decade, the availability of novel tools to study mitophagy both in vitro and in vivo has significantly advanced our understanding of the molecular mechanisms governing this fundamental process in normal physiology and in various disease models. We here give an overview of the known mitophagy pathways and how they are induced, with a particular emphasis on the early events governing mitophagosome formation.
Collapse
Affiliation(s)
- Maria Zachari
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | | |
Collapse
|
74
|
Mutvei AP, Nagiec MJ, Hamann JC, Kim SG, Vincent CT, Blenis J. Rap1-GTPases control mTORC1 activity by coordinating lysosome organization with amino acid availability. Nat Commun 2020; 11:1416. [PMID: 32184389 PMCID: PMC7078236 DOI: 10.1038/s41467-020-15156-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/17/2020] [Indexed: 02/08/2023] Open
Abstract
The kinase mTOR complex 1 (mTORC1) promotes cellular growth and is frequently dysregulated in cancers. In response to nutrients, mTORC1 is activated on lysosomes by Rag and Rheb guanosine triphosphatases (GTPases) and drives biosynthetic processes. How limitations in nutrients suppress mTORC1 activity remains poorly understood. We find that when amino acids are limited, the Rap1-GTPases confine lysosomes to the perinuclear region and reduce lysosome abundance, which suppresses mTORC1 signaling. Rap1 activation, which is independent of known amino acid signaling factors, limits the lysosomal surface available for mTORC1 activation. Conversely, Rap1 depletion expands the lysosome population, which markedly increases association between mTORC1 and its lysosome-borne activators, leading to mTORC1 hyperactivity. Taken together, we establish Rap1 as a critical coordinator of the lysosomal system, and propose that aberrant changes in lysosomal surface availability can impact mTORC1 signaling output.
Collapse
Affiliation(s)
- Anders P Mutvei
- Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, Belfer Research Building, 413 E. 69th St., New York, NY, 10021, USA.
- Karolinska Institutet, Department of Microbiology, Tumor and Cell biology, Nobels väg 16, KI Solna Campus Karolinska Institutet, Box 280, SE-171 77, Stockholm, Sweden.
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, 751 85, Uppsala, Sweden.
| | - Michal J Nagiec
- Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, Belfer Research Building, 413 E. 69th St., New York, NY, 10021, USA
| | - Jens C Hamann
- Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, Belfer Research Building, 413 E. 69th St., New York, NY, 10021, USA
| | - Sang Gyun Kim
- Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, Belfer Research Building, 413 E. 69th St., New York, NY, 10021, USA
| | - C Theresa Vincent
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, 751 85, Uppsala, Sweden
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - John Blenis
- Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, Belfer Research Building, 413 E. 69th St., New York, NY, 10021, USA.
| |
Collapse
|
75
|
Brunkard JO, Xu M, Scarpin MR, Chatterjee S, Shemyakina EA, Goodman HM, Zambryski P. TOR dynamically regulates plant cell-cell transport. Proc Natl Acad Sci U S A 2020; 117:5049-5058. [PMID: 32051250 PMCID: PMC7060719 DOI: 10.1073/pnas.1919196117] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The coordinated redistribution of sugars from mature "source" leaves to developing "sink" leaves requires tight regulation of sugar transport between cells via plasmodesmata (PD). Although fundamental to plant physiology, the mechanisms that control PD transport and thereby support development of new leaves have remained elusive. From a forward genetic screen for altered PD transport, we discovered that the conserved eukaryotic glucose-TOR (TARGET OF RAPAMYCIN) metabolic signaling network restricts PD transport in leaves. Genetic approaches and chemical or physiological treatments to either promote or disrupt TOR activity demonstrate that glucose-activated TOR decreases PD transport in leaves. We further found that TOR is significantly more active in mature leaves photosynthesizing excess sugars than in young, growing leaves, and that this increase in TOR activity correlates with decreased rates of PD transport. We conclude that leaf cells regulate PD trafficking in response to changing carbohydrate availability monitored by the TOR pathway.
Collapse
Affiliation(s)
- Jacob O Brunkard
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720;
- Plant Gene Expression Center, US Department of Agriculture, Agricultural Research Service, Albany, CA 94710
- Innovative Genomics Institute, Berkeley, CA 94720
| | - Min Xu
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720
- Department of Biology, Northwest University, 710069 Xi'an, China
| | - M Regina Scarpin
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720
- Plant Gene Expression Center, US Department of Agriculture, Agricultural Research Service, Albany, CA 94710
| | - Snigdha Chatterjee
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720
- Plant Gene Expression Center, US Department of Agriculture, Agricultural Research Service, Albany, CA 94710
- Innovative Genomics Institute, Berkeley, CA 94720
| | - Elena A Shemyakina
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720
- Plant Gene Expression Center, US Department of Agriculture, Agricultural Research Service, Albany, CA 94710
| | - Howard M Goodman
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720
| | - Patricia Zambryski
- Department of Plant and Microbial Biology, University of California, Berkeley CA 94720;
| |
Collapse
|
76
|
De Pasquale V, Costanzo M, Siciliano RA, Mazzeo MF, Pistorio V, Bianchi L, Marchese E, Ruoppolo M, Pavone LM, Caterino M. Proteomic Analysis of Mucopolysaccharidosis IIIB Mouse Brain. Biomolecules 2020; 10:biom10030355. [PMID: 32111039 PMCID: PMC7175334 DOI: 10.3390/biom10030355] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Mucopolysaccharidosis IIIB (MPS IIIB) is an inherited metabolic disease due to deficiency of α-N-Acetylglucosaminidase (NAGLU) enzyme with subsequent storage of undegraded heparan sulfate (HS). The main clinical manifestations of the disease are profound intellectual disability and neurodegeneration. A label-free quantitative proteomic approach was applied to compare the proteome profile of brains from MPS IIIB and control mice to identify altered neuropathological pathways of MPS IIIB. Proteins were identified through a bottom up analysis and 130 were significantly under-represented and 74 over-represented in MPS IIIB mouse brains compared to wild type (WT). Multiple bioinformatic analyses allowed to identify three major clusters of the differentially abundant proteins: proteins involved in cytoskeletal regulation, synaptic vesicle trafficking, and energy metabolism. The proteome profile of NAGLU-/- mouse brain could pave the way for further studies aimed at identifying novel therapeutic targets for the MPS IIIB. Data are available via ProteomeXchange with the identifier PXD017363.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
- CEINGE-Biotecnologie Avanzate scarl, 80145 Naples, Italy;
| | | | | | - Valeria Pistorio
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
| | - Laura Bianchi
- Laboratory of Functional Proteomics, Department of Life Sciences, University of Siena, 53100 Siena, Italy;
| | - Emanuela Marchese
- CEINGE-Biotecnologie Avanzate scarl, 80145 Naples, Italy;
- Department of Mental Health and Preventive Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
- CEINGE-Biotecnologie Avanzate scarl, 80145 Naples, Italy;
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
- Correspondence: ; Tel.: +39-081-7463043
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.D.P.); (M.C.); (V.P.); (M.R.); (M.C.)
- CEINGE-Biotecnologie Avanzate scarl, 80145 Naples, Italy;
| |
Collapse
|
77
|
Nazemi M, Rainero E. Cross-Talk Between the Tumor Microenvironment, Extracellular Matrix, and Cell Metabolism in Cancer. Front Oncol 2020; 10:239. [PMID: 32175281 PMCID: PMC7054479 DOI: 10.3389/fonc.2020.00239] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of secreted proteins which provides support for tissues and organs. Additionally, the ECM controls a plethora of cell functions, including cell polarity, migration, proliferation, and oncogenic transformation. One of the hallmarks of cancer is altered cell metabolism, which is currently being exploited to develop anti-cancer therapies. Several pieces of evidence indicate that the tumor microenvironment and the ECM impinge on tumor cell metabolism. Therefore, it is essential to understand the contribution of the complex 3D microenvironment in controlling metabolic plasticity and responsiveness to therapies targeting cell metabolism. In this mini-review, we will describe how the tumor microenvironment and cancer-associated fibroblasts dictate cancer cell metabolism, resulting in increased tumor progression. Moreover, we will define the cross-talk between nutrient signaling and the trafficking of the ECM receptors of the integrin family. Finally, we will present recent data highlighting the contribution of nutrient scavenging from the microenvironment to support cancer cells growth under nutrient starvation conditions.
Collapse
Affiliation(s)
- Mona Nazemi
- Biomedical Science Department, The University of Sheffield, Sheffield, United Kingdom
| | - Elena Rainero
- Biomedical Science Department, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
78
|
Meng D, Yang Q, Wang H, Melick CH, Navlani R, Frank AR, Jewell JL. Glutamine and asparagine activate mTORC1 independently of Rag GTPases. J Biol Chem 2020; 295:2890-2899. [PMID: 32019866 DOI: 10.1074/jbc.ac119.011578] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/24/2020] [Indexed: 12/14/2022] Open
Abstract
Nutrient sensing by cells is crucial, and when this sensing mechanism is disturbed, human disease can occur. mTOR complex 1 (mTORC1) senses amino acids to control cell growth, metabolism, and autophagy. Leucine, arginine, and methionine signal to mTORC1 through the well-characterized Rag GTPase signaling pathway. In contrast, glutamine activates mTORC1 through a Rag GTPase-independent mechanism that requires ADP-ribosylation factor 1 (Arf1). Here, using several biochemical and genetic approaches, we show that eight amino acids filter through the Rag GTPase pathway. Like glutamine, asparagine signals to mTORC1 through Arf1 in the absence of the Rag GTPases. Both the Rag-dependent and Rag-independent pathways required the lysosome and lysosomal function for mTORC1 activation. Our results show that mTORC1 is differentially regulated by amino acids through two distinct pathways.
Collapse
Affiliation(s)
- Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Qianmei Yang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Huanyu Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Chase H Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Rishika Navlani
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Anderson R Frank
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390.
| |
Collapse
|
79
|
Vernizzi L, Paiardi C, Licata G, Vitali T, Santarelli S, Raneli M, Manelli V, Rizzetto M, Gioria M, Pasini ME, Grifoni D, Vanoni MA, Gellera C, Taroni F, Bellosta P. Glutamine Synthetase 1 Increases Autophagy Lysosomal Degradation of Mutant Huntingtin Aggregates in Neurons, Ameliorating Motility in a Drosophila Model for Huntington's Disease. Cells 2020; 9:cells9010196. [PMID: 31941072 PMCID: PMC7016901 DOI: 10.3390/cells9010196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
Glutamine Synthetase 1 (GS1) is a key enzyme that catalyzes the ATP-dependent synthesis of l-glutamine from l-glutamate and is also member of the Glutamate Glutamine Cycle, a complex physiological process between glia and neurons that controls glutamate homeostasis and is often found compromised in neurodegenerative diseases including Huntington's disease (HD). Here we report that the expression of GS1 in neurons ameliorates the motility defects induced by the expression of the mutant Htt, using a Drosophila model for HD. This phenotype is associated with the ability of GS1 to favor the autophagy that we associate with the presence of reduced Htt toxic protein aggregates in neurons expressing mutant Htt. Expression of GS1 prevents the TOR activation and phosphorylation of S6K, a mechanism that we associate with the reduced levels of essential amino acids, particularly of arginine and asparagine important for TOR activation. This study reveals a novel function for GS1 to ameliorate neuronal survival by changing amino acids' levels that induce a "starvation-like" condition responsible to induce autophagy. The identification of novel targets that inhibit TOR in neurons is of particular interest for the beneficial role that autophagy has in preserving physiological neuronal health and in the mechanisms that eliminate the formation of toxic aggregates in proteinopathies.
Collapse
Affiliation(s)
- Luisa Vernizzi
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Chiara Paiardi
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Giusimaria Licata
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Teresa Vitali
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Stefania Santarelli
- Department of Cellular, Computational and Integrative Biology (CiBio), University of Trento, 38123 Trento, Italy;
| | - Martino Raneli
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Vera Manelli
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Manuela Rizzetto
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.R.); (C.G.); (F.T.)
| | - Mariarosa Gioria
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Maria E. Pasini
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Daniela Grifoni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Maria A. Vanoni
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.R.); (C.G.); (F.T.)
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.R.); (C.G.); (F.T.)
| | - Paola Bellosta
- Department of Biosciences, University of Milan, 20133 Milan, Italy; (L.V.); (C.P.); (T.V.); (M.R.); (V.M.); (M.G.); (M.E.P.); (M.A.V.)
- Department of Cellular, Computational and Integrative Biology (CiBio), University of Trento, 38123 Trento, Italy;
- Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA
- Correspondence: ; Tel.: +39-0461-283070
| |
Collapse
|
80
|
Translational Landscape of mTOR Signaling in Integrating Cues Between Cancer and Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:69-80. [PMID: 32030685 DOI: 10.1007/978-3-030-35582-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mammalian target of rapamycin (mTOR) represents a critical hub for the regulation of different processes in both normal and tumor cells. Furthermore, it is now well established the role of mTOR in integrating and shaping different environmental paracrine and autocrine stimuli in tumor microenvironment (TME) constituents. Recently, further efforts have been employed to understand how the mTOR signal transduction mechanisms modulate the sensitivity and resistance to targeted therapies, also for its involvement of mTOR also in modulating angiogenesis and tumor immunity. Indeed, interest in mTOR targeting was increased to improve immune response against cancer and to develop new long-term efficacy strategies, as demonstrated by clinical success of mTOR and immune checkpoint inhibitor combinations. In this chapter, we will describe the role of mTOR in modulating TME elements and the implication in its targeting as a great promise in clinical trials.
Collapse
|
81
|
Kakihara Y, Kiguchi T, Ohazama A, Saeki M. R2TP/PAQosome as a promising chemotherapeutic target in cancer. JAPANESE DENTAL SCIENCE REVIEW 2020; 56:38-42. [PMID: 31890057 PMCID: PMC6926247 DOI: 10.1016/j.jdsr.2019.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/27/2019] [Accepted: 08/21/2019] [Indexed: 11/30/2022] Open
Abstract
R2TP/PAQosome (particle for arrangement of quaternary structure) is a novel multisubunit chaperone specialized in the assembly/maturation of protein complexes that are involved in essential cellular processes such as PIKKs (phosphatidylinositol 3-kinase-like kinases) signaling, snoRNP (small nucleolar ribonucleoprotein) biogenesis, and RNAP II (RNA polymerase II) complex formation. In this review article, we describe the current understanding of R2TP/PAQosome functions and characteristics as well as how the chaperone complex is involved in oncogenesis, highlighting DNA damage response, mTOR (mammalian target of rapamycin) pathway as well as snoRNP biogenesis. Also, we discuss its possible involvement in HNSCC (head and neck squamous cell carcinoma) including OSCC (oral squamous cell carcinoma). Finally, we provide an overview of current anti-cancer drug development efforts targeting R2TP/PAQosome.
Collapse
Affiliation(s)
- Yoshito Kakihara
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tetsuo Kiguchi
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Atsushi Ohazama
- Division of Oral Anatomy, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makio Saeki
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
82
|
Mello T, Materozzi M, Zanieri F, Simeone I, Ceni E, Bereshchenko O, Polvani S, Tarocchi M, Marroncini G, Nerlov C, Guasti D, Bani D, Pinzani M, Galli A. Liver haploinsufficiency of RuvBL1 causes hepatic insulin resistance and enhances hepatocellular carcinoma progression. Int J Cancer 2019; 146:3410-3422. [PMID: 31721195 DOI: 10.1002/ijc.32787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/29/2022]
Abstract
RuvBL1 is an AAA+ ATPase whose expression in hepatocellular carcinoma (HCC) correlates with a poor prognosis. In vitro models suggest that targeting RuvBL1 could be an effective strategy against HCC. However, the role of RuvBL1 in the onset and progression of HCC remains unknown. To address this question, we developed a RuvBL1hep+/- mouse model and evaluated the outcome of DEN-induced liver carcinogenesis up to 12 months of progression. We found that RuvBL1 haploinsufficiency initially delayed the onset of liver cancer, due to a reduced hepatocyte turnover in RuvBL1hep+/- mice. However, RuvBL1hep+/- mice eventually developed HCC nodules that, with aging, grew larger than in the control mice. Moreover, RuvBL1hep+/- mice developed hepatic insulin resistance and impaired glucose homeostasis. We could determine that RuvBL1 regulates insulin signaling through the Akt/mTOR pathway in liver physiology in vivo as well as in normal hepatocytic and HCC cells in vitro. Whole transcriptome analysis of mice livers confirmed the major role of RuvBL1 in the regulation of hepatic glucose metabolism. Finally, RuvBL1 expression was found significantly correlated to glucose metabolism and mTOR signaling by bioinformatic analysis of human HCC sample from the publicly available TGCA database. These data uncover a role of RuvBL1 at the intersection of liver metabolism, hepatocyte proliferation and HCC development, providing a molecular rationale for its overexpression in liver cancer.
Collapse
Affiliation(s)
- Tommaso Mello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Maria Materozzi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Francesca Zanieri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Irene Simeone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Ceni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | | | - Simone Polvani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Mirko Tarocchi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Giada Marroncini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Claus Nerlov
- MRC Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Daniele Guasti
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Daniele Bani
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, London, United Kingdom
| | - Andrea Galli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
83
|
New insights into the evolutionary conservation of the sole PIKK pseudokinase Tra1/TRRAP. Biochem Soc Trans 2019; 47:1597-1608. [DOI: 10.1042/bst20180496] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/25/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Phosphorylation by protein kinases is a fundamental mechanism of signal transduction. Many kinase families contain one or several members that, although evolutionarily conserved, lack the residues required for catalytic activity. Studies combining structural, biochemical, and functional approaches revealed that these pseudokinases have crucial roles in vivo and may even represent attractive targets for pharmacological intervention. Pseudokinases mediate signal transduction by a diversity of mechanisms, including allosteric regulation of their active counterparts, assembly of signaling hubs, or modulation of protein localization. One such pseudokinase, named Tra1 in yeast and transformation/transcription domain-associated protein (TRRAP) in mammals, is the only member lacking all catalytic residues within the phosphatidylinositol 3-kinase related kinase (PIKK) family of kinases. PIKKs are related to the PI3K family of lipid kinases, but function as Serine/Threonine protein kinases and have pivotal roles in diverse processes such as DNA damage sensing and repair, metabolic control of cell growth, nonsense-mediated decay, or transcription initiation. Tra1/TRRAP is the largest subunit of two distinct transcriptional co-activator complexes, SAGA and NuA4/TIP60, which it recruits to promoters upon transcription factor binding. Here, we review our current knowledge on the Tra1/TRRAP pseudokinase, focusing on its role as a scaffold for SAGA and NuA4/TIP60 complex assembly and recruitment to chromatin. We further discuss its evolutionary history within the PIKK family and highlight recent findings that reveal the importance of molecular chaperones in pseudokinase folding, function, and conservation.
Collapse
|
84
|
Wilson TG, Hanna A, Recknagel J, Pruetz BL, Baschnagel AM, Wilson GD. Prognostic significance of MTOR expression in HPV positive and negative head and neck cancers treated by chemoradiation. Head Neck 2019; 42:153-162. [PMID: 31657099 DOI: 10.1002/hed.25983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/29/2019] [Accepted: 09/17/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The mechanistic target of rapamycin (MTOR) plays a key role in regulating cell growth and metabolism and is commonly overexpressed in head and neck cancer (HNSCC). This study investigated the association of MTOR with clinical outcome in human papilloma virus (HPV) positive and negative HNSCC patients treated by chemoradiation. METHODS A tissue microarray (TMA) consisting of cores from 109 HNSCC patients treated by definitive chemoradiation was constructed and stained with antibodies against p16 and MTOR and expression correlated with clinicopathological features and clinical outcome. RESULTS MTOR varied widely between tumor cores and was not associated with HPV status or clinicopathological features. There was a positive correlation with pre-treatment FDG uptake. (P = .01). In HPV negative patients, MTOR predicted for shorter locoregional control (P = .02), diseases free survival (P = .02), and overall survival (P = .04). MTOR expression was not associated with outcome in HPV positive patients. CONCLUSIONS Prognostic significance of MTOR expression depends on HPV status.
Collapse
Affiliation(s)
- Thomas G Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Alaa Hanna
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Johnathon Recknagel
- Oakland University William Beaumont School of Medicine, Oakland University, Rochester, Michigan
| | - Barbara L Pruetz
- Beaumont BioBank, William Beaumont Hospital, Royal Oak, Michigan
| | - Andrew M Baschnagel
- Department of Human Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| |
Collapse
|
85
|
Yamada D, Kawabe K, Tosa I, Tsukamoto S, Nakazato R, Kou M, Fujikawa K, Nakamura S, Ono M, Oohashi T, Kaneko M, Go S, Hinoi E, Yoneda Y, Takarada T. Inhibition of the glutamine transporter SNAT1 confers neuroprotection in mice by modulating the mTOR-autophagy system. Commun Biol 2019; 2:346. [PMID: 31552299 PMCID: PMC6751179 DOI: 10.1038/s42003-019-0582-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 08/19/2019] [Indexed: 01/31/2023] Open
Abstract
The pathophysiological role of mammalian target of rapamycin complex 1 (mTORC1) in neurodegenerative diseases is established, but possible therapeutic targets responsible for its activation in neurons must be explored. Here we identified solute carrier family 38a member 1 (SNAT1, Slc38a1) as a positive regulator of mTORC1 in neurons. Slc38a1flox/flox and Synapsin I-Cre mice were crossed to generate mutant mice in which Slc38a1 was selectively deleted in neurons. Measurement of 2,3,5-triphenyltetrazolium chloride (TTC) or the MAP2-negative area in a mouse model of middle cerebral artery occlusion (MCAO) revealed that Slc38a1 deficiency decreased infarct size. We found a transient increase in the phosphorylation of p70S6k1 (pp70S6k1) and a suppressive effect of rapamycin on infarct size in MCAO mice. Autophagy inhibitors completely mitigated the suppressive effect of SNAT1 deficiency on neuronal cell death under in vitro stroke culture conditions. These results demonstrate that SNAT1 promoted ischemic brain damage via mTOR-autophagy system.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Kenji Kawabe
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Ikue Tosa
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Shunpei Tsukamoto
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Ryota Nakazato
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Miki Kou
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Koichi Fujikawa
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Saki Nakamura
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Mari Kaneko
- Laboratory for Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima Minami, Chuou-ku, Kobe, Hyogo 650-0047 Japan
| | - Shioi Go
- Laboratory for Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima Minami, Chuou-ku, Kobe, Hyogo 650-0047 Japan
| | - Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| |
Collapse
|
86
|
Structural and functional analysis of the role of the chaperonin CCT in mTOR complex assembly. Nat Commun 2019; 10:2865. [PMID: 31253771 PMCID: PMC6599039 DOI: 10.1038/s41467-019-10781-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/01/2019] [Indexed: 01/01/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase forms two multi-protein signaling complexes, mTORC1 and mTORC2, which are master regulators of cell growth, metabolism, survival and autophagy. Two of the subunits of these complexes are mLST8 and Raptor, β-propeller proteins that stabilize the mTOR kinase and recruit substrates, respectively. Here we report that the eukaryotic chaperonin CCT plays a key role in mTORC assembly and signaling by folding both mLST8 and Raptor. A high resolution (4.0 Å) cryo-EM structure of the human mLST8-CCT intermediate isolated directly from cells shows mLST8 in a near-native state bound to CCT deep within the folding chamber between the two CCT rings, and interacting mainly with the disordered N- and C-termini of specific CCT subunits of both rings. These findings describe a unique function of CCT in mTORC assembly and a distinct binding site in CCT for mLST8, far from those found for similar β-propeller proteins. β-propeller domains are an important class of folding substrates for the eukaryotic cytosolic chaperonin CTT. Here the authors find that CTT contributes to the folding and assembly of two β-propeller proteins from mTOR complexes, mLST8 and Raptor, and determine the 4.0 Å cryoEM structure of a human mLST8-CCT intermediate that shows mLST8 in a near-native state.
Collapse
|
87
|
Dihydrocapsaicin Inhibits Epithelial Cell Transformation through Targeting Amino Acid Signaling and c-Fos Expression. Nutrients 2019; 11:nu11061269. [PMID: 31167465 PMCID: PMC6627986 DOI: 10.3390/nu11061269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 01/09/2023] Open
Abstract
Chili peppers are one of the most widely consumed spices worldwide. However, research on the health benefits of chili peppers and some of its constituents has raised controversy as to whether chili pepper compounds possess cancer-promoting or cancer-preventive effects. While ample studies have been carried out to examine the effect of capsaicin in carcinogenesis, the chemopreventive effect of other major components in chili pepper, including dihydrocapsaicin, capsiate, and capsanthin, is relatively unclear. Herein, we investigated the inhibitory effect of chili pepper components on malignant cell transformation. Among the tested chili pepper compounds, dihydrocapsaicin displayed the strongest inhibitory activity against epidermal growth factor (EGF)-induced neoplastic transformation. Dihydrocapsaicin specifically suppressed EGF-induced phosphorylations of the p70S6K1-S6 pathway and the expression of c-Fos. A reduction in c-Fos levels by dihydrocapsaicin led to a concomitant downregulation of AP-1 activation. Further analysis of the molecular mechanism responsible for the dihydrocapsaicin-mediated decrease in phospho-p70S6K1, revealed that dihydrocapsaicin can block amino acid-dependent mechanistic targets of rapamycin complex 1 (mTORC1)-p70S6K1-S6 signal activation. Additionally, dihydrocapsaicin was able to selectively augment amino acid deprivation-induced cell death in mTORC1-hyperactive cells. Collectively, dihydrocapsaicin exerted chemopreventive effects through inhibiting amino acid signaling and c-Fos pathways and, thus, might be a promising cancer preventive natural agent.
Collapse
|
88
|
Jewell JL, Fu V, Hong AW, Yu FX, Meng D, Melick CH, Wang H, Lam WLM, Yuan HX, Taylor SS, Guan KL. GPCR signaling inhibits mTORC1 via PKA phosphorylation of Raptor. eLife 2019; 8:43038. [PMID: 31112131 PMCID: PMC6529218 DOI: 10.7554/elife.43038] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/30/2019] [Indexed: 01/14/2023] Open
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth, metabolism, and autophagy. Extensive research has focused on pathways that activate mTORC1 like growth factors and amino acids; however, much less is known about signaling cues that directly inhibit mTORC1 activity. Here, we report that G-protein coupled receptors (GPCRs) paired to Gαs proteins increase cyclic adenosine 3’5’ monophosphate (cAMP) to activate protein kinase A (PKA) and inhibit mTORC1. Mechanistically, PKA phosphorylates the mTORC1 component Raptor on Ser 791, leading to decreased mTORC1 activity. Consistently, in cells where Raptor Ser 791 is mutated to Ala, mTORC1 activity is partially rescued even after PKA activation. Gαs-coupled GPCRs stimulation leads to inhibition of mTORC1 in multiple cell lines and mouse tissues. Our results uncover a signaling pathway that directly inhibits mTORC1, and suggest that GPCRs paired to Gαs proteins may be potential therapeutic targets for human diseases with hyperactivated mTORC1.
Collapse
Affiliation(s)
- Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Vivian Fu
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Moores Cancer Center, University of California San Diego, La Jolla, United States
| | - Audrey W Hong
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Moores Cancer Center, University of California San Diego, La Jolla, United States
| | - Fa-Xing Yu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chase H Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Huanyu Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Wai-Ling Macrina Lam
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Moores Cancer Center, University of California San Diego, La Jolla, United States
| | - Hai-Xin Yuan
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Moores Cancer Center, University of California San Diego, La Jolla, United States
| | - Susan S Taylor
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, United States
| | - Kun-Liang Guan
- Department of Pharmacology, University of California, San Diego, La Jolla, United States.,Moores Cancer Center, University of California San Diego, La Jolla, United States
| |
Collapse
|
89
|
Liao GY, Lee MT, Fan JJ, Hsiao PW, Lee CS, Su SY, Hwang JJ, Ke FC. Blockage of glutamine-dependent anaplerosis affects mTORC1/2 activity and ultimately leads to cellular senescence-like response. Biol Open 2019; 8:bio.038257. [PMID: 31097446 PMCID: PMC6550068 DOI: 10.1242/bio.038257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The purpose of study was to explore the role of glutamine-dependent anaplerosis in cell fate determination (proliferation and senescence) and the potential associated mechanism by employing a pharmacological inhibitor of glutamine-dependent anaplerosis, amino-oxyacetate (AOA). Using the WI38 normal human embryonic fibroblast cell line, we found that exposure to AOA induced mTORC1 inactivation−mTORC2 activation (within day 1), cell cycle arrest (day 2–6) and cellular senescence (day 4–6). These AOA effects were blocked by concomitantly providing anaplerotic factors [α-ketoglutarate (αKG), pyruvate or oxaloacetate], and not affected by ROS scavenger N-acetyl-cysteine (NAC). Moreover, AOA-induced cellular senescence in WI38 cells is associated with elevated protein levels of p53, p21CIP1 and p16INK4A and decreased Rb protein level, which was blocked by αKG supplementation. In p16INK4A-deficient U2OS human osteosarcoma cells and p16INK4A-knockdown WI38 cells, AOA exposure also induced similar effects on cell proliferation, and protein level of P-Rb-S807/811 and Rb. Interestingly, no AOA induction of cellular senescence was observed in U2OS cells, yet was still seen in p16INK4A-knockdown WI38 cells accompanied by the presence of p16 antibody-reactive p12. In summary, we disclose that glutamine-dependent anaplerosis is essential to cell growth and closely associated with mTORC1 activation and mTORC2 inactivation, and impedes cellular senescence particularly associated with p16INK4A. Summary: Glutamine-dependent anaplerosis is essential to cell growth and closely associated with mTORC1 activation and mTORC2 inactivation, and impedes cellular senescence particularly associated with p16INK4A.
Collapse
Affiliation(s)
- Geng-You Liao
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan.,Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Ming-Ting Lee
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Jhen-Jia Fan
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chun-Sheng Lee
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Shou-Yi Su
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Jiuan-Jiuan Hwang
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Ferng-Chun Ke
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
90
|
Muñoz-Hernández H, Pal M, Rodríguez CF, Fernandez-Leiro R, Prodromou C, Pearl LH, Llorca O. Structural mechanism for regulation of the AAA-ATPases RUVBL1-RUVBL2 in the R2TP co-chaperone revealed by cryo-EM. SCIENCE ADVANCES 2019; 5:eaaw1616. [PMID: 31049401 PMCID: PMC6494491 DOI: 10.1126/sciadv.aaw1616] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/16/2019] [Indexed: 05/04/2023]
Abstract
The human R2TP complex (RUVBL1-RUVBL2-RPAP3-PIH1D1) is an HSP90 co-chaperone required for the maturation of several essential multiprotein complexes, including RNA polymerase II, small nucleolar ribonucleoproteins, and PIKK complexes such as mTORC1 and ATR-ATRIP. RUVBL1-RUVBL2 AAA-ATPases are also primary components of other essential complexes such as INO80 and Tip60 remodelers. Despite recent efforts, the molecular mechanisms regulating RUVBL1-RUVBL2 in these complexes remain elusive. Here, we report cryo-EM structures of R2TP and show how access to the nucleotide-binding site of RUVBL2 is coupled to binding of the client recruitment component of R2TP (PIH1D1) to its DII domain. This interaction induces conformational rearrangements that lead to the destabilization of an N-terminal segment of RUVBL2 that acts as a gatekeeper to nucleotide exchange. This mechanism couples protein-induced motions of the DII domains with accessibility of the nucleotide-binding site in RUVBL1-RUVBL2, and it is likely a general mechanism shared with other RUVBL1-RUVBL2-containing complexes.
Collapse
Affiliation(s)
- Hugo Muñoz-Hernández
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Mohinder Pal
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Carlos F. Rodríguez
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Rafael Fernandez-Leiro
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Chrisostomos Prodromou
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Laurence H. Pearl
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Corresponding author.
| |
Collapse
|
91
|
Jamsheer K M, Jindal S, Laxmi A. Evolution of TOR-SnRK dynamics in green plants and its integration with phytohormone signaling networks. JOURNAL OF EXPERIMENTAL BOTANY 2019; 70:2239-2259. [PMID: 30870564 DOI: 10.1093/jxb/erz107] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/26/2019] [Indexed: 05/07/2023]
Abstract
The target of rapamycin (TOR)-sucrose non-fermenting 1 (SNF1)-related protein kinase 1 (SnRK1) signaling is an ancient regulatory mechanism that originated in eukaryotes to regulate nutrient-dependent growth. Although the TOR-SnRK1 signaling cascade shows highly conserved functions among eukaryotes, studies in the past two decades have identified many important plant-specific innovations in this pathway. Plants also possess SnRK2 and SnRK3 kinases, which originated from the ancient SnRK1-related kinases and have specialized roles in controlling growth, stress responses and nutrient homeostasis in plants. Recently, an integrative picture has started to emerge in which different SnRKs and TOR kinase are highly interconnected to control nutrient and stress responses of plants. Further, these kinases are intimately involved with phytohormone signaling networks that originated at different stages of plant evolution. In this review, we highlight the evolution and divergence of TOR-SnRK signaling components in plants and their communication with each other as well as phytohormone signaling to fine-tune growth and stress responses in plants.
Collapse
Affiliation(s)
- Muhammed Jamsheer K
- Amity Food & Agriculture Foundation, Amity University Uttar Pradesh, Noida, India
| | - Sunita Jindal
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, India
| | - Ashverya Laxmi
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
92
|
Wu Y, Shi L, Li L, Fu L, Liu Y, Xiong Y, Sheen J. Integration of nutrient, energy, light, and hormone signalling via TOR in plants. JOURNAL OF EXPERIMENTAL BOTANY 2019; 70:2227-2238. [PMID: 30715492 PMCID: PMC6463029 DOI: 10.1093/jxb/erz028] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/08/2019] [Indexed: 05/04/2023]
Abstract
The multidomain target of rapamycin (TOR) is an atypical serine/threonine protein kinase resembling phosphatidylinositol lipid kinases, but retains high sequence identity and serves a remarkably conserved role as a master signalling integrator in yeasts, plants, and humans. TOR dynamically orchestrates cell metabolism, biogenesis, organ growth, and development transitions in response to nutrient, energy, hormone, and environmental cues. Here we review recent findings on the versatile and complex roles of TOR in transcriptome reprogramming, seedling, root, and shoot growth, and root hair production activated by sugar and energy signalling. We explore how co-ordination of TOR-mediated light and hormone signalling is involved in root and shoot apical meristem activation, proliferation of leaf primordia, cotyledon/leaf greening, and hypocotyl elongation. We also discuss the emerging TOR functions in response to sulfur assimilation and metabolism and consider potential molecular links and positive feedback loops between TOR, sugar, energy, and other essential macronutrients.
Collapse
Affiliation(s)
- Yue Wu
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lin Shi
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lei Li
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Liwen Fu
- Basic Forestry and Proteomics Research Centre, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fujian Province, PR China
| | - Yanlin Liu
- Basic Forestry and Proteomics Research Centre, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fujian Province, PR China
| | - Yan Xiong
- Basic Forestry and Proteomics Research Centre, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fujian Province, PR China
| | - Jen Sheen
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
93
|
Van Leene J, Han C, Gadeyne A, Eeckhout D, Matthijs C, Cannoot B, De Winne N, Persiau G, Van De Slijke E, Van de Cotte B, Stes E, Van Bel M, Storme V, Impens F, Gevaert K, Vandepoele K, De Smet I, De Jaeger G. Capturing the phosphorylation and protein interaction landscape of the plant TOR kinase. NATURE PLANTS 2019; 5:316-327. [PMID: 30833711 DOI: 10.1038/s41477-019-0378-z] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/28/2019] [Indexed: 05/18/2023]
Abstract
The target of rapamycin (TOR) kinase is a conserved regulatory hub that translates environmental and nutritional information into permissive or restrictive growth decisions. Despite the increased appreciation of the essential role of the TOR complex in plants, no large-scale phosphoproteomics or interactomics studies have been performed to map TOR signalling events in plants. To fill this gap, we combined a systematic phosphoproteomics screen with a targeted protein complex analysis in the model plant Arabidopsis thaliana. Integration of the phosphoproteome and protein complex data on the one hand shows that both methods reveal complementary subspaces of the plant TOR signalling network, enabling proteome-wide discovery of both upstream and downstream network components. On the other hand, the overlap between both data sets reveals a set of candidate direct TOR substrates. The integrated network embeds both evolutionarily-conserved and plant-specific TOR signalling components, uncovering an intriguing complex interplay with protein synthesis. Overall, the network provides a rich data set to start addressing fundamental questions about how TOR controls key processes in plants, such as autophagy, auxin signalling, chloroplast development, lipid metabolism, nucleotide biosynthesis, protein translation or senescence.
Collapse
Affiliation(s)
- Jelle Van Leene
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Chao Han
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
- The Key Laboratory of Plant Cell Engineering and Germplasm Innovation, College of Life Sciences, Shandong University, Jinan, China
| | - Astrid Gadeyne
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Dominique Eeckhout
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Caroline Matthijs
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Bernard Cannoot
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Nancy De Winne
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Geert Persiau
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Eveline Van De Slijke
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Brigitte Van de Cotte
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Elisabeth Stes
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Michiel Van Bel
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, Ghent, Belgium
| | - Veronique Storme
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Francis Impens
- Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Center for Medical Biotechnology, Ghent, Belgium
- VIB Proteomics Core, Ghent, Belgium
| | - Kris Gevaert
- Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Center for Medical Biotechnology, Ghent, Belgium
- VIB Proteomics Core, Ghent, Belgium
| | - Klaas Vandepoele
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, Ghent, Belgium
| | - Ive De Smet
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Geert De Jaeger
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium.
- VIB Center for Plant Systems Biology, Ghent, Belgium.
| |
Collapse
|
94
|
Wu J, Xiao S, Ren J, Zhang D. A unified mitochondria mechanistic target of rapamycin acyl-coenzyme A dehydrogenase 10 signal relay modulation for metformin growth inhibition in human immortalized keratinocytes cells. J Cell Biochem 2019; 120:1773-1782. [PMID: 30206977 DOI: 10.1002/jcb.27481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 07/19/2018] [Indexed: 01/24/2023]
Abstract
Metformin exhibits antiproliferative and proapoptotic effects in a variety of diseases, characterized by malignant and nonmalignant hyperplastic cells; however, the underlying molecular mechanism of metformin in psoriasis has not been elucidated. In the current study, we found that after metformin treatment the proliferation of human immortalized keratinocytes (HaCaT) was significantly inhibited, while cell apoptosis was increased in a dose-dependent manner, accompanied with enhanced protein expression of acyl-coenzyme A dehydrogenase 10 (ACAD10). Furthermore, mechanism analysis revealed that ACAD10 expression is induced by downregulated activities of mechanistic target of rapamycin 1 (mTORC1) signaling rather than AMP-activated protein kinase signaling. The inactivation of mTORC1 by rapamycin pretreatment or rotenone-induced mitochondrial complex inhibition showed a similar effect because of the metformin treatment on the proliferation and apoptosis of HaCaT keratinocytes. Overexpression of mTORC1 almost reversed the antiproliferation and proapoptosis effects induced by metformin. This study showed that the metformin treatment inhibited HaCaT cells proliferation and promoted apoptosis by affecting the mitochondrial-mTORC1 signaling and elevated the ACAD10 expression. Hence, metformin can be used as a potential therapeutic agent for psoriasis.
Collapse
Affiliation(s)
- Jiawen Wu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shengxiang Xiao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianwen Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dingwei Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
95
|
Lucarelli G, Rutigliano M, Sallustio F, Ribatti D, Giglio A, Signorile ML, Grossi V, Sanese P, Napoli A, Maiorano E, Bianchi C, Perego RA, Ferro M, Ranieri E, Serino G, Bell LN, Ditonno P, Simone C, Battaglia M. Integrated multi-omics characterization reveals a distinctive metabolic signature and the role of NDUFA4L2 in promoting angiogenesis, chemoresistance, and mitochondrial dysfunction in clear cell renal cell carcinoma. Aging (Albany NY) 2018; 10:3957-3985. [PMID: 30538212 PMCID: PMC6326659 DOI: 10.18632/aging.101685] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/22/2018] [Indexed: 11/25/2022]
Abstract
An altered metabolism is involved in the development of clear cell - renal cell carcinoma (ccRCC), and in this tumor many altered genes play a fundamental role in controlling cell metabolic activities. We delineated a large-scale metabolomic profile of human ccRCC, and integrated it with transcriptomic data to connect the variations in cancer metabolism with gene expression changes. Moreover, to better analyze the specific contribution of metabolic gene alterations potentially associated with tumorigenesis and tumor progression, we evaluated the transcription profile of primary renal tumor cells. Untargeted metabolomic analysis revealed a signature of an increased glucose uptake and utilization in ccRCC. In addition, metabolites related to pentose phosphate pathway were also altered in the tumor samples in association with changes in Krebs cycle intermediates and related metabolites. We identified NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4-like 2 (NDUFA4L2) as the most highly expressed gene in renal cancer cells and evaluated its role in sustaining angiogenesis, chemoresistance, and mitochondrial dysfunction. Finally, we showed that silencing of NDUFA4L2 affects cell viability, increases mitochondrial mass, and induces ROS generation in hypoxia.
Collapse
Affiliation(s)
- Giuseppe Lucarelli
- Department of Emergency and Organ Transplantation- Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
- Equal contribution
| | - Monica Rutigliano
- Department of Emergency and Organ Transplantation- Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
- Equal contribution
| | - Fabio Sallustio
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Andrea Giglio
- Department of Emergency and Organ Transplantation- Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Martina Lepore Signorile
- Department of Biomedical Sciences and Human Oncology, Medical Genetics, University of Bari, Bari, Italy
| | - Valentina Grossi
- Department of Biomedical Sciences and Human Oncology, Medical Genetics, University of Bari, Bari, Italy
| | - Paola Sanese
- Department of Biomedical Sciences and Human Oncology, Medical Genetics, University of Bari, Bari, Italy
| | - Anna Napoli
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Eugenio Maiorano
- Department of Emergency and Organ Transplantation, Pathology Unit, University of Bari, Bari, Italy
| | - Cristina Bianchi
- Department of Health Sciences, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Roberto A. Perego
- Department of Health Sciences, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, Milan, Italy
| | - Elena Ranieri
- Department of Medical and Surgical Sciences, Molecular Medicine Center, Section of Clinical Pathology, University of Foggia, Foggia, Italy
| | - Grazia Serino
- National Institute of Gastroenterology, ‘S de Bellis’, Castellana Grotte, Bari, Italy
| | - Lauren N. Bell
- Metabolon, Inc., Research Triangle Park, Morrisville, NC 27560, USA
| | - Pasquale Ditonno
- Department of Emergency and Organ Transplantation- Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Cristiano Simone
- Department of Biomedical Sciences and Human Oncology, Medical Genetics, University of Bari, Bari, Italy
- National Institute of Gastroenterology, ‘S de Bellis’, Castellana Grotte, Bari, Italy
| | - Michele Battaglia
- Department of Emergency and Organ Transplantation- Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| |
Collapse
|
96
|
Javary J, Allain-Courtois N, Saucisse N, Costet P, Heraud C, Benhamed F, Pierre R, Bure C, Pallares-Lupon N, Do Cruzeiro M, Postic C, Cota D, Dubus P, Rosenbaum J, Benhamouche-Trouillet S. Liver Reptin/RUVBL2 controls glucose and lipid metabolism with opposite actions on mTORC1 and mTORC2 signalling. Gut 2018; 67:2192-2203. [PMID: 29074727 DOI: 10.1136/gutjnl-2017-314208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The AAA+ ATPase Reptin is overexpressed in hepatocellular carcinoma and preclinical studies indicate that it could be a relevant therapeutic target. However, its physiological and pathophysiological roles in vivo remain unknown. This study aimed to determine the role of Reptin in mammalian adult liver. DESIGN AND RESULTS We generated an inducible liver-specific Reptin knockout (RepinLKO ) mouse model. Following Reptin invalidation, mice displayed decreased body and fat mass, hypoglycaemia and hypolipidaemia. This was associated with decreased hepatic mTOR protein abundance. Further experiments in primary hepatocytes demonstrated that Reptin maintains mTOR protein level through its ATPase activity. Unexpectedly, loss or inhibition of Reptin induced an opposite effect on mTORC1 and mTORC2 signalling, with: (1) strong inhibition of hepatic mTORC1 activity, likely responsible for the reduction of hepatocytes cell size, for decreased de novo lipogenesis and cholesterol transcriptional programmes and (2) enhancement of mTORC2 activity associated with inhibition of the gluconeogenesis transcriptional programme and hepatic glucose production. Consequently, the role of hepatic Reptin in the pathogenesis of insulin resistance (IR) and non-alcoholic fatty liver disease consecutive to a high-fat diet was investigated. We found that Reptin deletion completely rescued pathological phenotypes associated with IR, including glucose intolerance, hyperglycaemia, hyperlipidaemia and hepatic steatosis. CONCLUSION We show here that the AAA +ATPase Reptin is a regulator of mTOR signalling in the liver and global glucido-lipidic homeostasis. Inhibition of hepatic Reptin expression or activity represents a new therapeutic perspective for metabolic syndrome.
Collapse
Affiliation(s)
- Joaquim Javary
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Nathalie Allain-Courtois
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Nicolas Saucisse
- Université de Bordeaux, Bordeaux, France.,Physiopathologie de la Plasticité Neuronale, INSERM, Neuro Centre Magendie, Bordeaux, France
| | | | - Capucine Heraud
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Fadila Benhamed
- INSERM, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France
| | - Rémi Pierre
- Université Paris Descartes, Paris, France.,Plate-forme de Recombinaison Homologue, Institut Cochin, INSERM, Paris, France
| | - Corinne Bure
- Université de Bordeaux, Bordeaux, France.,UMR , Chimie et Biologie des Membranes et des Nano objets, Bordeaux, France
| | - Nestor Pallares-Lupon
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Marcio Do Cruzeiro
- Université Paris Descartes, Paris, France.,Plate-forme de Recombinaison Homologue, Institut Cochin, INSERM, Paris, France
| | - Catherine Postic
- INSERM, Institut Cochin, Paris, France.,Université Paris Descartes, Paris, France
| | - Daniela Cota
- Université de Bordeaux, Bordeaux, France.,Physiopathologie de la Plasticité Neuronale, INSERM, Neuro Centre Magendie, Bordeaux, France
| | - Pierre Dubus
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Jean Rosenbaum
- INSERM, Bordeaux Research in Translational Oncology, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | | |
Collapse
|
97
|
Lynham J, Houry WA. The Multiple Functions of the PAQosome: An R2TP- and URI1 Prefoldin-Based Chaperone Complex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1106:37-72. [DOI: 10.1007/978-3-030-00737-9_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
98
|
Mészáros G, Pasquier A, Vivot K, Goginashvili A, Ricci R. Lysosomes in nutrient signalling: A focus on pancreatic β-cells. Diabetes Obes Metab 2018; 20 Suppl 2:104-115. [PMID: 30230186 DOI: 10.1111/dom.13389] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 01/12/2023]
Abstract
Regulated insulin secretion from pancreatic β-cells is a major process maintaining glucose homeostasis in mammals. Enhancing insulin release in response to chronic nutrient overload and obesity-related insulin resistance (pre-diabetes) requires several adaptive cellular mechanisms maintaining β-cell health under such stresses. Once these mechanisms are overwhelmed, β-cell failure occurs leading to full-blown Type 2 Diabetes (T2D). Nutrient-dependent macroautophagy represents one such adaptive mechanism in β-cells. While macroautophagy levels are high and protective in β-cells in pre-diabetes, they decrease at later stages contributing to β-cell failure. However, mechanisms compromising macroautophagy in β-cells remain poorly understood. In this review, we discuss how recently discovered signalling cascades that emanate from the limiting membrane of lysosomes contribute to changes in macroautophagy flux in physiology and disease. In particular, these mechanisms are put into context with β-cell function highlighting most recently described links between nutrient-dependent lysosomal signalling pathways and insulin secretion. Understanding these mechanisms in response to metabolic stress might pave the way for development of more tailored treatment strategies aimed at preserving β-cell health.
Collapse
Affiliation(s)
- Gergő Mészáros
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Laboratoire de Biochimie et de Biologie Moléculaire, Nouvel Hôpital Civil, Strasbourg, France
| | - Adrien Pasquier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Kevin Vivot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Alexander Goginashvili
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California
| | - Romeo Ricci
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Laboratoire de Biochimie et de Biologie Moléculaire, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
99
|
Nguyen TL, Nokin MJ, Egorov M, Tomé M, Bodineau C, Di Primo C, Minder L, Wdzieczak-Bakala J, Garcia-Alvarez MC, Bignon J, Thoison O, Delpech B, Surpateanu G, Frapart YM, Peyrot F, Abbas K, Terés S, Evrard S, Khatib AM, Soubeyran P, Iorga BI, Durán RV, Collin P. mTOR Inhibition via Displacement of Phosphatidic Acid Induces Enhanced Cytotoxicity Specifically in Cancer Cells. Cancer Res 2018; 78:5384-5397. [PMID: 30054335 DOI: 10.1158/0008-5472.can-18-0232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/09/2018] [Accepted: 07/23/2018] [Indexed: 11/16/2022]
Abstract
The mTOR is a central regulator of cell growth and is highly activated in cancer cells to allow rapid tumor growth. The use of mTOR inhibitors as anticancer therapy has been approved for some types of tumors, albeit with modest results. We recently reported the synthesis of ICSN3250, a halitulin analogue with enhanced cytotoxicity. We report here that ICSN3250 is a specific mTOR inhibitor that operates through a mechanism distinct from those described for previous mTOR inhibitors. ICSN3250 competed with and displaced phosphatidic acid from the FRB domain in mTOR, thus preventing mTOR activation and leading to cytotoxicity. Docking and molecular dynamics simulations evidenced not only the high conformational plasticity of the FRB domain, but also the specific interactions of both ICSN3250 and phosphatidic acid with the FRB domain in mTOR. Furthermore, ICSN3250 toxicity was shown to act specifically in cancer cells, as noncancer cells showed up to 100-fold less sensitivity to ICSN3250, in contrast to other mTOR inhibitors that did not show selectivity. Thus, our results define ICSN3250 as a new class of mTOR inhibitors that specifically targets cancer cells.Significance: ICSN3250 defines a new class of mTORC1 inhibitors that displaces phosphatidic acid at the FRB domain of mTOR, inducing cell death specifically in cancer cells but not in noncancer cells. Cancer Res; 78(18); 5384-97. ©2018 AACR.
Collapse
Affiliation(s)
- Tra-Ly Nguyen
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Marie-Julie Nokin
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.,Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Liège, Belgium
| | - Maxim Egorov
- ATLANTHERA, Cedex, France.,Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Mercedes Tomé
- Laboratoire de l'Angiogénèse et du Microenvironnement des Cancers, INSERM U1029, Université de Bordeaux, Allée Geoffroy Saint Hilaire, Bâtiment, Pessac, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Carmelo Di Primo
- Université de Bordeaux, Laboratoire ARNA, Bordeaux, France; INSERM U1212, CNRS UMR 5320, Institut Européen de Chimie et Biologie, CNRS UMS3033/INSERMUS001, Pessac, France
| | - Lætitia Minder
- Université de Bordeaux, CNRS UMS3033/INSERM US001, Institut Européen de Chimie et Biologie, Pessac, France
| | | | | | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Odile Thoison
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Bernard Delpech
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Georgiana Surpateanu
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Yves-Michel Frapart
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fabienne Peyrot
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Ecole Supérieure du Professorat et de l'Education de l'Académie de Paris, Sorbonne Université, Paris, France
| | - Kahina Abbas
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Silvia Terés
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Serge Evrard
- Institut Bergonié, Digestive Tumours Unit, Université de Bordeaux, Bordeaux, France
| | - Abdel-Majid Khatib
- Laboratoire de l'Angiogénèse et du Microenvironnement des Cancers, INSERM U1029, Université de Bordeaux, Allée Geoffroy Saint Hilaire, Bâtiment, Pessac, France
| | - Pierre Soubeyran
- Institut Bergonié, INSERM U1218, Université de Bordeaux, Bordeaux, France
| | - Bogdan I Iorga
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France.
| | - Raúl V Durán
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.
| | - Pascal Collin
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France.,Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Diderot, UFR Odontologie, Paris, France
| |
Collapse
|
100
|
Abstract
Target of rapamycin (TOR) is an evolutionarily conserved protein kinase that plays a central role in both plants and animals, despite their distinct developmental programs and survival strategies. Indeed, TOR integrates nutrient, energy, hormone, growth factor and environmental inputs to control proliferation, growth and metabolism in diverse multicellular organisms. Here, we compare the molecular composition, upstream regulators and downstream signaling relays of TOR complexes in plants and animals. We also explore and discuss the pivotal functions of TOR signaling in basic cellular processes, such as translation, cell division and stem/progenitor cell regulation during plant development.
Collapse
Affiliation(s)
- Lin Shi
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | - Yue Wu
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| | - Jen Sheen
- Department of Molecular Biology and Centre for Computational and Integrative Biology, Massachusetts General Hospital, and Department of Genetics, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|