201
|
Wee P, Wang Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers (Basel) 2017; 9:cancers9050052. [PMID: 28513565 PMCID: PMC5447962 DOI: 10.3390/cancers9050052] [Citation(s) in RCA: 1211] [Impact Index Per Article: 151.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is commonly upregulated in cancers such as in non-small-cell lung cancer, metastatic colorectal cancer, glioblastoma, head and neck cancer, pancreatic cancer, and breast cancer. Various mechanisms mediate the upregulation of EGFR activity, including common mutations and truncations to its extracellular domain, such as in the EGFRvIII truncations, as well as to its kinase domain, such as the L858R and T790M mutations, or the exon 19 truncation. These EGFR aberrations over-activate downstream pro-oncogenic signaling pathways, including the RAS-RAF-MEK-ERK MAPK and AKT-PI3K-mTOR pathways. These pathways then activate many biological outputs that are beneficial to cancer cell proliferation, including their chronic initiation and progression through the cell cycle. Here, we review the molecular mechanisms that regulate EGFR signal transduction, including the EGFR structure and its mutations, ligand binding and EGFR dimerization, as well as the signaling pathways that lead to G1 cell cycle progression. We focus on the induction of CYCLIN D expression, CDK4/6 activation, and the repression of cyclin-dependent kinase inhibitor proteins (CDKi) by EGFR signaling pathways. We also discuss the successes and challenges of EGFR-targeted therapies, and the potential for their use in combination with CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Ping Wee
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Zhixiang Wang
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
202
|
Liu L, Liao JZ, He XX, Li PY. The role of autophagy in hepatocellular carcinoma: friend or foe. Oncotarget 2017; 8:57707-57722. [PMID: 28915706 PMCID: PMC5593678 DOI: 10.18632/oncotarget.17202] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved lysosome-dependent catabolic process which degrades cell’s components in order to recycle substrates to exert optimally and adapt to tough circumstances. It is a critical cellular homeostatic mechanism with stress resistance, immunity, antiaging, and pro-tumor or anti-tumor effects. Among these, the role of autophagy in cancer is the most eye-catching that is not immutable but dynamic and highly complex. Basal autophagy acts as a tumor suppressor by maintaining genomic stability in normal cells. However, once a tumor is established, unbalanced autophagy will contribute to carcinoma cell survival under tumor microenvironment and in turn promote tumor growth and development. The dynamic role of autophagy can also apply on hepatocellular carcinoma (HCC). HCC is a highly malignant cancer with high morbidity and poor survival rate. Decline or overexpression of autophagic essential genes such as ATG7, ATG5 or Beclin 1 plays a key role in the occurrence and development of HCC but the exact mechanisms are still highly controversial. Signaling pathways or molecules involving in autophagy, for example PI3K/AKT/mTOR pathway, ERK/MAPK pathway, PERK pathway, p53, LncRNA PTENP1 (Long non-coding RNA PTENP1), microRNA-375 and so on, occupy an important position in the complex role of autophagy in HCC. Here, we discuss the dynamic role, the signaling pathways and the potential prognostic and therapy value of autophagy in HCC.
Collapse
Affiliation(s)
- Lian Liu
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Zhi Liao
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-Xing He
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Yuan Li
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
203
|
Abstract
INTRODUCTION Dishevelled, Egl-10 and Pleckstrin (DEP) domain-containing protein 5 (DEPDC5) is a protein subunit of the GTPase-activating proteins towards Rags 1 (GATOR1) complex. GATOR1 is a recently identified modulator of mechanistic target of rapamycin (mTOR) activity. mTOR is a key regulator of cell proliferation and metabolism; disruption of the mTOR pathway is implicated in focal epilepsy, both acquired and genetic. Tuberous sclerosis is the prototypic mTOR genetic syndrome with epilepsy, however GATOR1 gene mutations have recently been shown to cause lesional and non-lesional focal epilepsy. Areas covered: This review summarizes the mTOR pathway, including regulators and downstream effectors, emphasizing recent developments in the understanding of the complex role of the GATOR1 complex. We review the epilepsy types associated with mTOR overactivity, including tuberous sclerosis, polyhydramnios megalencephaly symptomatic epilepsy, cortical dysplasia, non-lesional focal epilepsy and post-traumatic epilepsy. Currently available mTOR inhibitors are discussed, primarily rapamycin analogs and ATP competitive mTOR inhibitors. Expert opinion: DEPDC5 is an attractive therapeutic target in focal epilepsy, as effects of DEPDC5 agonists would likely be anti-epileptogenic and more selective than currently available mTOR inhibitors. Therapeutic effects might be synergistic with certain existing dietary therapies, including the ketogenic diet.
Collapse
Affiliation(s)
- Kenneth A Myers
- a Epilepsy Research Centre, Department of Medicine , The University of Melbourne, Austin Health , Heidelberg , Victoria , Australia.,b Department of Paediatrics , Royal Children's Hospital, The University of Melbourne , Flemington , Victoria , Australia
| | - Ingrid E Scheffer
- a Epilepsy Research Centre, Department of Medicine , The University of Melbourne, Austin Health , Heidelberg , Victoria , Australia.,b Department of Paediatrics , Royal Children's Hospital, The University of Melbourne , Flemington , Victoria , Australia.,c The Florey Institute of Neuroscience and Mental Health , Heidelberg , Victoria , Australia
| |
Collapse
|
204
|
Moderate lifelong overexpression of tuberous sclerosis complex 1 (TSC1) improves health and survival in mice. Sci Rep 2017; 7:834. [PMID: 28400571 PMCID: PMC5429778 DOI: 10.1038/s41598-017-00970-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The tuberous sclerosis complex 1/2 (TSC1/2) is an endogenous regulator of the mechanistic target of rapamycin (mTOR). While mTOR has been shown to play an important role in health and aging, the role of TSC1/2 in aging has not been fully investigated. In the current study, a constitutive TSC1 transgenic (Tsc1tg) mouse model was generated and characterized. mTORC1 signaling was reduced in majority of the tissues, except the brain. In contrast, mTORC2 signaling was enhanced in Tsc1tg mice. Tsc1tg mice are more tolerant to exhaustive exercises and less susceptible to isoproterenol-induced cardiac hypertrophy at both young and advanced ages. Tsc1tg mice have less fibrosis and inflammation in aged as well as isoproterenol-challenged heart than age-matched wild type mice. The female Tsc1tg mice exhibit a higher fat to lean mass ratio at advanced ages than age-matched wild type mice. More importantly, the lifespan increased significantly in female Tsc1tg mice, but not in male Tsc1tg mice. Collectively, our data demonstrated that moderate increase of TSC1 expression can enhance overall health, particularly cardiovascular health, and improve survival in a gender-specific manner.
Collapse
|
205
|
Li D, Zhao K, Yang X, Xiao X, Tang S. TCS2 Increases Olaquindox-Induced Apoptosis by Upregulation of ROS Production and Downregulation of Autophagy in HEK293 Cells. Molecules 2017; 22:E595. [PMID: 28387735 PMCID: PMC6154664 DOI: 10.3390/molecules22040595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 11/16/2022] Open
Abstract
Olaquindox, a feed additive, has drawn public attention due to its potential mutagenicity, genotoxicity, hepatoxicity and nephrotoxicity. The purpose of this study was to investigate the role of tuberous sclerosis complex (TSC2) pathways in olaquindox-induced autophagy in human embryonic kidney 293 (HEK293) cells. The results revealed that olaquindox treatment reduced the cell viability of HEK293 cells and downregulated the expression of TSC2 in a dose- and time-dependent manner. Meanwhile, olaquindox treatment markedly induced the production of reactive oxygen species (ROS), cascaded to autophagy, oxidative stress, and apoptotic cell death, which was effectively eliminated by the antioxidant N-acetylcysteine (NAC). Furthermore, overexpression of TSC2 attenuated olaquindox-induced autophagy in contrast to inducing the production of ROS, oxidative stress and apoptosis. Consistently, knockdown of TSC2 upregulated autophagy, and decreased olaquindox-induced cell apoptosis. In conclusion, our findings indicate that TSC2 partly participates in olaquindox-induced autophagy, oxidative stress and apoptosis, and demonstrate that TSC2 has a negative regulation role in olaquindox-induced autophagy in HEK293 cells.
Collapse
Affiliation(s)
- Daowen Li
- College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China.
| | - Kena Zhao
- College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China.
| | - Xiayun Yang
- College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China.
| | - Xilong Xiao
- College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China.
| | - Shusheng Tang
- College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China.
| |
Collapse
|
206
|
Jacobs BL, McNally RM, Kim KJ, Blanco R, Privett RE, You JS, Hornberger TA. Identification of mechanically regulated phosphorylation sites on tuberin (TSC2) that control mechanistic target of rapamycin (mTOR) signaling. J Biol Chem 2017; 292:6987-6997. [PMID: 28289099 PMCID: PMC5409467 DOI: 10.1074/jbc.m117.777805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/08/2017] [Indexed: 12/31/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) signaling is necessary to generate a mechanically induced increase in skeletal muscle mass, but the mechanism(s) through which mechanical stimuli regulate mTOR signaling remain poorly defined. Recent studies have suggested that Ras homologue enriched in brain (Rheb), a direct activator of mTOR, and its inhibitor, the GTPase-activating protein tuberin (TSC2), may play a role in this pathway. To address this possibility, we generated inducible and skeletal muscle-specific knock-out mice for Rheb (iRhebKO) and TSC2 (iTSC2KO) and mechanically stimulated muscles from these mice with eccentric contractions (EC). As expected, the knock-out of TSC2 led to an elevation in the basal level of mTOR signaling. Moreover, we found that the magnitude of the EC-induced activation of mTOR signaling was significantly blunted in muscles from both inducible and skeletal muscle-specific knock-out mice for Rheb and iTSC2KO mice. Using mass spectrometry, we identified six sites on TSC2 whose phosphorylation was significantly altered by the EC treatment. Employing a transient transfection-based approach to rescue TSC2 function in muscles of the iTSC2KO mice, we demonstrated that these phosphorylation sites are required for the role that TSC2 plays in the EC-induced activation of mTOR signaling. Importantly, however, these phosphorylation sites were not required for an insulin-induced activation of mTOR signaling. As such, our results not only establish a critical role for Rheb and TSC2 in the mechanical activation of mTOR signaling, but they also expose the existence of a previously unknown branch of signaling events that can regulate the TSC2/mTOR pathway.
Collapse
Affiliation(s)
- Brittany L Jacobs
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel M McNally
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Kook-Joo Kim
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rocky Blanco
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel E Privett
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Jae-Sung You
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Troy A Hornberger
- From the Department of Comparative Biosciences and .,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| |
Collapse
|
207
|
Mitchell R, Barton S, Harvey AS, Williams K. Risk factors for the development of autism spectrum disorder in children with tuberous sclerosis complex: protocol for a systematic review. Syst Rev 2017; 6:49. [PMID: 28270230 PMCID: PMC5341363 DOI: 10.1186/s13643-017-0448-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is an autosomal dominant condition, caused by mutations in either the TSC1 or TSC2 gene. It has widespread systemic manifestations and is associated with significant neurological morbidity. In addition to seizures and cerebral pathology including cortical tubers, subependymal nodules, subependymal giant cell astrocytoma and abnormal white matter, there are recognised neuropsychiatric difficulties including intellectual disability, autism spectrum disorder (ASD) and a range of learning and behaviour problems, recently conceptualised as "tuberous sclerosis-associated neuropsychiatric disorders", or "TAND". ASD in TSC is of particular importance because (1) it affects up to 50% of people with TSC and is a source of considerable difficulty for them and their families and (2) it provides a model for considering neurobiological pathways involved in ASD. Multiple factors are implicated in the development of ASD in TSC, including (1) seizures and related electrophysiological factors, (2) cerebral pathology, (3) genotype and (4) child characteristics. However, the neurobiological pathway remains unclear. We will conduct a systematic review to investigate and synthesise existing evidence about the role of these risk factors, individually and in combination, in leading to the development of ASD. METHODS Our review will report on all studies that include one or more of four predefined risk factors in the development of ASD in children with TSC. We will search five databases: MEDLINE, EMBASE, PubMed, The Cochrane Library and Web of Science (Conference Proceedings Citation Index). Studies will be selected for reporting after two authors independently (1) review all titles and abstracts, (2) read full text of all appropriate papers and (3) assess for bias using the Newcastle-Ottawa Scale recommended by the Guidelines for Meta-Analysis and Systematic Reviews of Observational Studies (MOOSE guidelines) and the ROBINS-I. DISCUSSION To our knowledge, this is the first systematic review investigating multiple risk factors in the development of ASD in children with TSC. Clarifying the evidence in this area will be important to researchers in the field and to clinicians providing prognostic information to families. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42016042841.
Collapse
Affiliation(s)
- Rebecca Mitchell
- Developmental Medicine, The Royal Children's Hospital, 50 Flemington Road, Parkville, Victoria, 3052, Australia.
| | - Sarah Barton
- Department of Neurology, The Royal Children's Hospital, 50 Flemington Road, Parkville, Victoria, 3052, Australia
| | - A Simon Harvey
- Department of Neurology, The Royal Children's Hospital, 50 Flemington Road, Parkville, Victoria, 3052, Australia
| | - Katrina Williams
- Developmental Medicine, The Royal Children's Hospital, 50 Flemington Road, Parkville, Victoria, 3052, Australia
| |
Collapse
|
208
|
Lien EC, Lyssiotis CA, Cantley LC. Metabolic Reprogramming by the PI3K-Akt-mTOR Pathway in Cancer. Recent Results Cancer Res 2017; 207:39-72. [PMID: 27557534 DOI: 10.1007/978-3-319-42118-6_3] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the past decade, there has been a resurgence of interest in elucidating how metabolism is altered in cancer cells and how such dependencies can be targeted for therapeutic gain. At the core of this research is the concept that metabolic pathways are reprogrammed in cancer cells to divert nutrients toward anabolic processes to facilitate enhanced growth and proliferation. Importantly, physiological cellular signaling mechanisms normally tightly regulate the ability of cells to gain access to and utilize nutrients, posing a fundamental barrier to transformation. This barrier is often overcome by aberrations in cellular signaling that drive tumor pathogenesis by enabling cancer cells to make critical cellular decisions in a cell-autonomous manner. One of the most frequently altered pathways in human cancer is the PI3K-Akt-mTOR signaling pathway. Here, we describe mechanisms by which this signaling network is responsible for controlling cellular metabolism. Through both the post-translational regulation and the induction of transcriptional programs, the PI3K-Akt-mTOR pathway coordinates the uptake and utilization of multiple nutrients, including glucose, glutamine, nucleotides, and lipids, in a manner best suited for supporting the enhanced growth and proliferation of cancer cells. These regulatory mechanisms illustrate how metabolic changes in cancer are closely intertwined with oncogenic signaling pathways that drive tumor initiation and progression.
Collapse
Affiliation(s)
- Evan C Lien
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, EC/CLS-628C, Boston, MA, 02215, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, 1150 E. Medical Center Drive, Room 6308, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, 1150 E. Medical Center Drive, Room 6308, Ann Arbor, MI, 48109, USA
| | - Lewis C Cantley
- Department of Medicine, the Cancer Center, Weill Cornell Medical College, The Belfer Research Building, 413 East 69th Street, Floor 13 Room BB-1362, New York, NY, 10021, USA.
| |
Collapse
|
209
|
Glomerulocystic kidney presenting as a unilateral kidney mass in a newborn with tuberous sclerosis: Report of a case and review of the literature. Pathol Res Pract 2017; 213:286-291. [DOI: 10.1016/j.prp.2016.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/11/2016] [Indexed: 11/19/2022]
|
210
|
Palmieri M, Pal R, Nelvagal HR, Lotfi P, Stinnett GR, Seymour ML, Chaudhury A, Bajaj L, Bondar VV, Bremner L, Saleem U, Tse DY, Sanagasetti D, Wu SM, Neilson JR, Pereira FA, Pautler RG, Rodney GG, Cooper JD, Sardiello M. mTORC1-independent TFEB activation via Akt inhibition promotes cellular clearance in neurodegenerative storage diseases. Nat Commun 2017; 8:14338. [PMID: 28165011 PMCID: PMC5303831 DOI: 10.1038/ncomms14338] [Citation(s) in RCA: 302] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Neurodegenerative diseases characterized by aberrant accumulation of undigested cellular components represent unmet medical conditions for which the identification of actionable targets is urgently needed. Here we identify a pharmacologically actionable pathway that controls cellular clearance via Akt modulation of transcription factor EB (TFEB), a master regulator of lysosomal pathways. We show that Akt phosphorylates TFEB at Ser467 and represses TFEB nuclear translocation independently of mechanistic target of rapamycin complex 1 (mTORC1), a known TFEB inhibitor. The autophagy enhancer trehalose activates TFEB by diminishing Akt activity. Administration of trehalose to a mouse model of Batten disease, a prototypical neurodegenerative disease presenting with intralysosomal storage, enhances clearance of proteolipid aggregates, reduces neuropathology and prolongs survival of diseased mice. Pharmacological inhibition of Akt promotes cellular clearance in cells from patients with a variety of lysosomal diseases, thus suggesting broad applicability of this approach. These findings open new perspectives for the clinical translation of TFEB-mediated enhancement of cellular clearance in neurodegenerative storage diseases.
Collapse
Affiliation(s)
- Michela Palmieri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| | - Rituraj Pal
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hemanth R. Nelvagal
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9RT, UK
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| | - Gary R. Stinnett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michelle L. Seymour
- Huffington Center on Aging and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Arindam Chaudhury
- Department of Molecular Physiology and Biophysics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Lakshya Bajaj
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| | - Vitaliy V. Bondar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| | - Laura Bremner
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9RT, UK
| | - Usama Saleem
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9RT, UK
| | - Dennis Y. Tse
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, USA
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Deepthi Sanagasetti
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| | - Samuel M. Wu
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Joel R. Neilson
- Department of Molecular Physiology and Biophysics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Fred A. Pereira
- Huffington Center on Aging and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robia G. Pautler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - George G. Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jonathan D. Cooper
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9RT, UK
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas 77030, USA
| |
Collapse
|
211
|
Calimeri T, Ferreri AJM. m-TOR inhibitors and their potential role in haematological malignancies. Br J Haematol 2017; 177:684-702. [PMID: 28146265 DOI: 10.1111/bjh.14529] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/28/2016] [Indexed: 12/22/2022]
Abstract
It is widely demonstrated that the PI3K-AKT-mTOR signalling is critical in normal myeloid and lymphoid development and function. Thus, it is not strange that this pathway is often deregulated in haematological tumours, providing a strong preclinical rationale for the use of drugs targeting the PI3K-AKT-mTOR axis in haematological malignancies. The main focus of this review is to examine the mammalian target of rapamycin (mTOR, also termed mechanistic target of rapamycin [MTOR]) signalling pathways and to provide a brief overview of rapalogs and second-generation mTOR inhibitors used to target its aberrant activation in cancer treatment. We will also discuss the results obtained with the use of these agents in patients with acute leukaemia, Hodgkin lymphoma, non-Hodgkin lymphomas, multiple myeloma and Waldenström macroglobulinaemia. Ongoing clinical trials in haematological malignancies that are investigating first- and second-generation mTOR inhibitors as single agents and as components of combination regimens are also presented.
Collapse
Affiliation(s)
- Teresa Calimeri
- Unit of Lymphoid Malignancies, Department of Onco-Haematology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Andrés J M Ferreri
- Unit of Lymphoid Malignancies, Department of Onco-Haematology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
212
|
González A, Hall MN. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J 2017; 36:397-408. [PMID: 28096180 DOI: 10.15252/embj.201696010] [Citation(s) in RCA: 537] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 01/13/2023] Open
Abstract
Coordinating cell growth with nutrient availability is critical for cell survival. The evolutionarily conserved TOR (target of rapamycin) controls cell growth in response to nutrients, in particular amino acids. As a central controller of cell growth, mTOR (mammalian TOR) is implicated in several disorders, including cancer, obesity, and diabetes. Here, we review how nutrient availability is sensed and transduced to TOR in budding yeast and mammals. A better understanding of how nutrient availability is transduced to TOR may allow novel strategies in the treatment for mTOR-related diseases.
Collapse
|
213
|
Abstract
Renal anomalies are common birth defects that may manifest as a wide spectrum of anomalies from hydronephrosis (dilation of the renal pelvis and calyces) to renal aplasia (complete absence of the kidney(s)). Aneuploidies and mosaicisms are the most common syndromes associated with CAKUT. Syndromes with single gene and renal developmental defects are less common but have facilitated insight into the mechanism of renal and other organ development. Analysis of underlying genetic mutations with transgenic and mutant mice has also led to advances in our understanding of mechanisms of renal development.
Collapse
|
214
|
mTOR and MAPK: from localized translation control to epilepsy. BMC Neurosci 2016; 17:73. [PMID: 27855659 PMCID: PMC5114760 DOI: 10.1186/s12868-016-0308-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/09/2016] [Indexed: 01/03/2023] Open
Abstract
Background Epilepsy is one of the most common neurological diseases characterized by excessive hyperexcitability of neurons. Molecular mechanisms of epilepsy are diverse and not really understood. All in common is the misregulation of proteins that determine excitability such as potassium and sodium channels as well as GABA receptors; which are all known as biomarkers for epilepsy. Two recently identified key pathways involve the kinases mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinases (MAPK). Interestingly, mRNAs coding for those biomarkers are found to be localized at or near synapses indicating a local misregulation of synthesis and activity. Results Research in the last decade indicates that RNA-binding proteins (RBPs) responsible for mRNA localization, stability and translation mediate local expression control. Among others, they are affected by mTOR and MAPK to guide expression of epileptic factors. These results suggest that mTOR/MAPK act on RBPs to regulate the fate of mRNAs, indicating a misregulation of protein expression at synapses in epilepsy. Conclusion We propose that mTOR and MAPK regulate RBPs, thereby guiding the local expression of their target-mRNAs encoding for markers of epilepsy. Thus, misregulated mTOR/MAPK-RBP interplay may result in excessive local synthesis of ion channels and receptors thereby leading to hyperexcitability. Continuous stimulation of synapses further activates mTOR/MAPK pathway reinforcing their effect on RBP-mediated expression control establishing the basis for epilepsy. Here, we highlight findings showing the tight interplay between mTOR as well as MAPK with RBPs to control expression for epileptic biomarkers.
Collapse
|
215
|
Wada S, Neinast M, Jang C, Ibrahim YH, Lee G, Babu A, Li J, Hoshino A, Rowe GC, Rhee J, Martina JA, Puertollano R, Blenis J, Morley M, Baur JA, Seale P, Arany Z. The tumor suppressor FLCN mediates an alternate mTOR pathway to regulate browning of adipose tissue. Genes Dev 2016; 30:2551-2564. [PMID: 27913603 PMCID: PMC5159669 DOI: 10.1101/gad.287953.116] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022]
Abstract
Noncanonical mechanistic target of rapamycin (mTOR) pathways remain poorly understood. Mutations in the tumor suppressor folliculin (FLCN) cause Birt-Hogg-Dubé syndrome, a hamartomatous disease marked by mitochondria-rich kidney tumors. FLCN functionally interacts with mTOR and is expressed in most tissues, but its role in fat has not been explored. We show here that FLCN regulates adipose tissue browning via mTOR and the transcription factor TFE3. Adipose-specific deletion of FLCN relieves mTOR-dependent cytoplasmic retention of TFE3, leading to direct induction of the PGC-1 transcriptional coactivators, drivers of mitochondrial biogenesis and the browning program. Cytoplasmic retention of TFE3 by mTOR is sensitive to ambient amino acids, is independent of growth factor and tuberous sclerosis complex (TSC) signaling, is driven by RagC/D, and is separable from canonical mTOR signaling to S6K. Codeletion of TFE3 in adipose-specific FLCN knockout animals rescues adipose tissue browning, as does codeletion of PGC-1β. Conversely, inducible expression of PGC-1β in white adipose tissue is sufficient to induce beige fat gene expression in vivo. These data thus unveil a novel FLCN-mTOR-TFE3-PGC-1β pathway-separate from the canonical TSC-mTOR-S6K pathway-that regulates browning of adipose tissue.
Collapse
Affiliation(s)
- Shogo Wada
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael Neinast
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Cholsoon Jang
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Chemistry and Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
| | - Yasir H Ibrahim
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York, 10021, USA
| | - Gina Lee
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York, 10021, USA
| | - Apoorva Babu
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jian Li
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Atsushi Hoshino
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Glenn C Rowe
- Division of Cardiovascular Disease, University of Alabama, Birmingham, Alabama 35294, USA
| | - James Rhee
- Department of Anesthesia and Critical Care, Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - José A Martina
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20814, USA
| | - John Blenis
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York, 10021, USA
| | - Michael Morley
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Joseph A Baur
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Patrick Seale
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Zoltan Arany
- Department of Medicine and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
216
|
Guri Y, Hall MN. mTOR Signaling Confers Resistance to Targeted Cancer Drugs. Trends Cancer 2016; 2:688-697. [PMID: 28741507 DOI: 10.1016/j.trecan.2016.10.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 12/19/2022]
Abstract
Cancer is a complex disease and a leading cause of death worldwide. Extensive research over decades has led to the development of therapies that target cancer-specific signaling pathways. However, the clinical benefits of such drugs are at best transient due to tumors displaying intrinsic or adaptive resistance. The underlying compensatory pathways that allow cancer cells to circumvent a drug blockade are poorly understood. We review here recent studies suggesting that mammalian TOR (mTOR) signaling is a major compensatory pathway conferring resistance to many cancer drugs. mTOR-mediated resistance can be cell-autonomous or non-cell-autonomous. These findings suggest that mTOR signaling should be monitored routinely in tumors and that an mTOR inhibitor should be considered as a co-therapy.
Collapse
Affiliation(s)
- Yakir Guri
- Biozentrum, University of Basel, Basel, Switzerland
| | | |
Collapse
|
217
|
Samueli S, Abraham K, Dressler A, Gröppel G, Mühlebner-Fahrngruber A, Scholl T, Kasprian G, Laccone F, Feucht M. Efficacy and safety of Everolimus in children with TSC - associated epilepsy - Pilot data from an open single-center prospective study. Orphanet J Rare Dis 2016; 11:145. [PMID: 27809914 PMCID: PMC5094073 DOI: 10.1186/s13023-016-0530-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/24/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Epilepsy occurs in up to 90 % of all individuals with tuberous sclerosis complex (TSC). In 67 % disease onset is during childhood. In ≥ 50 % seizures are refractory to currently available treatment options. The mTOR-Inhibitor Everolimus (Votubia®) was approved for the treatment of subependymal giant cell astrocytoma (SEGA) and renal angiomyolipoma (AML) in Europe in 2011. It's anticonvulsive/antiepileptic properties are promising, but evidence is still limited. Study aim was to evaluate the efficacy and safety of Everolimus in children and adolescents with TSC-associated epilepsies. METHODS Inclusion-criteria of this investigator-initiated, single-center, open, prospective study were: 1) the ascertained diagnosis of TSC; 2) age ≤ 18 years; 3) treatment indication for Votubia® according to the European Commission guidelines; 4) drug-resistant TSC-associated epilepsy, 5) prospective continuous follow-up for at least 6 months after treatment initiation and 6) informed consent to participate. Votubia® was orally administered once/day, starting with 4.5 mg/m2 and titrated to achieve blood trough concentrations between 5 and 15 ng/ml. Primary endpoint was the reduction in seizure frequency of ≥ 50 % compared to baseline. RESULTS Fifteen patients (nine male) with a median age of six (range; 1-18) years fulfilled the inclusion criteria. 26 % (4/15) had TSC1, 66 % (10/15) had TSC2 mutations. In one patient no mutation was found. Time of observation after treatment initiation was median 22 (range; 6-50) months. At last observation, 80 % (12/15) of the patients were responders, 58 % of them (7/12) were seizure free. The overall reduction in seizure frequency was 60 % in focal seizures, 80 % in generalized tonic clonic seizures and 87 % in drop attacks. The effect of Everolimus was seen already at low doses, early after treatment initiation. Loss of efficacy over time was not observed. Transient side effects were seen in 93 % (14/15) of the patients. In no case the drug had to be withdrawn. CONCLUSION Everolimus seems to be an effective treatment option not only for SEGA and AML, but also for TSC-related epilepsies. Although there are potential serious side effects, treatment was tolerated well by the majority of patients, provided that patients are under close surveillance of epileptologists who are familiar with immunosuppressive agents.
Collapse
Affiliation(s)
- Sharon Samueli
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus Abraham
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anastasia Dressler
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Gudrun Gröppel
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Theresa Scholl
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Franco Laccone
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Martha Feucht
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
218
|
Abstract
The etiology of most childhood cancer remains largely unknown, but is likely attributable to random or induced genetic aberrations in somatic tissue. However, a subset of children develops cancer in the setting of an underlying inheritable condition involving a germline genetic mutation or chromosomal aberration. The term "neurocutaneous syndrome" encompasses a group of multisystem, hereditary disorders that are associated with skin manifestations as well as central and/or peripheral nervous system lesions of variable severity. This review outlines the central nervous system tumors associated with underlying neurocutaneous disorders, including neurofibromatosis type 1, neurofibromatosis type 2, schwannomatosis, tuberous sclerosis complex, Von Hippel Lindau, and nevoid basal cell carcinoma syndrome. Recognizing the presence of an underlying syndrome is critically important to both optimizing clinical care and treatment as well as genetic counseling and monitoring of these affected patients and their families.
Collapse
Affiliation(s)
- Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital and Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
219
|
Roach ES. Applying the Lessons of Tuberous Sclerosis: The 2015 Hower Award Lecture. Pediatr Neurol 2016; 63:6-22. [PMID: 27543366 DOI: 10.1016/j.pediatrneurol.2016.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Tuberous sclerosis complex is a dominantly inherited disorder that variably affects the brain, skin, kidneys, heart, and other organs. Its neurological manifestations include epilepsy, autism, cognitive and behavioral dysfunction, and giant cell tumors. A mutation of either TSC1 or TSC2 can cause tuberous sclerosis complex. Their two gene products, hamartin and tuberin, form a physical complex which normally inhibits protein synthesis mediated through the mechanistic target of rapamycin, so a TSC1 or TSC2 mutation results in overactivation of the mechanistic target of rapamycin cascade. In addition to their tumor suppressor roles, TSC1 and TSC2 help to regulate cell size, neuronal migration, axon formation, and synaptic plasticity. Clinical trials of two different the mechanistic target of rapamycin inhibitors have demonstrated substantial improvement of tuberous sclerosis complex-related tumors, and a recent trial also showed a benefit from the mechanistic target of rapamycin inhibitor everolimus in the treatment of refractory epilepsy due to tuberous sclerosis complex. Effective mechanism-based therapy is now available for some manifestations of tuberous sclerosis complex.
Collapse
Affiliation(s)
- E Steve Roach
- Division of Child Neurology, Ohio State University College of Medicine, Nationwide Children's Hospital, Columbus, Ohio.
| |
Collapse
|
220
|
Semba RD, Trehan I, Gonzalez-Freire M, Kraemer K, Moaddel R, Ordiz MI, Ferrucci L, Manary MJ. Perspective: The Potential Role of Essential Amino Acids and the Mechanistic Target of Rapamycin Complex 1 (mTORC1) Pathway in the Pathogenesis of Child Stunting. Adv Nutr 2016; 7:853-65. [PMID: 27633102 PMCID: PMC5015042 DOI: 10.3945/an.116.013276] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Stunting is the best summary measure of chronic malnutrition in children. Approximately one-quarter of children under age 5 worldwide are stunted. Lipid-based or micronutrient supplementation has little to no impact in reducing stunting, which suggests that other critical dietary nutrients are missing. A dietary pattern of poor-quality protein is associated with stunting. Stunted children have significantly lower circulating essential amino acids than do nonstunted children. Inadequate dietary intakes of essential amino acids could adversely affect growth, because amino acids are required for synthesis of proteins. The master growth regulation pathway, the mechanistic target of rapamycin complex 1 (mTORC1) pathway, is exquisitely sensitive to amino acid availability. mTORC1 integrates cues such as nutrients, growth factors, oxygen, and energy to regulate growth of bone, skeletal muscle, nervous system, gastrointestinal tract, hematopoietic cells, immune effector cells, organ size, and whole-body energy balance. mTORC1 represses protein and lipid synthesis and cell and organismal growth when amino acids are deficient. Over the past 4 decades, the main paradigm for child nutrition in developing countries has been micronutrient malnutrition, with relatively less attention paid to protein. In this Perspective, we present the view that essential amino acids and the mTORC1 pathway play a key role in child growth. The current assumption that total dietary protein intake is adequate for growth among most children in developing countries needs re-evaluation.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD;
| | - Indi Trehan
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | - Klaus Kraemer
- Sight and Life, Basel, Switzerland; and Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | - M Isabel Ordiz
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | | - Mark J Manary
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
221
|
Liu R, Kenney JW, Manousopoulou A, Johnston HE, Kamei M, Woelk CH, Xie J, Schwarzer M, Garbis SD, Proud CG. Quantitative Non-canonical Amino Acid Tagging (QuaNCAT) Proteomics Identifies Distinct Patterns of Protein Synthesis Rapidly Induced by Hypertrophic Agents in Cardiomyocytes, Revealing New Aspects of Metabolic Remodeling. Mol Cell Proteomics 2016; 15:3170-3189. [PMID: 27512079 PMCID: PMC5054342 DOI: 10.1074/mcp.m115.054312] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 01/16/2023] Open
Abstract
Cardiomyocytes undergo growth and remodeling in response to specific pathological or physiological conditions. In the former, myocardial growth is a risk factor for cardiac failure and faster protein synthesis is a major factor driving cardiomyocyte growth. Our goal was to quantify the rapid effects of different pro-hypertrophic stimuli on the synthesis of specific proteins in ARVC and to determine whether such effects are caused by alterations on mRNA abundance or the translation of specific mRNAs. Cardiomyocytes have very low rates of protein synthesis, posing a challenging problem in terms of studying changes in the synthesis of specific proteins, which also applies to other nondividing primary cells. To study the rates of accumulation of specific proteins in these cells, we developed an optimized version of the Quantitative Noncanonical Amino acid Tagging LC/MS proteomic method to label and selectively enrich newly synthesized proteins in these primary cells while eliminating the suppressive effects of pre-existing and highly abundant nonisotope-tagged polypeptides. Our data revealed that a classical pathologic (phenylephrine; PE) and the recently identified insulin stimulus that also contributes to the development of pathological cardiac hypertrophy (insulin), both increased the synthesis of proteins involved in, e.g. glycolysis, the Krebs cycle and beta-oxidation, and sarcomeric components. However, insulin increased synthesis of many metabolic enzymes to a greater extent than PE. Using a novel validation method, we confirmed that synthesis of selected candidates is indeed up-regulated by PE and insulin. Synthesis of all proteins studied was up-regulated by signaling through mammalian target of rapamycin complex 1 without changes in their mRNA levels, showing the key importance of translational control in the rapid effects of hypertrophic stimuli. Expression of PKM2 was up-regulated in rat hearts following TAC. This isoform possesses specific regulatory properties, so this finding indicates it may be involved in metabolic remodeling and also serve as a novel candidate biomarker. Levels of translation factor eEF1 also increased during TAC, likely contributing to faster cell mass accumulation. Interestingly those two candidates were not up-regulated in pregnancy or exercise induced CH, indicating PKM2 and eEF1 were pathological CH specific markers. We anticipate that the methodologies described here will be valuable for other researchers studying protein synthesis in primary cells.
Collapse
Affiliation(s)
- Rui Liu
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Justin W Kenney
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Antigoni Manousopoulou
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Harvey E Johnston
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Makoto Kamei
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Christopher H Woelk
- ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jianling Xie
- §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Michael Schwarzer
- **Department of Cardiovascular Surgery, Jena University Hospital-Friedrich Schiller University of Jena, Erlanger Allee 101, 07747 Jena, Germany
| | - Spiros D Garbis
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; ¶Clinical and Experimental Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; ‖Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK;
| | - Christopher G Proud
- From the ‡Center for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom; §South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA5005, Australia
| |
Collapse
|
222
|
Abbosh PH, McConkey DJ, Plimack ER. Targeting Signaling Transduction Pathways in Bladder Cancer. Curr Oncol Rep 2016; 17:58. [PMID: 26472299 DOI: 10.1007/s11912-015-0477-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Systemic therapy for urothelial carcinoma (UC) of the bladder has largely revolved around cytotoxic chemotherapy regimens. However, several recent clinical trials have explored the roles of targeted therapies which specifically inhibit signal transduction pathways. Simultaneously, a rationale for such therapies has come to the forefront of management of this disease because an overabundance of signaling pathways are genetically deranged as a result of point mutation or copy number alteration (CNA) as identified by several recent next generation sequencing (NGS) studies. Importantly, these derangements are found in all stages of disease, and therefore targeted therapies hold promise as a next step in the evolution of the medical management of both localized and metastatic UCC. We review the rationale for and progress in studying inhibition of signal transduction as a means of treatment of UCC.
Collapse
Affiliation(s)
- Phillip H Abbosh
- Department of Surgical Oncology, Division of Urologic Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA.
| | - David J McConkey
- Departments of Urology and Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,The University of Texas-Graduate School of Biomedical Sciences (GSBS) at Houston, Houston, TX, 77030, USA.
| | - Elizabeth R Plimack
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA.
| |
Collapse
|
223
|
Multi-compartment microscopic diffusion imaging. Neuroimage 2016; 139:346-359. [PMID: 27282476 PMCID: PMC5517363 DOI: 10.1016/j.neuroimage.2016.06.002] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/30/2016] [Accepted: 06/02/2016] [Indexed: 12/03/2022] Open
Abstract
This paper introduces a multi-compartment model for microscopic diffusion anisotropy imaging. The aim is to estimate microscopic features specific to the intra- and extra-neurite compartments in nervous tissue unconfounded by the effects of fibre crossings and orientation dispersion, which are ubiquitous in the brain. The proposed MRI method is based on the Spherical Mean Technique (SMT), which factors out the neurite orientation distribution and thus provides direct estimates of the microscopic tissue structure. This technique can be immediately used in the clinic for the assessment of various neurological conditions, as it requires only a widely available off-the-shelf sequence with two b-shells and high-angular gradient resolution achievable within clinically feasible scan times. To demonstrate the developed method, we use high-quality diffusion data acquired with a bespoke scanner system from the Human Connectome Project. This study establishes the normative values of the new biomarkers for a large cohort of healthy young adults, which may then support clinical diagnostics in patients. Moreover, we show that the microscopic diffusion indices offer direct sensitivity to pathological tissue alterations, exemplified in a preclinical animal model of Tuberous Sclerosis Complex (TSC), a genetic multi-organ disorder which impacts brain microstructure and hence may lead to neurological manifestations such as autism, epilepsy and developmental delay.
Collapse
|
224
|
Abstract
As a key regulator of cell metabolism and survival, mechanistic target of rapamycin (mTOR) emerges as a novel therapeutic target for Parkinson's disease (PD). A growing body of research indicates that restoring perturbed mTOR signaling in PD models can prevent neuronal cell death. Nevertheless, molecular mechanisms underlying mTOR-mediated effects in PD have not been fully understood yet. Here, we review recent progress in characterizing the association of mTOR signaling with PD risk factors and further discuss the potential roles of mTOR in PD.
Collapse
|
225
|
Clinical response to everolimus in a patient with Hodgkin's lymphoma harboring a TSC2 mutation. Blood Cancer J 2016; 6:e420. [PMID: 27176796 PMCID: PMC4916294 DOI: 10.1038/bcj.2016.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
226
|
Abstract
The movement toward precision medicine with targeted therapeutics for cancer treatment has been hindered by both innate and acquired resistance. Understanding the molecular wiring and plasticity of oncogenic signaling networks is essential to the development of therapeutic strategies to avoid or overcome resistance. The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) represents a highly integrated signaling node that is dysregulated in the majority of human cancers. Several studies have revealed that sustained mTORC1 inhibition is essential to avoid resistance to targeted therapeutics against the driving oncogenic pathway in a given cancer. Here we discuss the role of mTORC1 in dictating the response of tumors to targeted therapeutics and review recent examples from lung cancer, breast cancer, and melanoma.
Collapse
Affiliation(s)
- Erika Ilagan
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
227
|
Moavero R, Folgiero V, Carai A, Miele E, Ferretti E, Po A, Diomedi Camassei F, Lepri FR, Vigevano F, Curatolo P, Valeriani M, Colafati GS, Locatelli F, Tornesello A, Mastronuzzi A. Metastatic Group 3 Medulloblastoma in a Patient With Tuberous Sclerosis Complex: Case Description and Molecular Characterization of the Tumor. Pediatr Blood Cancer 2016; 63:719-22. [PMID: 26626406 DOI: 10.1002/pbc.25851] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 01/23/2023]
Abstract
Medulloblastoma is the most common pediatric brain tumor. We describe a child with tuberous sclerosis complex that developed a Group 3, myc overexpressed, metastatic medulloblastoma (MB). Considering the high risk of treatment-induced malignancies, a tailored therapy, omitting radiation, was given. Based on the evidence of mammalian target of rapamycin mTORC, mTOR Complex; RAS, Rat sarcoma; RAF, rapidly accelerated fibrosarcoma (mTOR) pathway activation in the tumor, targeted therapy was applied resulting in complete remission of disease. Although the PI3K/AKT/mTOR signaling pathway plays a role in MB, we did not find TSC1/TSC2 (TSC, tuberous sclerosis complex) mutation in our patient. We speculate that a different pathway resulting in mTOR activation is the basis of both TSC and MB in this child; H&E, haematoxilin and eosin; Gd, gadolinium.
Collapse
Affiliation(s)
- Romina Moavero
- Neuroscience and Neurorehabilitation Department, Neurology Unit, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy.,Systems Medicine Department, Child Neurology and Psychiatry Unit, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Valentina Folgiero
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Andrea Carai
- Department of Neuroscience and Neurorehabilitation, Neurosurgery Unit, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Molecular Medicine, Sapienza University, Rome, Italy.,Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Agnese Po
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | - Francesca Romana Lepri
- Department of Laboratories, Medical Genetics laboratory, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Federico Vigevano
- Neuroscience and Neurorehabilitation Department, Neurology Unit, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Paolo Curatolo
- Systems Medicine Department, Child Neurology and Psychiatry Unit, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Massimiliano Valeriani
- Neuroscience and Neurorehabilitation Department, Neurology Unit, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Giovanna S Colafati
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy.,Department of Pediatric Science, University of Pavia, Italy
| | - Assunta Tornesello
- Department of Pediatrics, "Università Cattolica del Sacro Cuore,", Rome, Italy
| | - Angela Mastronuzzi
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, , IRCCS, Rome, Italy
| |
Collapse
|
228
|
McVey D, Aronov M, Rizzi G, Cowan A, Scott C, Megill J, Russell R, Tirosh B. CHO cells knocked out for TSC2 display an improved productivity of antibodies under fed batch conditions. Biotechnol Bioeng 2016; 113:1942-52. [DOI: 10.1002/bit.25951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Duncan McVey
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Michael Aronov
- Institute for Drug Research; The School of Pharmacy; The Hebrew University of Jerusalem; Jerusalem 91120 Israel
| | - Giovanni Rizzi
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Alexis Cowan
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Charo Scott
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - John Megill
- Discovery Toxicology; Bristol Myers Squibb Company; Pennington New Jersey
| | - Reb Russell
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Boaz Tirosh
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
- Institute for Drug Research; The School of Pharmacy; The Hebrew University of Jerusalem; Jerusalem 91120 Israel
| |
Collapse
|
229
|
Aronov M, Tirosh B. Metabolic Control of Plasma Cell Differentiation- What We Know and What We Don't Know. J Clin Immunol 2016; 36 Suppl 1:12-7. [PMID: 26910101 DOI: 10.1007/s10875-016-0246-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 02/16/2016] [Indexed: 12/21/2022]
Abstract
Antibody secretion is executed by plasma cells that are generated in the periphery and migrate to the bone marrow to establish a long lived pool. The terminal differentiation of B lymphocytes into plasma cells is executed by a network of transcription factors that cross-regulate each other in order to irreversibly promote this transition. While major progress has been made in the understanding the transcriptional activity of the underlying master regulators, much less is known on the metabolic regulation of plasma cell differentiation that is required to support antibody synthesis, folding and secretion at high levels and allow their long-lasting survival. In this review we will address the known cross talks between the transcription and metabolic control of plasma cells and elaborate on the gaps of knowledge in the field.
Collapse
Affiliation(s)
- Michael Aronov
- Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Boaz Tirosh
- Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
230
|
Tee AR, Sampson JR, Pal DK, Bateman JM. The role of mTOR signalling in neurogenesis, insights from tuberous sclerosis complex. Semin Cell Dev Biol 2016; 52:12-20. [PMID: 26849906 DOI: 10.1016/j.semcdb.2016.01.040] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/05/2016] [Accepted: 01/25/2016] [Indexed: 11/30/2022]
Abstract
Understanding the development and function of the nervous system is one of the foremost aims of current biomedical research. The nervous system is generated during a relatively short period of intense neurogenesis that is orchestrated by a number of key molecular signalling pathways. Even subtle defects in the activity of these molecules can have serious repercussions resulting in neurological, neurodevelopmental and neurocognitive problems including epilepsy, intellectual disability and autism. Tuberous sclerosis complex (TSC) is a monogenic disease characterised by these problems and by the formation of benign tumours in multiple organs, including the brain. TSC is caused by mutations in the TSC1 or TSC2 gene leading to activation of the mechanistic target of rapamycin (mTOR) signalling pathway. A desire to understand the neurological manifestations of TSC has stimulated research into the role of the mTOR pathway in neurogenesis. In this review we describe TSC neurobiology and how the use of animal model systems has provided insights into the roles of mTOR signalling in neuronal differentiation and migration. Recent progress in this field has identified novel mTOR pathway components regulating neuronal differentiation. The roles of mTOR signalling and aberrant neurogenesis in epilepsy are also discussed. Continuing efforts to understand mTOR neurobiology will help to identify new therapeutic targets for TSC and other neurological diseases.
Collapse
Affiliation(s)
- Andrew R Tee
- Institute of Cancer & Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff CF14 4XN UK
| | - Julian R Sampson
- Institute of Cancer & Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff CF14 4XN UK
| | - Deb K Pal
- Department of Basic & Clinical Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, King's College, London SE5 8RX UK
| | - Joseph M Bateman
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL UK.
| |
Collapse
|
231
|
Abstract
Type 2 diabetes is a complex disease. It results from a failure of the body to maintain energy homoeostasis. Multicellular organisms have evolved complex strategies to preserve a relatively stable internal nutrient environment, despite fluctuations in external nutrient availability. This complex strategy involves the co-ordinated responses of multiple organs to promote storage or mobilization of energy sources according to the availability of nutrients and cellular bioenergetics needs. The endocrine pancreas plays a central role in these processes by secreting insulin and glucagon. When this co-ordinated effort fails, hyperglycaemia and hyperlipidaemia develops, characterizing a state of metabolic imbalance and ultimately overt diabetes. Although diabetes is most likely a collection of diseases, scientists are starting to identify genetic components and environmental triggers. Genome-wide association studies revealed that by and large, gene variants associated with type 2 diabetes are implicated in pancreatic β-cell function, suggesting that the β-cell may be the weakest link in the chain of events that results in diabetes. Thus, it is critical to understand how environmental cues affect the β-cell. Phosphoinositides are important 'decoders' of environmental cues. As such, these lipids have been implicated in cellular responses to a wide range of growth factors, hormones, stress agents, nutrients and metabolites. Here we will review some of the well-established and potential new roles for phosphoinositides in β-cell function/dysfunction and discuss how our knowledge of phosphoinositide signalling could aid in the identification of potential strategies for treating or preventing type 2 diabetes.
Collapse
Affiliation(s)
- Lucia E Rameh
- Department of Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, U.S.A.
| | - Jude T Deeney
- Department of Medicine, Boston University School of Medicine, 650 Albany Street, Boston, MA 02118, U.S.A
| |
Collapse
|
232
|
Tuberous sclerosis--A model for tumour growth. Semin Cell Dev Biol 2016; 52:3-11. [PMID: 26816112 DOI: 10.1016/j.semcdb.2016.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/18/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder where patients develop benign tumours in several organ systems. Central to TSC pathology is hyper-activation of the mammalian target of rapamycin complex 1 (mTORC1) signalling pathway, which is a key controller of cell growth. As a result, TSC model systems are a valuable tool for examining mTORC1-driven cellular processes. The immunosuppressant, rapamycin, is a specific inhibitor of mTORC1 and has shown promise as a therapeutic agent in TSC as well as in malignancy. This review will focus on the cellular processes controlled by mTORC1 and how TSC-deficient cell lines and mouse models have broadened our understanding of the mTORC1 signalling network. It will also discuss how our knowledge of TSC signalling can help us understand sporadic conditions where mTORC1 activity is implicated in disease onset or progression, and the possibility of using rapamycin to treat sporadic disease.
Collapse
|
233
|
Sadowski K, Kotulska K, Jóźwiak S. Management of side effects of mTOR inhibitors in tuberous sclerosis patients. Pharmacol Rep 2016; 68:536-42. [PMID: 26891243 DOI: 10.1016/j.pharep.2016.01.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 01/20/2023]
Abstract
mTOR inhibitors represent a relatively new therapeutic option in the management of patients affected by tuberous sclerosis complex (TSC). Randomized clinical trials support the use of everolimus in the treatment of subependymal giant cell astrocytomas (SEGA) and renal angiomyolipomas (AML) related to TSC. Accumulating data suggest also systemic disease-modifying potential of mTOR inhibitors. Given that increasing number of patients with TSC receive mTOR inhibitors, the issue of adverse events associated with this therapy becomes practically important. In the present study we provide the overview of clinical manifestations and therapeutic options for the most common adverse events related to mTOR inhibitors in TSC patients.
Collapse
Affiliation(s)
- Krzysztof Sadowski
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warszawa, Poland.
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warszawa, Poland.
| | - Sergiusz Jóźwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warszawa, Poland; Department of Pediatric Neurology, Warsaw Medical University, Warszawa, Poland.
| |
Collapse
|
234
|
The spectrum of nephrocutaneous diseases and associations: Genetic causes of nephrocutaneous disease. J Am Acad Dermatol 2016; 74:231-44; quiz 245-6. [PMID: 26775773 DOI: 10.1016/j.jaad.2015.05.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
There are a significant number of diseases and treatment considerations of considerable importance relating to the skin and renal systems. This emphasizes the need for dermatologists in practice or in clinical training to be aware of these associations. Part I of this 2-part continuing medical education article reviews the genetic syndromes with both renal and cutaneous involvement that are most important for the dermatologist to be able to identify, manage, and appropriately refer to nephrology colleagues. Part II reviews the inflammatory syndromes with relevant renal manifestations and therapeutic agents commonly used by dermatologists that have drug-induced effects on or require close consideration of renal function. In addition, we will likewise review therapeutic agents commonly used by nephrologists that have drug-induced effects on the skin that dermatologists are likely to encounter in clinical practice. In both parts of this continuing medical education article, we discuss diagnosis, management, and appropriate referral to our nephrology colleagues in the context of each nephrocutaneous association. There are a significant number of dermatoses associated with renal abnormalities and disease, emphasizing the need for dermatologists to be keenly aware of their presence in order to avoid overlooking important skin conditions with potentially devastating renal complications. This review discusses important nephrocutaneous disease associations with recommendations for the appropriate urgency of referral to nephrology colleagues for diagnosis, surveillance, and early management of potential renal sequelae.
Collapse
|
235
|
Kazdoba TM, Leach PT, Crawley JN. Behavioral phenotypes of genetic mouse models of autism. GENES, BRAIN, AND BEHAVIOR 2016; 15:7-26. [PMID: 26403076 PMCID: PMC4775274 DOI: 10.1111/gbb.12256] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/27/2015] [Accepted: 09/18/2015] [Indexed: 12/11/2022]
Abstract
More than a hundred de novo single gene mutations and copy-number variants have been implicated in autism, each occurring in a small subset of cases. Mutant mouse models with syntenic mutations offer research tools to gain an understanding of the role of each gene in modulating biological and behavioral phenotypes relevant to autism. Knockout, knockin and transgenic mice incorporating risk gene mutations detected in autism spectrum disorder and comorbid neurodevelopmental disorders are now widely available. At present, autism spectrum disorder is diagnosed solely by behavioral criteria. We developed a constellation of mouse behavioral assays designed to maximize face validity to the types of social deficits and repetitive behaviors that are central to an autism diagnosis. Mouse behavioral assays for associated symptoms of autism, which include cognitive inflexibility, anxiety, hyperactivity, and unusual reactivity to sensory stimuli, are frequently included in the phenotypic analyses. Over the past 10 years, we and many other laboratories around the world have employed these and additional behavioral tests to phenotype a large number of mutant mouse models of autism. In this review, we highlight mouse models with mutations in genes that have been identified as risk genes for autism, which work through synaptic mechanisms and through the mTOR signaling pathway. Robust, replicated autism-relevant behavioral outcomes in a genetic mouse model lend credence to a causal role for specific gene contributions and downstream biological mechanisms in the etiology of autism.
Collapse
Affiliation(s)
- T. M. Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - P. T. Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - J. N. Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
236
|
Abstract
The evolutionarily conserved target of rapamycin complex 1 (TORC1) is a master regulator of cell growth and metabolism. In mammals, growth factors and cellular energy stimulate mTORC1 activity through inhibition of the TSC complex (TSC1-TSC2-TBC1D7), a negative regulator of mTORC1. Amino acids signal to mTORC1 independently of the TSC complex. Here, we review recently identified regulators that link amino acid sufficiency to mTORC1 activity and how mutations affecting these regulators cause human disease.
Collapse
|
237
|
Yang X, Hei C, Liu P, Song Y, Thomas T, Tshimanga S, Wang F, Niu J, Sun T, Li PA. Inhibition of mTOR Pathway by Rapamycin Reduces Brain Damage in Rats Subjected to Transient Forebrain Ischemia. Int J Biol Sci 2015; 11:1424-35. [PMID: 26681922 PMCID: PMC4672000 DOI: 10.7150/ijbs.12930] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/21/2015] [Indexed: 12/19/2022] Open
Abstract
The aims of this study are to clarify the role of mTOR in mediating cerebral ischemic brain damage and the effects of rapamycin on ischemic outcomes. Ten minutes of forebrain ischemia was induced in rats, and their brains were sampled after 3 h, 16 h, and 7 days reperfusion for histology, immunohistochemistry and biochemical analysis. Our data demonstrated that cerebral ischemia resulted in both apoptotic and necrotic neuronal death; cerebral ischemia and reperfusion led to significant increases of mRNA and protein levels of p-mTOR and its downstream p-P70S6K and p-S6; elevation of LC3-II, and release of cytochrome c into the cytoplasm in both the cortex and hippocampus. Inhibition of mTOR by rapamycin markedly reduced ischemia-induced damage; suppressed p-Akt, p-mTOR, p-P70S6K and p-S6 protein levels; decreased LC3-II and Beclin-1; and prevented cytochrome c release in the two structures. All together, these data provide evidence that cerebral ischemia activates mTOR and autophagy pathways. Inhibition of mTOR deactivates the mTOR pathway, suppresses autophagy, prevents cytochrome c release and reduces ischemic brain damage.
Collapse
Affiliation(s)
- Xiao Yang
- 1. Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 75004, China ; 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Changhun Hei
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA ; 3. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - Ping Liu
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA ; 4. Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yaozu Song
- 1. Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 75004, China
| | - Taylor Thomas
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Sylvie Tshimanga
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Feng Wang
- 1. Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 75004, China
| | - Jianguo Niu
- 1. Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 75004, China
| | - Tao Sun
- 1. Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 75004, China
| | - P Andy Li
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
238
|
Detection of low-prevalence somatic TSC2 mutations in sporadic pulmonary lymphangioleiomyomatosis tissues by deep sequencing. Hum Genet 2015; 135:61-8. [PMID: 26563443 DOI: 10.1007/s00439-015-1611-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/06/2015] [Indexed: 01/30/2023]
Abstract
Lymphangioleiomyomatosis (LAM) (MIM #606690) is a rare lung disorder leading to respiratory failure associated with progressive cystic destruction due to the proliferation and infiltration of abnormal smooth muscle-like cells (LAM cells). LAM can occur alone (sporadic LAM, S-LAM) or combined with tuberous sclerosis complex (TSC-LAM). TSC is caused by a germline heterozygous mutation in either TSC1 or TSC2, and TSC-LAM is thought to occur as a result of a somatic mutation (second hit) in addition to a germline mutation in TSC1 or TSC2 (first hit). S-LAM is also thought to occur under the two-hit model involving a somatic mutation and/or loss of heterozygosity in TSC2. To identify TSC1 or TSC2 changes in S-LAM patients, the two genes were analyzed by deep next-generation sequencing (NGS) using genomic DNA from blood leukocytes (n = 9), LAM tissue from lung (n = 7), LAM cultured cells (n = 4), or LAM cell clusters (n = 1). We identified nine somatic mutations in six of nine S-LAM patients (67 %) with mutant allele frequencies of 1.7-46.2 %. Three of these six patients (50 %) showed two different TSC2 mutations with allele frequencies of 1.7-28.7 %. Furthermore, at least five mutations with low prevalence (<20 % of allele frequency) were confirmed by droplet digital PCR. As LAM tissues are likely to be composed of heterogeneous cell populations, mutant allele frequencies can be low. Our results confirm the consistent finding of TSC2 mutations in LAM samples, and highlight the benefit of laser capture microdissection and in-depth allele analyses for detection, such as NGS.
Collapse
|
239
|
Yang Z, Liu F, Qu H, Wang H, Xiao X, Deng H. 1, 25(OH)2D3 protects β cell against high glucose-induced apoptosis through mTOR suppressing. Mol Cell Endocrinol 2015. [PMID: 26213322 DOI: 10.1016/j.mce.2015.07.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Diabetes mellitus is a leading cause of death and disability worldwide, which presents a serious public health crisis in China nowadays. It has been well recognized that excessive β-cell apoptosis is the key pathogenesis of diabetes, of which the mammalian target of rapamycin (mTOR) serves as the critical signaling pathway. Emerging evidence indicates that vitamin D deficiency acts as a potential risk factor for diabetes. The present study aims to test the hypothesis that 1 alpha, 25-dihydroxyvitamin D(3) [1, 25(OH)2D3] can inhibit β-cell apoptosis via the suppression of mTOR signaling pathway. β-cells (INS-1) were cultured in the context of normal glucose or high glucose media with or without 1, 25(OH)2D3 treatment. β-cell apoptosis was evaluated by inverted fluorescence microscope, flow cytometry and electron microscope, respectively. Quantitative RT-PCR and Western blotting were performed to assess the possible perturbations in mTOR signaling pathway. High glucose significantly increased β-cell apoptosis. Of importance, RT-PCR and Western blotting demonstrated that high glucose inhibited DNA-damage-inducible transcript 4 (DDIT4) and TSC1/TSC2, up-regulated Rheb/mTOR/p70S6K and enhanced expression of the apoptosis regulating proteins, such as phospho-Bcl-2, cytochrome C and cleaved caspase. Interestingly, 1, 25(OH)2D3 treatment reversed high glucose induced pathological changes in mTOR signaling pathway, restored expression of DDIT4 and TSC1/TSC2, blocked aberrant up-regulation of Rheb/mTOR/p70S6K and the apoptosis regulating proteins, and effectively inhibited β-cell apoptosis. Therefore, 1, 25(OH)2D3 treatment can effectively protects β cell against high glucose-induced apoptosis mainly via the suppression of mTOR signaling pathway, which may be considered as a potential therapy for patients with diabetes.
Collapse
Affiliation(s)
- Zesong Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Fang Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Hua Qu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Hang Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Xiaoqiu Xiao
- Laboratory of Lipid and Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China
| | - Huacong Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, PR China.
| |
Collapse
|
240
|
Ess KC, Chugani HT. Dynamic tubers in tuberous sclerosis complex: A window for intervention? Neurology 2015; 85:1530-1. [PMID: 26432847 DOI: 10.1212/wnl.0000000000002056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Kevin C Ess
- From the Division of Pediatric Neurology (K.C.E.), Vanderbilt University Medical Center, Nashville, TN; and Division of Pediatric Neurology (H.T.C.), PET Center Children's Hospital of Michigan, Detroit.
| | - Harry T Chugani
- From the Division of Pediatric Neurology (K.C.E.), Vanderbilt University Medical Center, Nashville, TN; and Division of Pediatric Neurology (H.T.C.), PET Center Children's Hospital of Michigan, Detroit
| |
Collapse
|
241
|
Gao Y, Gartenhaus RB, Lapidus RG, Hussain A, Zhang Y, Wang X, Dan HC. Differential IKK/NF-κB Activity Is Mediated by TSC2 through mTORC1 in PTEN-Null Prostate Cancer and Tuberous Sclerosis Complex Tumor Cells. Mol Cancer Res 2015; 13:1602-14. [PMID: 26374334 DOI: 10.1158/1541-7786.mcr-15-0213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/05/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED The serine/threonine protein kinase Akt plays a critical role in regulating proliferation, growth, and survival through phosphorylation of different downstream substrates. The mTOR is a key target for Akt to promote tumorigenesis. It has been reported that Akt activates mTOR through phosphorylation and inhibition of the tuberous sclerosis complex (TSC) protein TSC2. Previously, it was demonstrated that mTOR activates IKK/NF-κB signaling by promoting IκB kinase (IKK) activity downstream of Akt in conditions deficient of PTEN. In this study, the mechanistic role of the tumor-suppressor TSC2 was investigated in the regulation of IKK/NF-κB activity in PTEN-null prostate cancer and in TSC2-mutated tumor cells. The results demonstrate that TSC2 inhibits IKK/NF-κB activity downstream of Akt and upstream of mTORC1 in a PTEN-deficient environment. However, TSC2 promotes IKK/NF-κB activity upstream of Akt and mTORC1 in TSC2 mutated tumor cells. These data indicate that TSC2 negatively or positively regulates IKK/NF-κB activity in a context-dependent manner depending on the genetic background. IMPLICATIONS This study provides fundamental insight for understanding the molecular details by which TSC2/mTOR regulates NF-κB signaling in different tumors.
Collapse
Affiliation(s)
- Yu Gao
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning Province, P.R China
| | - Ronald B Gartenhaus
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rena G Lapidus
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Arif Hussain
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Baltimore VA Medical Center, Baltimore, Maryland
| | - Yanting Zhang
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Han C Dan
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
242
|
Curatolo P, Moavero R, de Vries PJ. Neurological and neuropsychiatric aspects of tuberous sclerosis complex. Lancet Neurol 2015; 14:733-45. [PMID: 26067126 DOI: 10.1016/s1474-4422(15)00069-1] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 01/22/2023]
Abstract
Tuberous sclerosis (also known as tuberous sclerosis complex [TSC]) is a multisystem genetic disorder that affects almost every organ in the body. Mutations in the TSC1 or TSC2 genes lead to disruption of the TSC1-TSC2 intracellular protein complex, causing overactivation of the mammalian target of rapamycin (mTOR) protein complex. The surveillance and management guidelines and clinical criteria for tuberous sclerosis were revised in 2012, and mTOR inhibitors are now recommended as treatment options for subependymal giant cell astrocytomas and renal angiomyolipomas-two common features of the disease. However, most morbidity and mortality caused by tuberous sclerosis is associated with neurological and neuropsychiatric manifestations. Treatment of epilepsy associated with tuberous sclerosis remains a major challenge, with more than 60% of patients having ongoing seizures. Tuberous-sclerosis-associated neuropsychiatric disorders (TAND) are multilevel and occur in most individuals with the disorder, but are rarely assessed and treated. Clinical trials of mTOR inhibitors to treat seizures and TAND are underway. Management of the neurological and neuropsychiatric manifestations of the disorder should be coordinated with treatment of other organ systems. In view of the age-related expression of manifestations from infancy to adulthood, continuity of clinical care and ongoing monitoring is paramount, and particular attention is needed to plan transition of patient care from childhood to adult services.
Collapse
Affiliation(s)
- Paolo Curatolo
- Neuroscience Department, Child Neurology and Psychiatry Division, University Hospital of Tor Vergata, Rome, Italy.
| | - Romina Moavero
- Neuroscience Department, Child Neurology and Psychiatry Division, University Hospital of Tor Vergata, Rome, Italy; Neuroscience Department, Child Neurology Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
243
|
Fouqué A, Delalande O, Jean M, Castellano R, Josselin E, Malleter M, Shoji KF, Hung MD, Rampanarivo H, Collette Y, van de Weghe P, Legembre P. A Novel Covalent mTOR Inhibitor, DHM25, Shows in Vivo Antitumor Activity against Triple-Negative Breast Cancer Cells. J Med Chem 2015; 58:6559-73. [PMID: 26237138 DOI: 10.1021/acs.jmedchem.5b00991] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Constitutive activation of the PI3K/mTOR signaling pathway contributes to carcinogenesis and metastasis in most, if not all, breast cancers. From a chromene backbone reported to inhibit class I PI3K catalytic subunits, several rounds of chemical syntheses led to the generation of a new collection of chromologues that showed enhanced ability to kill PI3K-addicted cancer cells and to inhibit Akt phosphorylation at serine 473, a hallmark of PI3K/mTOR activation. This initial screen uncovered a chromene designated DHM25 that exerted potent antitumor activity against breast tumor cell lines. Strikingly, DHM25 was shown to be a selective and covalent inhibitor of mTOR using biochemical and cellular analyses, modeling, and a large panel of kinase activity assays spanning the human kinome (243 kinases). Finally, in vivo, this novel drug was an efficient inhibitor of growth and metastasis of triple-negative breast cancer cells, paving the way for its clinical application in oncology.
Collapse
Affiliation(s)
- Amélie Fouqué
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,CLCC Eugène Marquis, INSERM ER440 Oncogenesis, Stress & Signaling, rue bataille Flandres Dunkerque, 35042 Rennes, France.,Equipe Labellisée Ligue Contre le Cancer, rue bataille Flandres Dunkerque, Rennes, France
| | - Olivier Delalande
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,IGDR, CNRS UMR 6290, UFR des Sciences Biologiques et Pharmaceutiques, 2 Av. du Professeur Léon Bernard, 35043 Rennes, France
| | - Mickael Jean
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,UMR6226, CNRS, Institut des Sciences Chimiques de Rennes, Equipe Produits Naturels, Synthèses, Chimie Médicinale, UFR des Sciences Biologiques et Pharmaceutiques, 2 Av. du Professeur Léon Bernard, 35043 Rennes, France
| | - Rémy Castellano
- CRCM, INSERM, U1068; Institut Paoli-Calmettes; Aix-Marseille Université; CNRS, UMR 7258, Marseille, France
| | - Emmanuelle Josselin
- CRCM, INSERM, U1068; Institut Paoli-Calmettes; Aix-Marseille Université; CNRS, UMR 7258, Marseille, France
| | - Marine Malleter
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France
| | - Kenji F Shoji
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,INSERM, U1085, 2 avenue du Prof. Leon Bernard, 35043 Rennes, France
| | - Mac Dinh Hung
- Faculty of Chemistry, National Vietnam University , 19 Le Thanh Tong, Hanoi, Vietnam
| | - Hariniaina Rampanarivo
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,CLCC Eugène Marquis, INSERM ER440 Oncogenesis, Stress & Signaling, rue bataille Flandres Dunkerque, 35042 Rennes, France.,Equipe Labellisée Ligue Contre le Cancer, rue bataille Flandres Dunkerque, Rennes, France.,IGDR, CNRS UMR 6290, UFR des Sciences Biologiques et Pharmaceutiques, 2 Av. du Professeur Léon Bernard, 35043 Rennes, France
| | - Yves Collette
- CRCM, INSERM, U1068; Institut Paoli-Calmettes; Aix-Marseille Université; CNRS, UMR 7258, Marseille, France
| | - Pierre van de Weghe
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,UMR6226, CNRS, Institut des Sciences Chimiques de Rennes, Equipe Produits Naturels, Synthèses, Chimie Médicinale, UFR des Sciences Biologiques et Pharmaceutiques, 2 Av. du Professeur Léon Bernard, 35043 Rennes, France
| | - Patrick Legembre
- Université de Rennes-1 , 2 avenue du Prof. Leon Bernard, 35043 Rennes, France.,CLCC Eugène Marquis, INSERM ER440 Oncogenesis, Stress & Signaling, rue bataille Flandres Dunkerque, 35042 Rennes, France.,Equipe Labellisée Ligue Contre le Cancer, rue bataille Flandres Dunkerque, Rennes, France
| |
Collapse
|
244
|
Qiu B, Simon MC. Oncogenes strike a balance between cellular growth and homeostasis. Semin Cell Dev Biol 2015; 43:3-10. [PMID: 26277544 DOI: 10.1016/j.semcdb.2015.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/28/2015] [Accepted: 08/09/2015] [Indexed: 12/28/2022]
Abstract
Altered tumor cell metabolism is now firmly established as a hallmark of human cancer. Downstream of oncogenic events, metabolism is re-wired to support cellular energetics and supply the building blocks for biomass. Rapid, uncontrolled proliferation results in tumor growth beyond the reach of existing vasculature and triggers cellular adaptations to overcome limiting nutrient and oxygen delivery. However, oncogenic activation and metabolic re-programming also elicit cell intrinsic stresses, independent of the tumor microenvironment. To ensure metabolic robustness and stress resistance, pro-growth signals downstream of oncogene activation or tumor suppressor loss simultaneously activate homeostatic processes. Here, we summarize recent literature describing the adaptive mechanisms co-opted by common oncogenes, including mTOR, MYC, and RAS. Recurrent themes in our review include: (1) coordination of oncogene-induced changes in protein and lipid metabolism to sustain endoplasmic reticulum homeostasis, (2) maintenance of mitochondrial functional capacity to support anabolic metabolism, (3) adaptations to sustain intracellular metabolite concentrations required for growth, and (4) prevention of oxidative stress. We also include a discussion of the hypoxia inducible factors (HIFs) and the AMP-dependent protein kinase (AMPK)--stress sensors that are co-opted to support tumor growth. Ultimately, an understanding of the adaptations required downstream of specific oncogenes could reveal targetable metabolic vulnerabilities.
Collapse
Affiliation(s)
- Bo Qiu
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
245
|
Wataya-Kaneda M. Mammalian target of rapamycin and tuberous sclerosis complex. J Dermatol Sci 2015; 79:93-100. [DOI: 10.1016/j.jdermsci.2015.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/16/2015] [Indexed: 12/25/2022]
|
246
|
Dutchak PA, Laxman S, Estill SJ, Wang C, Wang Y, Wang Y, Bulut GB, Gao J, Huang LJ, Tu BP. Regulation of Hematopoiesis and Methionine Homeostasis by mTORC1 Inhibitor NPRL2. Cell Rep 2015; 12:371-9. [PMID: 26166573 DOI: 10.1016/j.celrep.2015.06.042] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/08/2015] [Accepted: 06/08/2015] [Indexed: 01/23/2023] Open
Abstract
Nitrogen permease regulator-like 2 (NPRL2) is a component of a conserved complex that inhibits mTORC1 (mammalian Target Of Rapamycin Complex 1) in response to amino acid insufficiency. Here, we show that NPRL2 is required for mouse viability and that its absence significantly compromises fetal liver hematopoiesis in developing embryos. Moreover, NPRL2 KO embryos have significantly reduced methionine levels and exhibit phenotypes reminiscent of cobalamin (vitamin B12) deficiency. Consistent with this idea, NPRL2 KO liver and mouse embryonic fibroblasts (MEFs) show defective processing of the cobalamin-transport protein transcobalamin 2, along with impaired lysosomal acidification and lysosomal gene expression. NPRL2 KO MEFs exhibit a significant defect in the cobalamin-dependent synthesis of methionine from homocysteine, which can be rescued by supplementation with cyanocobalamin. Taken together, these findings demonstrate a role for NPRL2 and mTORC1 in the regulation of lysosomal-dependent cobalamin processing, methionine synthesis, and maintenance of cellular re-methylation potential, which are important during hematopoiesis.
Collapse
Affiliation(s)
- Paul A Dutchak
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA
| | - Sunil Laxman
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA
| | - Sandi Jo Estill
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA
| | - Chensu Wang
- Simmons Comprehensive Cancer Center and Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-8807, USA
| | - Yun Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA
| | - Yiguang Wang
- Simmons Comprehensive Cancer Center and Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-8807, USA
| | - Gamze B Bulut
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9039, USA
| | - Jinming Gao
- Simmons Comprehensive Cancer Center and Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390-8807, USA
| | - Lily J Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-9039, USA
| | - Benjamin P Tu
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA.
| |
Collapse
|
247
|
Dibble CC, Cantley LC. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol 2015; 25:545-55. [PMID: 26159692 DOI: 10.1016/j.tcb.2015.06.002] [Citation(s) in RCA: 607] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 06/08/2015] [Accepted: 06/08/2015] [Indexed: 11/29/2022]
Abstract
The class I phosphoinositide 3-kinase (PI3K)-mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) signaling network directs cellular metabolism and growth. Activation of mTORC1 [composed of mTOR, regulatory-associated protein of mTOR (Raptor), mammalian lethal with SEC13 protein 8(mLST8), 40-kDa proline-rich Akt substrate (PRAS40), and DEP domain-containing mTOR-interacting protein (DEPTOR)] depends on the Ras-related GTPases (Rags) and Ras homolog enriched in brain (Rheb) GTPase and requires signals from amino acids, glucose, oxygen, energy (ATP), and growth factors (including cytokines and hormones such as insulin). Here we discuss the signal transduction mechanisms through which growth factor-responsive PI3K signaling activates mTORC1. We focus on how PI3K-dependent activation of Akt and spatial regulation of the tuberous sclerosis complex (TSC) complex (TSC complex) [composed of TSC1, TSC2, and Tre2-Bub2-Cdc16-1 domain family member 7 (TBC1D7)] switches on Rheb at the lysosome, where mTORC1 is activated. Integration of PI3K- and amino acid-dependent signals upstream of mTORC1 at the lysosome is detailed in a working model. A coherent understanding of the PI3K-mTORC1 network is imperative as its dysregulation has been implicated in diverse pathologies including cancer, diabetes, autism, and aging.
Collapse
Affiliation(s)
- Christian C Dibble
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
248
|
Gamsiz ED, Sciarra LN, Maguire AM, Pescosolido MF, van Dyck LI, Morrow EM. Discovery of Rare Mutations in Autism: Elucidating Neurodevelopmental Mechanisms. Neurotherapeutics 2015; 12:553-71. [PMID: 26105128 PMCID: PMC4489950 DOI: 10.1007/s13311-015-0363-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of highly genetic neurodevelopmental disorders characterized by language, social, cognitive, and behavioral abnormalities. ASD is a complex disorder with a heterogeneous etiology. The genetic architecture of autism is such that a variety of different rare mutations have been discovered, including rare monogenic conditions that involve autistic symptoms. Also, de novo copy number variants and single nucleotide variants contribute to disease susceptibility. Finally, autosomal recessive loci are contributing to our understanding of inherited factors. We will review the progress that the field has made in the discovery of these rare genetic variants in autism. We argue that mutation discovery of this sort offers an important opportunity to identify neurodevelopmental mechanisms in disease. The hope is that these mechanisms will show some degree of convergence that may be amenable to treatment intervention.
Collapse
Affiliation(s)
- Ece D. Gamsiz
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
- />Developmental Disorders Genetics Research Program, Emma Pendleton Bradley Hospital and Department of Psychiatry and Human Behavior, Brown University Medical School, Providence, RI USA
| | - Laura N. Sciarra
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
- />Neuroscience Graduate Program (NSGP), Brown University, Providence, RI USA
| | - Abbie M. Maguire
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
- />Molecular Biology, Cell Biology and Biochemistry (MCB) Graduate Training Program, Brown University, Providence, RI USA
| | - Matthew F. Pescosolido
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
- />Neuroscience Graduate Program (NSGP), Brown University, Providence, RI USA
| | - Laura I. van Dyck
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
| | - Eric M. Morrow
- />Department of Molecular Biology, Cell Biology and Biochemistry (MCB), and Institute for Brain Science, Brown University, Providence, RI USA
- />Developmental Disorders Genetics Research Program, Emma Pendleton Bradley Hospital and Department of Psychiatry and Human Behavior, Brown University Medical School, Providence, RI USA
| |
Collapse
|
249
|
Ray RM, Bavaria M, Johnson LR. Interaction of polyamines and mTOR signaling in the synthesis of antizyme (AZ). Cell Signal 2015; 27:1850-9. [PMID: 26093026 DOI: 10.1016/j.cellsig.2015.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/11/2015] [Accepted: 06/11/2015] [Indexed: 01/01/2023]
Abstract
Tissue polyamine levels are largely determined by the activity of ornithine decarboxylase (ODC, EC 4.1.17), which catalyzes the conversion of ornithine to the diamine putrescine. The activity of the enzyme is primarily regulated by a negative feedback mechanism involving ODC antizyme (AZ). Our previous studies demonstrated that AZ synthesis is stimulated by the absence of amino acids, the levels of which are sensed by the mTOR complex containing TORC1, which is stimulated by amino acids and inhibited by their absence, and TORC2 the function of which is not well defined. Polyamines, which cause a +1 ribosomal frameshift during the translation of AZ mRNA are required to increase AZ synthesis in both the presence and absence of amino acids. Amino acid starvation increases TORC2 activity. We have demonstrated that mTORC2 activity is necessary for AZ synthesis in the absence of amino acids. Tuberous sclerosis protein (TSC), a negative regulator of mTOR function regulates the activities of both the TORC1 and TORC2. TSC2 knockdown increased mTORC1 activity with concomitant inhibition of mTORC2 activity eliminating AZ induction in the absence of amino acids as well as that induced by spermidine. Thus, these results clearly demonstrate that in addition to polyamines, mTORC2 activity is necessary for AZ synthesis. Moreover, our results support a role for mTORC2 in the synthesis of a specific protein, AZ, which regulates growth of intestinal epithelial cells.
Collapse
Affiliation(s)
- Ramesh M Ray
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Mitul Bavaria
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Leonard R Johnson
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
250
|
Ishii R, Wataya-Kaneda M, Canuet L, Nonomura N, Nakai Y, Takeda M. Everolimus improves behavioral deficits in a patient with autism associated with tuberous sclerosis: a case report. ACTA ACUST UNITED AC 2015. [DOI: 10.1186/s40810-015-0004-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|