251
|
Casar Tena T, Burkhalter MD, Philipp M. Left-right asymmetry in the light of TOR: An update on what we know so far. Biol Cell 2015; 107:306-18. [PMID: 25943139 PMCID: PMC4744706 DOI: 10.1111/boc.201400094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/29/2015] [Indexed: 01/06/2023]
Abstract
The internal left‐right (LR) asymmetry is a characteristic that exists throughout the animal kingdom from roundworms over flies and fish to mammals. Cilia, which are antenna‐like structures protruding into the extracellular space, are involved in establishing LR asymmetry during early development. Humans who suffer from dysfunctional cilia often develop conditions such as heterotaxy, where internal organs appear to be placed randomly. As a consequence to this failure in asymmetry development, serious complications such as congenital heart defects (CHD) occur. The mammalian (or mechanistic) target of rapamycin (mTOR) pathway has recently emerged as an important regulator regarding symmetry breaking. The mTOR pathway governs fundamental processes such as protein translation or metabolism. Its activity can be transduced by two complexes, which are called TORC1 and TORC2, respectively. So far, only TORC1 has been implicated with asymmetry development and appears to require very precise regulation. A number of recent papers provided evidence that dysregulated TORC1 results in alterations of motile cilia and asymmetry defects. In here, we give an update on what we know so far of mTORC1 in LR asymmetry development.
Collapse
Affiliation(s)
- Teresa Casar Tena
- Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| | - Martin D Burkhalter
- Leibniz Institute for Age Research Fritz Lippmann Institute, Jena, 07745, Germany
| | - Melanie Philipp
- Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| |
Collapse
|
252
|
Abstract
Tuberous sclerosis complex is an autosomal-dominant, neurocutaneous, multisystem disorder characterized by cellular hyperplasia and tissue dysplasia. The genetic cause is mutations in the TSC1 gene, found on chromosome 9q34, and TSC2 gene, found on chromosome 16p13. The clinical phenotypes resulting from mutations in either of the 2 genes are variable in each individual. Herein, advances in the understanding of molecular mechanisms in tuberous sclerosis complex are reviewed, and current guidelines for diagnosis, treatment, follow-up, and management are summarized.
Collapse
Affiliation(s)
- Francis J DiMario
- Department of Pediatrics, Neurogenetics-Tuberous Sclerosis Clinic, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06070, USA.
| | - Mustafa Sahin
- Multidisciplinary Tuberous Sclerosis Program, Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
253
|
Reduction in retinal nerve fiber layer thickness in tuberous sclerosis complex. Childs Nerv Syst 2015; 31:857-61. [PMID: 25910752 DOI: 10.1007/s00381-015-2702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 04/05/2015] [Indexed: 01/26/2023]
Abstract
PURPOSE The aim of our study was to non-invasively investigate central nervous system axonal integrity in patients with tuberous sclerosis complex (TSC). Diffuse microstructural white matter abnormalities reflecting axonal disorganization, reduced/altered myelination, or gliosis have been described in individuals with TSC. Optical coherence tomography (OCT) is a fast, easy-to-perform, non-invasive, and cost-efficient method to assess retinal morphology in vivo and to measure the thickness of the retinal nerve fiber layer (RNFL). METHODS In order to assess central nervous system axonal integrity, eight subjects with TSC have been investigated by OCT to evaluate RNFL and they have been compared with matched healthy controls. RESULTS When comparing mean overall RNFL thicknesses of the TSC group with those of the control group, the TSC group presented with significantly lower RNFL values, compared to the control group, in the temporal quadrant (62.5 ± 6.9 vs. 76.9 ± 5.4; t = 14.438; p < 0.0001). CONCLUSIONS Since a reduced RNFL thickness might be seen as an indicator of chronic axonal degeneration or lack of appropriate neuronal development, our results support the presence of axonal alterations in TSC and also that white matter disorganization could be much more diffuse than originally thought. Since axonal alterations directly derive from mammalian target of rapamycin (mTOR) overactivation, which occurs early during fetus development, the RNFL thinning we observed could represent one of the facets of such early neurodevelopmental abnormalities.
Collapse
|
254
|
Chantranupong L, Wolfson RL, Sabatini DM. Nutrient-sensing mechanisms across evolution. Cell 2015; 161:67-83. [PMID: 25815986 DOI: 10.1016/j.cell.2015.02.041] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 12/11/2022]
Abstract
For organisms to coordinate their growth and development with nutrient availability, they must be able to sense nutrient levels in their environment. Here, we review select nutrient-sensing mechanisms in a few diverse organisms. We discuss how these mechanisms reflect the nutrient requirements of specific species and how they have adapted to the emergence of multicellularity in eukaryotes.
Collapse
Affiliation(s)
- Lynne Chantranupong
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, 9 Cambridge Center, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Rachel L Wolfson
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, 9 Cambridge Center, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, 9 Cambridge Center, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
255
|
Ci X, Kuraoka M, Wang H, Carico Z, Hopper K, Shin J, Deng X, Qiu Y, Unniraman S, Kelsoe G, Zhong XP. TSC1 Promotes B Cell Maturation but Is Dispensable for Germinal Center Formation. PLoS One 2015; 10:e0127527. [PMID: 26000908 PMCID: PMC4441391 DOI: 10.1371/journal.pone.0127527] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 04/16/2015] [Indexed: 01/10/2023] Open
Abstract
Accumulating evidence indicates that the tuberous sclerosis complex 1 (TSC1), a tumor suppressor that acts by inhibiting mTOR signaling, plays an important role in the immune system. We report here that TSC1 differentially regulates mTOR complex 1 (mTORC1) and mTORC2/Akt signaling in B cells. TSC1 deficiency results in the accumulation of transitional-1 (T1) B cells and progressive losses of B cells as they mature beyond the T1 stage. Moreover, TSC1KO mice exhibit a mild defect in the serum antibody responses or rate of Ig class-switch recombination after immunization with a T-cell-dependent antigen. In contrast to a previous report, we demonstrate that both constitutive Peyer’s patch germinal centers (GCs) and immunization-induced splenic GCs are unimpaired in TSC1-deficient (TSC1KO) mice and that the ratio of GC B cells to total B cells is comparable in WT and TSC1KO mice. Together, our data demonstrate that TSC1 plays important roles for B cell development, but it is dispensable for GC formation and serum antibody responses.
Collapse
Affiliation(s)
- Xinxin Ci
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710, United States of America
- Key Laboratory of Zoonosis Ministry of Education, Institute of Zoonosis, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Masayuki Kuraoka
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
| | - Hongxia Wang
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710, United States of America
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zachary Carico
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
| | - Kristen Hopper
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
| | - Jinwook Shin
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710, United States of America
| | - Xuming Deng
- Key Laboratory of Zoonosis Ministry of Education, Institute of Zoonosis, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yirong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shyam Unniraman
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
| | - Garnett Kelsoe
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
- * E-mail: (XPZ); (GK)
| | - Xiao-Ping Zhong
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710, United States of America
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America
- * E-mail: (XPZ); (GK)
| |
Collapse
|
256
|
Kimura T, Kitaura H, Masuda H, Kameyama S, Saito Y, Sugai K, Otsuki T, Nakazawa A, Morota N, Yamamoto T, Iida K, Nakagawa M, Mizuno T, Takahashi H, Kakita A. Characteristic expression of p57/Kip2 in balloon cells in focal cortical dysplasia. Neuropathology 2015; 35:401-9. [PMID: 25950494 DOI: 10.1111/neup.12199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 11/29/2022]
Abstract
Balloon cells are a pathognomonic cellular feature of various cortical malformations, including focal cortical dysplasia type IIb (FCD IIb), cortical tubers of tuberous sclerosis (TSC) and hemimegalencephaly (HME). In the present study, we investigated the immunohistochemical expression of p57/Kip2, a member of the Cip/Kip family of cyclin-dependent kinase inhibitory proteins, in balloon cells in surgical specimens taken from 26, 17 and six patients with FCD IIb, TSC and HME, respectively. Characteristic dot-like reactivity with a faint, intense, reticular and process-like pattern was confined to the proximal portion of the cytoplasmic processes of the cells. Immunoelectron microscopy revealed the p57/Kip2 reactivity on intermediate filaments in the proximal portion of the processes. The immunohistochemical profile appeared similar to that of CD34; however, a double immunofluorescence study demonstrated that no cells showed reactivity for both p57/Kip2 and CD34. The frequencies of the p57/Kip2-positive cells in FCD IIb and HME were significantly higher than those in TSC, suggesting that the balloon cells may be heterogeneous. These findings suggest some functional significance of the protein on the cytoplasmic processes of balloon cells and appear consistent with the notion that the cells are abnormally differentiated progenitor cells.
Collapse
Affiliation(s)
- Tadashi Kimura
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan.,Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Kitaura
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Hiroshi Masuda
- Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Shigeki Kameyama
- Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Yuko Saito
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kenji Sugai
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Taisuke Otsuki
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Atsuko Nakazawa
- National Medical Center for Children and Mothers, National Center for Child Health and Development, Tokyo, Japan
| | - Nobuhito Morota
- National Medical Center for Children and Mothers, National Center for Child Health and Development, Tokyo, Japan
| | - Takamichi Yamamoto
- Comprehensive Epilepsy Center, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Kouji Iida
- Department of Neurosurgery, University of Hiroshima, Hiroshima, Japan
| | - Masanori Nakagawa
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| |
Collapse
|
257
|
Pollizzi KN, Patel CH, Sun IH, Oh MH, Waickman AT, Wen J, Delgoffe GM, Powell JD. mTORC1 and mTORC2 selectively regulate CD8⁺ T cell differentiation. J Clin Invest 2015; 125:2090-108. [PMID: 25893604 DOI: 10.1172/jci77746] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 03/12/2015] [Indexed: 12/16/2022] Open
Abstract
Activation of mTOR-dependent pathways regulates the specification and differentiation of CD4+ T effector cell subsets. Herein, we show that mTOR complex 1 (mTORC1) and mTORC2 have distinct roles in the generation of CD8+ T cell effector and memory populations. Evaluation of mice with a T cell-specific deletion of the gene encoding the negative regulator of mTORC1, tuberous sclerosis complex 2 (TSC2), resulted in the generation of highly glycolytic and potent effector CD8+ T cells; however, due to constitutive mTORC1 activation, these cells retained a terminally differentiated effector phenotype and were incapable of transitioning into a memory state. In contrast, CD8+ T cells deficient in mTORC1 activity due to loss of RAS homolog enriched in brain (RHEB) failed to differentiate into effector cells but retained memory characteristics, such as surface marker expression, a lower metabolic rate, and increased longevity. However, these RHEB-deficient memory-like T cells failed to generate recall responses as the result of metabolic defects. While mTORC1 influenced CD8+ T cell effector responses, mTORC2 activity regulated CD8+ T cell memory. mTORC2 inhibition resulted in metabolic reprogramming, which enhanced the generation of CD8+ memory cells. Overall, these results define specific roles for mTORC1 and mTORC2 that link metabolism and CD8+ T cell effector and memory generation and suggest that these functions have the potential to be targeted for enhancing vaccine efficacy and antitumor immunity.
Collapse
|
258
|
Sahin K, Cross B, Sahin N, Ciccone K, Suleiman S, Osunkoya AO, Master V, Harris W, Carthon B, Mohammad R, Bilir B, Wertz K, Moreno CS, Walker CL, Kucuk O. Lycopene in the prevention of renal cell cancer in the TSC2 mutant Eker rat model. Arch Biochem Biophys 2015; 572:36-39. [PMID: 25602702 PMCID: PMC5657428 DOI: 10.1016/j.abb.2015.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/03/2015] [Accepted: 01/08/2015] [Indexed: 01/05/2023]
Abstract
Renal cell carcinoma (RCC) is the most frequent upper urinary tract cancer in humans and accounts for 80-85% of malignant renal tumors. Eker rat represents a unique animal model to study RCC since these rats develop spontaneous renal tumors and leiomyoma, which may be due to tuberous sclerosis 2 (TSC2) mutation resulting in the activation of the mammalian target of rapamycin (mTOR) pathway. This study examines the role of a lycopene-rich diet in the development of RCC in the TSC2 mutant Eker rat model. Ten-week old female Eker rats (n=90) were assigned in equal numbers to receive 0, 100 or 200mg/kg of lycopene as part of their daily diet. After 18 months the rats were sacrificed and the kidneys were removed. Immunohistochemical staining with antibodies against mTOR, phospho-S6 and EGFR were performed, as well as hematoxylin-eosin staining for histologic examination of the tumors. Tumors were counted and measured in individual kidneys. Presence of tumor decreased from 94% in control animals to 65% in the experimental group, but the difference was not statistically significant (P<0.12). However, mean numbers of renal carcinomas were statistically significantly decreased in the lycopene-treated rats (P<0.008) when compared to untreated controls. In the lycopene group, tumor numbers decreased (P<0.002) and the numbers tended to decrease linearly (P<0.003) as supplemental lycopene increased from 0 to 200. Control rats fed only basal diet had a greater length of tumors (23.98 mm) than rats fed lycopene supplement groups (12.90 mm and 11.07 mm) (P<0.05). Moreover tumor length decreased (P<0.02) and tumor length tended to decrease linearly (P<0.03) as supplemental lycopene increased from 0 to 200mg/kg. All tumors showed strong staining with antibodies against mTOR, phospho-S6 and EGFR. In conclusion, dietary supplementation with lycopene attenuates the development of renal cell cancers in the predisposed TSC2 mutant Eker rat model. These results suggest that lycopene may play a role in the prevention of RCC.
Collapse
Affiliation(s)
- Kazim Sahin
- Department of Animal Nutrition, Veterinary Faculty, Firat University, Elazig, Turkey
| | - Brian Cross
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Nurhan Sahin
- Department of Animal Nutrition, Veterinary Faculty, Firat University, Elazig, Turkey
| | - Karina Ciccone
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Shadeah Suleiman
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | | | - Viraj Master
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Wayne Harris
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Bradley Carthon
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Ramzi Mohammad
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Birdal Bilir
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | | | - Carlos S Moreno
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Cheryl L Walker
- Molecular Carcinogenesis, Science Park - Research Division, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Omer Kucuk
- Winship Cancer Institute, Emory University, Atlanta, GA, United States.
| |
Collapse
|
259
|
Samueli S, Abraham K, Dressler A, Groeppel G, Jonak C, Muehlebner A, Prayer D, Reitner A, Feucht M. Tuberous Sclerosis Complex: new criteria for diagnostic work-up and management. Wien Klin Wochenschr 2015; 127:619-30. [PMID: 25860851 DOI: 10.1007/s00508-015-0758-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/27/2015] [Indexed: 12/24/2022]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic multisystem disorder, characterized by predominantly benign tumors in potentially all organ systems. System involvement, severity of clinical symptoms and the response to treatment are age-dependent and heterogeneous. Consequently, the disorder is still not recognized in a considerable number of patients. The diagnostic criteria and the guidelines for surveillance and management of patients with TSC were revised, and the establishment of specialized TSC-centers was strongly recommended during an International Consensus Conference in 2012. TOSCA (TuberOus SClerosis registry to increase disease Awareness), an international patient registry, was started to allow new insights into the causes of different courses. Finally, there are-since the approval of the mTOR inhibitor Everolimus-promising new therapeutic approaches.This review focuses on the various TSC related symptoms occurring at different ages, the novel recommendations for diagnosis and treatment as well as the need for multidisciplinary follow-up.
Collapse
Affiliation(s)
- Sharon Samueli
- Universitätsklinik für Kinder- und Jugendheilkunde, AKH Wien, Wien, Österreich
| | | | | | | | | | | | | | | | | | | |
Collapse
|
260
|
Abstract
PURPOSE OF REVIEW Tuberous sclerosis complex (TSC) is a multisystem genetic disorder with physical and neuropsychiatric manifestations and significant research progress has been made in recent years. Here, we focus on the key advances over the last 18 months. RECENT FINDINGS Three main themes were identified in the literature. Firstly, the diagnostic criteria and surveillance guidelines for TSC were revised, incorporating a genetic criterion alongside clinical criteria, and making a positive step towards evidence-based treatment of TSC. Secondly, a new term - TSC-associated neuropsychiatric disorders (TAND) - was introduced as an umbrella term for all possible neuropsychiatric difficulties seen in TSC, and a TAND Checklist was developed as a screening tool. Thirdly, the risks and benefits of molecularly targeted treatments of the neuropsychiatric manifestations of TSC are being debated. SUMMARY The updated diagnostic criteria and management guidelines, the new concept of TAND and the TAND Checklist should lead to significant improvements in the quality of care for individuals with TSC. The promise of mammalian target of rapamycin inhibitors and other molecular treatments are still to be confirmed. We suggest that great care should be taken to identify 'optimal mammalian target of rapamycin signalling' in the therapeutic approach to the neuropsychiatric features of the disorder.
Collapse
|
261
|
Thien A, Prentzell MT, Holzwarth B, Kläsener K, Kuper I, Boehlke C, Sonntag AG, Ruf S, Maerz L, Nitschke R, Grellscheid SN, Reth M, Walz G, Baumeister R, Neumann-Haefelin E, Thedieck K. TSC1 activates TGF-β-Smad2/3 signaling in growth arrest and epithelial-to-mesenchymal transition. Dev Cell 2015; 32:617-30. [PMID: 25727005 DOI: 10.1016/j.devcel.2015.01.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 12/19/2014] [Accepted: 01/22/2015] [Indexed: 11/27/2022]
Abstract
The tuberous sclerosis proteins TSC1 and TSC2 are key integrators of growth factor signaling. They suppress cell growth and proliferation by acting in a heteromeric complex to inhibit the mammalian target of rapamycin complex 1 (mTORC1). In this study, we identify TSC1 as a component of the transforming growth factor β (TGF-β)-Smad2/3 pathway. Here, TSC1 functions independently of TSC2. TSC1 interacts with the TGF-β receptor complex and Smad2/3 and is required for their association with one another. TSC1 regulates TGF-β-induced Smad2/3 phosphorylation and target gene expression and controls TGF-β-induced growth arrest and epithelial-to-mesenchymal transition (EMT). Hyperactive Akt specifically activates TSC1-dependent cytostatic Smad signaling to induce growth arrest. Thus, TSC1 couples Akt activity to TGF-β-Smad2/3 signaling. This has implications for cancer treatments targeting phosphoinositide 3-kinases and Akt because they may impair tumor-suppressive cytostatic TGF-β signaling by inhibiting Akt- and TSC1-dependent Smad activation.
Collapse
Affiliation(s)
- Antje Thien
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Mirja Tamara Prentzell
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands
| | - Birgit Holzwarth
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Kathrin Kläsener
- Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ineke Kuper
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | | | - Annika G Sonntag
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Stefanie Ruf
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Lars Maerz
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Roland Nitschke
- BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | | | - Michael Reth
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Molecular Immunology, Max-Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Gerd Walz
- Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | - Ralf Baumeister
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Research Training Group (RTG) 1104, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), Albert-Ludwigs-University Freiburg, 79106 Freiburg, Germany
| | | | - Kathrin Thedieck
- Bioinformatics and Molecular Genetics (Faculty of Biology), Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands; BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany.
| |
Collapse
|
262
|
Zhou Y, Rychahou P, Wang Q, Weiss HL, Evers BM. TSC2/mTORC1 signaling controls Paneth and goblet cell differentiation in the intestinal epithelium. Cell Death Dis 2015; 6:e1631. [PMID: 25654764 PMCID: PMC4669793 DOI: 10.1038/cddis.2014.588] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 12/18/2022]
Abstract
The intestinal mucosa undergoes a continual process of proliferation, differentiation and apoptosis, which is regulated by multiple signaling pathways. Notch signaling is critical for the control of intestinal stem cell maintenance and differentiation. However, the precise mechanisms involved in the regulation of differentiation are not fully understood. Previously, we have shown that tuberous sclerosis 2 (TSC2) positively regulates the expression of the goblet cell differentiation marker, MUC2, in intestinal cells. Using transgenic mice constitutively expressing a dominant negative TSC2 allele, we observed that TSC2 inactivation increased mTORC1 and Notch activities, and altered differentiation throughout the intestinal epithelium, with a marked decrease in the goblet and Paneth cell lineages. Conversely, treatment of mice with either Notch inhibitor dibenzazepine (DBZ) or mTORC1 inhibitor rapamycin significantly attenuated the reduction of goblet and Paneth cells. Accordingly, knockdown of TSC2 activated, whereas knockdown of mTOR or treatment with rapamycin decreased, the activity of Notch signaling in the intestinal cell line LS174T. Importantly, our findings demonstrate that TSC2/mTORC1 signaling contributes to the maintenance of intestinal epithelium homeostasis by regulating Notch activity.
Collapse
Affiliation(s)
- Y Zhou
- Markey Cancer Center, The University of Kentucky, Lexington, KY, USA
| | - P Rychahou
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| | - Q Wang
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| | - H L Weiss
- Markey Cancer Center, The University of Kentucky, Lexington, KY, USA
| | - B M Evers
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| |
Collapse
|
263
|
Anti-EGFR antibody reduces lung nodules by inhibition of EGFR-pathway in a model of lymphangioleiomyomatosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:315240. [PMID: 25699271 PMCID: PMC4324894 DOI: 10.1155/2015/315240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/26/2014] [Indexed: 11/17/2022]
Abstract
EGFR belongs to the HER/ErbB family of tyrosine kinase receptors and its activation in cancer cells has been linked with increased proliferation, angiogenesis, and metastasis. Lymphangioleiomyomatosis (LAM) is a rare, low-grade neoplasm that occurs sporadically or in association with tuberous sclerosis complex (TSC), a genetic, multisystem disorder characterized by hamartomas in several organs. From chylous of a LAM/TSC patient, we previously isolated smooth muscle-like LAM/TSC cells whose proliferation depends on EGF and monoclonal anti-EGFR antibodies reduced proliferation and caused cell death. We demonstrated that the dependency from EGF was caused by the absence of tuberin. To study the role of EGFR pathway in vivo, we developed a mouse model by administration of LAM/TSC cells to female nude mice. LAM/TSC cells caused pulmonary airspace enlargement and, after 30 weeks, nodule formation which express EGFR. Anti-EGFR antibody decreased the number and dimension of lung nodules likely for the inhibition of Erk and S6 signaling, reversed the pulmonary alterations, and reduced lymphatic and blood vessels. Moreover, in pulmonary nodules anti-EGFR antibody reduced the positivity to estrogen and progesterone receptors which enhance survival of LAM cells and Snail expression. These results suggest that the inhibition of EGFR signalling has a potential in treatment of LAM/TSC lung alterations.
Collapse
|
264
|
Abstract
Tuberous sclerosis complex (TSC) is a neurocutaneous syndrome that can affect the brain, skin, eyes, kidneys, heart, and lungs. TSC alters cellular proliferation and differentiation, resulting in hamartomas of various organs, tumor formation, and altered neuronal migration. The phenotype is highly variable. Most individuals have seizures, commonly including infantile spasms, and there is variable intellectual disability and autism. Neonates can present with cardiac failure due to intracardiac rhabdomyomas. The likelihood of renal angiomyolipomas increases with age, and renal disease is the most common cause of death in adults with TSC. Pulmonary involvement occurs predominantly in women and carries a high morbidity and mortality. TSC is inherited as an autosomal dominant trait, but spontaneous mutations are common. A mutation of either TSC1 on chromosome 9 or TSC2 on chromosome 16 leads to dysfunction of hamartin or tuberin, respectively. These two proteins form a functional complex that modulates the mammalian target of rapamycin (mTOR) pathway. Medications that inhibit mTOR are being used to treat TSC-related tumors, and current studies are investigating whether these agents could alleviate other TSC complications. Consensus statements guide identification and optimal management of many of the TSC-related complications at diagnosis and throughout the lifespan. A multidisciplinary approach is necessary for optimal management of individuals with TSC.
Collapse
Affiliation(s)
- Monica P Islam
- Department of Child Neurology, Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, OH, USA.
| | - E Steve Roach
- Department of Child Neurology, Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
265
|
Wang Z, Liu S, Kakizaki M, Hirose Y, Ishikawa Y, Funato H, Yanagisawa M, Yu Y, Liu Q. Orexin/hypocretin activates mTOR complex 1 (mTORC1) via an Erk/Akt-independent and calcium-stimulated lysosome v-ATPase pathway. J Biol Chem 2014; 289:31950-31959. [PMID: 25278019 PMCID: PMC4231673 DOI: 10.1074/jbc.m114.600015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/30/2014] [Indexed: 02/05/2023] Open
Abstract
The lack of the neuropeptide orexin, also known as hypocretin, results in narcolepsy, a chronic sleep disorder characterized by frequent sleep/cataplexy attacks and rapid eye movement sleep abnormalities. However, the downstream pathways of orexin signaling are not clearly understood. Here, we show that orexin activates the mTOR pathway, a central regulator of cell growth and metabolism, in the mouse brain and multiple recombinant cell lines that express the G protein-coupled receptors (GPCRs), orexin 1 receptor (OX1R) or orexin 2 receptor (OX2R). This orexin/GPCR-stimulated mTOR activation is sensitive to rapamycin, an inhibitor of mTOR complex 1 (mTORC1) but is independent of two well known mTORC1 activators, Erk and Akt. Rather, our studies indicate that orexin activates mTORC1 via extracellular calcium influx and the lysosome pathway involving v-ATPase and Rag GTPases. Moreover, a cytoplasmic calcium transient is sufficient to mimic orexin/GPCR signaling to mTORC1 activation in a v-ATPase-dependent manner. Together, our studies suggest that the mTORC1 pathway functions downstream of orexin/GPCR signaling, which plays a crucial role in many physiological and metabolic processes.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Shimeng Liu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,; College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Miyo Kakizaki
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yuuki Hirose
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yukiko Ishikawa
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Hiromasa Funato
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and; Department of Anatomy, Toho University School of Medicine, Tokyo 143-8540, Japan
| | - Masashi Yanagisawa
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yonghao Yu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,.
| | - Qinghua Liu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,; International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and.
| |
Collapse
|
266
|
Leprivier G, Rotblat B, Khan D, Jan E, Sorensen PH. Stress-mediated translational control in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:845-60. [PMID: 25464034 DOI: 10.1016/j.bbagrm.2014.11.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/31/2014] [Accepted: 11/04/2014] [Indexed: 12/22/2022]
Abstract
Tumor cells are continually subjected to diverse stress conditions of the tumor microenvironment, including hypoxia, nutrient deprivation, and oxidative or genotoxic stress. Tumor cells must evolve adaptive mechanisms to survive these conditions to ultimately drive tumor progression. Tight control of mRNA translation is critical for this response and the adaptation of tumor cells to such stress forms. This proceeds though a translational reprogramming process which restrains overall translation activity to preserve energy and nutrients, but which also stimulates the selective synthesis of major stress adaptor proteins. Here we present the different regulatory signaling pathways which coordinate mRNA translation in the response to different stress forms, including those regulating eIF2α, mTORC1 and eEF2K, and we explain how tumor cells hijack these pathways for survival under stress. Finally, mechanisms for selective mRNA translation under stress, including the utilization of upstream open reading frames (uORFs) and internal ribosome entry sites (IRESes) are discussed in the context of cell stress. This article is part of a Special Issue entitled: Translation and Cancer.
Collapse
Affiliation(s)
- Gabriel Leprivier
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Barak Rotblat
- Department of Life Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Debjit Khan
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T1Z3, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L4, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada.
| |
Collapse
|
267
|
Shin SW, Park BL, Chang H, Park JS, Bae DJ, Song HJ, Choi IS, Kim MK, Park HS, Kim LH, Namgoong S, Kim JO, Shin HD, Park CS. Exonic variants associated with development of aspirin exacerbated respiratory diseases. PLoS One 2014; 9:e111887. [PMID: 25372592 PMCID: PMC4221198 DOI: 10.1371/journal.pone.0111887] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 09/29/2014] [Indexed: 12/11/2022] Open
Abstract
Aspirin-exacerbated respiratory disease (AERD) is one phenotype of asthma, often occurring in the form of a severe and sudden attack. Due to the time-consuming nature and difficulty of oral aspirin challenge (OAC) for AERD diagnosis, non-invasive biomarkers have been sought. The aim of this study was to identify AERD-associated exonic SNPs and examine the diagnostic potential of a combination of these candidate SNPs to predict AERD. DNA from 165 AERD patients, 397 subjects with aspirin-tolerant asthma (ATA), and 398 normal controls were subjected to an Exome BeadChip assay containing 240K SNPs. 1,023 models (210-1) were generated from combinations of the top 10 SNPs, selected by the p-values in association with AERD. The area under the curve (AUC) of the receiver operating characteristic (ROC) curves was calculated for each model. SNP Function Portal and PolyPhen-2 were used to validate the functional significance of candidate SNPs. An exonic SNP, exm537513 in HLA-DPB1, showed the lowest p-value (p = 3.40×10−8) in its association with AERD risk. From the top 10 SNPs, a combination model of 7 SNPs (exm537513, exm83523, exm1884673, exm538564, exm2264237, exm396794, and exm791954) showed the best AUC of 0.75 (asymptotic p-value of 7.94×10−21), with 34% sensitivity and 93% specificity to discriminate AERD from ATA. Amino acid changes due to exm83523 in CHIA were predicted to be “probably damaging” to the structure and function of the protein, with a high score of ‘1’. A combination model of seven SNPs may provide a useful, non-invasive genetic marker combination for predicting AERD.
Collapse
Affiliation(s)
- Seung-Woo Shin
- Genome Research Center for Allergy and Respiratory Diseases, Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Byung Lae Park
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
| | - HunSoo Chang
- Genome Research Center for Allergy and Respiratory Diseases, Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
- Department of Interdisciplinary Program in Biomedical Science Major Graduate School of Soonchunhyang University, Asan, Republic of Korea
| | - Jong Sook Park
- Genome Research Center for Allergy and Respiratory Diseases, Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Da-Jeong Bae
- Department of Interdisciplinary Program in Biomedical Science Major Graduate School of Soonchunhyang University, Asan, Republic of Korea
| | - Hyun-Ji Song
- Department of Interdisciplinary Program in Biomedical Science Major Graduate School of Soonchunhyang University, Asan, Republic of Korea
| | - Inseon S. Choi
- Department of Allergy, Chonnam National University Medical School and Research Institute of Medical Sciences, Gwangju, Republic of Korea
| | - Mi-Kyeong Kim
- Division of Allergy, Department of Internal Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Hea-Sim Park
- Department of Allergy & Clinical Immunology, Ajou University Hospital, Suwoon, Republic of Korea
| | - Lyoung Hyo Kim
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Suhg Namgoong
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Ji On Kim
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Hyoung Doo Shin
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Choon-Sik Park
- Genome Research Center for Allergy and Respiratory Diseases, Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
- * E-mail: , (SWS)
| |
Collapse
|
268
|
Mackey AM, Sarkes DA, Bettencourt I, Asara JM, Rameh LE. PIP4kγ is a substrate for mTORC1 that maintains basal mTORC1 signaling during starvation. Sci Signal 2014; 7:ra104. [PMID: 25372051 DOI: 10.1126/scisignal.2005191] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphatidylinositol-5-phosphate 4-kinases (PIP4ks) are a family of lipid kinases that specifically use phosphatidylinositol 5-monophosphate (PI-5-P) as a substrate to synthesize phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Suppression of PIP4k function in Drosophila results in smaller cells and reduced target of rapamycin complex 1 (TORC1) signaling. We showed that the γ isoform of PIP4k stimulated signaling through mammalian TORC1 (mTORC1). Knockdown of PIP4kγ reduced cell mass in cells in which mTORC1 is constitutively activated by Tsc2 deficiency. In Tsc2 null cells, mTORC1 activation was partially independent of amino acids or glucose and glutamine. PIP4kγ knockdown inhibited the nutrient-independent activation of mTORC1 in Tsc2 knockdown cells and reduced basal mTORC1 signaling in wild-type cells. PIP4kγ was phosphorylated by mTORC1 and associated with the complex. Phosphorylated PIP4kγ was enriched in light microsomal vesicles, whereas the unphosphorylated form was enriched in heavy microsomal vesicles associated with the Golgi. Furthermore, basal mTORC1 signaling was enhanced by overexpression of unphosphorylated wild-type PIP4kγ or a phosphorylation-defective mutant and decreased by overexpression of a phosphorylation-mimetic mutant. Together, these results demonstrate that PIP4kγ and mTORC1 interact in a self-regulated feedback loop to maintain low and tightly regulated mTORC1 activation during starvation.
Collapse
Affiliation(s)
- Ashley M Mackey
- Boston Biomedical Research Institute, Watertown, MA 02472, USA. Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Ian Bettencourt
- Boston Biomedical Research Institute, Watertown, MA 02472, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA. Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Lucia E Rameh
- Boston Biomedical Research Institute, Watertown, MA 02472, USA. Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
269
|
Peng ZF, Yang L, Wang TT, Han P, Liu ZH, Wei Q. Efficacy and Safety of Sirolimus for Renal Angiomyolipoma in Patients with Tuberous Sclerosis Complex or Sporadic Lymphangioleiomyomatosis: A Systematic Review. J Urol 2014; 192:1424-30. [PMID: 24813310 DOI: 10.1016/j.juro.2014.04.096] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Zhu-feng Peng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Yang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting-ting Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Han
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen-hua Liu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
270
|
Xu K, Liu P, Wei W. mTOR signaling in tumorigenesis. Biochim Biophys Acta Rev Cancer 2014; 1846:638-54. [PMID: 25450580 DOI: 10.1016/j.bbcan.2014.10.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/23/2014] [Accepted: 10/25/2014] [Indexed: 12/25/2022]
Abstract
mTOR (the mechanistic target of rapamycin) is an atypical serine/threonine kinase involved in regulating major cellular functions including growth and proliferation. Deregulation of the mTOR signaling pathway is one of the most commonly observed pathological alterations in human cancers. To this end, oncogenic activation of the mTOR signaling pathway contributes to cancer cell growth, proliferation and survival, highlighting the potential for targeting the oncogenic mTOR pathway members as an effective anti-cancer strategy. In order to do so, a thorough understanding of the physiological roles of key mTOR signaling pathway components and upstream regulators would guide future targeted therapies. Thus, in this review, we summarize available genetic mouse models for mTORC1 and mTORC2 components, as well as characterized mTOR upstream regulators and downstream targets, and assign a potential oncogenic or tumor suppressive role for each evaluated molecule. Together, our work will not only facilitate the current understanding of mTOR biology and possible future research directions, but more importantly, provide a molecular basis for targeted therapies aiming at key oncogenic members along the mTOR signaling pathway.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pengda Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
271
|
Familial or Infectious…or Both? The Complexity of Confirming Causality of Cortical Malformations. Epilepsy Curr 2014; 14:259-60. [PMID: 25346632 DOI: 10.5698/1535-7597-14.5.259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
272
|
Mir SUR, Schwarze SR, Jin L, Zhang J, Friend W, Miriyala S, St Clair D, Craven RJ. Progesterone receptor membrane component 1/Sigma-2 receptor associates with MAP1LC3B and promotes autophagy. Autophagy 2014; 9:1566-78. [DOI: 10.4161/auto.25889] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
273
|
Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A, Rosenberg M, Gray N, Barletta JA, Guo Y, Swanson SJ, Ruan DT, Hanna GJ, Haddad RI, Getz G, Kwiatkowski DJ, Carter SL, Sabatini DM, Jänne PA, Garraway LA, Lorch JH. Response and acquired resistance to everolimus in anaplastic thyroid cancer. N Engl J Med 2014; 371:1426-33. [PMID: 25295501 PMCID: PMC4564868 DOI: 10.1056/nejmoa1403352] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), is effective in treating tumors harboring alterations in the mTOR pathway. Mechanisms of resistance to everolimus remain undefined. Resistance developed in a patient with metastatic anaplastic thyroid carcinoma after an extraordinary 18-month response. Whole-exome sequencing of pretreatment and drug-resistant tumors revealed a nonsense mutation in TSC2, a negative regulator of mTOR, suggesting a mechanism for exquisite sensitivity to everolimus. The resistant tumor also harbored a mutation in MTOR that confers resistance to allosteric mTOR inhibition. The mutation remains sensitive to mTOR kinase inhibitors.
Collapse
Affiliation(s)
- Nikhil Wagle
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School (N.W., E.M.V.A., N.G., R.I.H., D.J.K., P.A.J., L.A.G., J.H.L.), the Department of Medicine, Brigham and Women's Hospital and Harvard Medical School (N.W., E.M.V.A., Y.G., R.I.H., D.J.K., P.A.J., L.A.G., J.H.L.), the Departments of Pathology (J.A.B.) and Surgery (S.J.S., D.T.R.), Brigham and Women's Hospital, the Department of Medicine, Beth Israel Deaconess Medical Center (G.J.H.), and Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute (P.A.J.) - all in Boston; and Broad Institute of the Massachusetts Institute of Technology (MIT) and Harvard (N.W., E.M.V.A., A.A.-M., A.T.-W., M.R., G.G., D.J.K., S.L.C., D.M.S., L.A.G.), Whitehead Institute for Biomedical Research and the MIT Department of Biology (B.C.G., D.M.S.), and Howard Hughes Medical Institute, MIT (B.C.G., D.M.S.) - all in Cambridge, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Malaguarnera R, Chen KY, Kim TY, Dominguez JM, Voza F, Ouyang B, Vundavalli SK, Knauf JA, Fagin JA. Switch in signaling control of mTORC1 activity after oncoprotein expression in thyroid cancer cell lines. J Clin Endocrinol Metab 2014; 99:E1976-87. [PMID: 25029414 PMCID: PMC4184069 DOI: 10.1210/jc.2013-3976] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Thyroid growth is regulated by TSH and requires mammalian target of rapamycin (mTOR). Thyroid cancers frequently exhibit mutations in MAPK and/or phosphoinositol-3-kinase-related kinase effectors. OBJECTIVE The objective of the study was to explore the contribution of RET/PTC, RAS, and BRAF to mTOR regulation and response to mTOR inhibitors. METHODS PCCL3 cells conditionally expressing RET/PTC3, HRAS(G12V), or BRAF(V600E) and human thyroid cancer cells harboring mutations of these genes were used to test pathways controlling mTOR and its requirement for growth. RESULTS TSH/cAMP-induced growth of PCCL3 cells requires mTOR, which is stimulated via protein kinase A in a MAPK kinase (MEK)- and AKT-independent manner. Expression of RET/PTC3, HRAS(G12V), or BRAF(V600E) in PCCL3 cells induces mTOR but does not entirely abrogate the cAMP-mediated control of its activity. Acute oncoprotein-induced mTOR activity is regulated by MEK and AKT, albeit to differing degrees. By contrast, mTOR was not activated by TSH/cAMP in human thyroid cancer cells. Tumor genotype did not predict the effects of rapamycin or the mTOR kinase inhibitor AZD8055 on growth, with the exception of a PTEN-null cell line. Selective blockade of MEK did not influence mTOR activity of BRAF or RAS mutant cells. Combined MEK and mTOR kinase inhibition was synergistic on growth of BRAF- and RAS-mutant thyroid cancer cells in vitro and in vivo. CONCLUSION Thyroid cancer cells lose TSH/cAMP dependency of mTOR signaling and cell growth. mTOR activity is not decreased by the MEK or AKT inhibitors in the RAS or BRAF human thyroid cancer cell lines. This may account for the augmented effects of combining the mTOR inhibitors with selective antagonists of these oncogenic drivers.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- Human Oncology and Pathogenesis Program (R.M., K.-Y.C., T.-Y.K., J.M.D., F.V., S.K.V., J.A.K., J.A.F.) and Department of Medicine (J.A.K., J.A.F.), Memorial Sloan-Kettering Cancer Center, New York, New York 10065; and Division of Endocrinology (B.O.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | | | | | | | | | | | | | | | | |
Collapse
|
275
|
Chantranupong L, Wolfson RL, Orozco JM, Saxton RA, Scaria SM, Bar-Peled L, Spooner E, Isasa M, Gygi SP, Sabatini DM. The Sestrins interact with GATOR2 to negatively regulate the amino-acid-sensing pathway upstream of mTORC1. Cell Rep 2014; 9:1-8. [PMID: 25263562 DOI: 10.1016/j.celrep.2014.09.014] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/03/2014] [Accepted: 09/12/2014] [Indexed: 12/21/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) kinase is a major regulator of cell growth that responds to numerous environmental cues. A key input is amino acids, which act through the heterodimeric Rag GTPases (RagA or RagB bound to RagC or RagD) in order to promote the translocation of mTORC1 to the lysosomal surface, its site of activation. GATOR2 is a complex of unknown function that positively regulates mTORC1 signaling by acting upstream of or in parallel to GATOR1, which is a GTPase-activating protein (GAP) for RagA or RagB and an inhibitor of the amino-acid-sensing pathway. Here, we find that the Sestrins, a family of poorly understood growth regulators (Sestrin1-Sestrin3), interact with GATOR2 in an amino-acid-sensitive fashion. Sestrin2-mediated inhibition of mTORC1 signaling requires GATOR1 and the Rag GTPases, and the Sestrins regulate the localization of mTORC1 in response to amino acids. Thus, we identify the Sestrins as GATOR2-interacting proteins that regulate the amino-acid-sensing branch of the mTORC1 pathway.
Collapse
Affiliation(s)
- Lynne Chantranupong
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA
| | - Rachel L Wolfson
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA
| | - Jose M Orozco
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA
| | - Robert A Saxton
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA
| | - Sonia M Scaria
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Liron Bar-Peled
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA
| | - Eric Spooner
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Marta Isasa
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - David M Sabatini
- Department of Biology, Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 7 Cambridge Center, Cambridge MA 02142, USA.
| |
Collapse
|
276
|
Huang K, Fingar DC. Growing knowledge of the mTOR signaling network. Semin Cell Dev Biol 2014; 36:79-90. [PMID: 25242279 DOI: 10.1016/j.semcdb.2014.09.011] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 12/14/2022]
Abstract
The kinase mTOR (mechanistic target of rapamycin) integrates diverse environmental signals and translates these cues into appropriate cellular responses. mTOR forms the catalytic core of at least two functionally distinct signaling complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). mTORC1 promotes anabolic cellular metabolism in response to growth factors, nutrients, and energy and functions as a master controller of cell growth. While significantly less well understood than mTORC1, mTORC2 responds to growth factors and controls cell metabolism, cell survival, and the organization of the actin cytoskeleton. mTOR plays critical roles in cellular processes related to tumorigenesis, metabolism, immune function, and aging. Consequently, aberrant mTOR signaling contributes to myriad disease states, and physicians employ mTORC1 inhibitors (rapamycin and analogs) for several pathological conditions. The clinical utility of mTOR inhibition underscores the important role of mTOR in organismal physiology. Here we review our growing knowledge of cellular mTOR regulation by diverse upstream signals (e.g. growth factors; amino acids; energy) and how mTORC1 integrates these signals to effect appropriate downstream signaling, with a greater emphasis on mTORC1 over mTORC2. We highlight dynamic subcellular localization of mTORC1 and associated factors as an important mechanism for control of mTORC1 activity and function. We will cover major cellular functions controlled by mTORC1 broadly. While significant advances have been made in the last decade regarding the regulation and function of mTOR within complex cell signaling networks, many important findings remain to be discovered.
Collapse
Affiliation(s)
- Kezhen Huang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, United States
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, United States; Division of Metabolism, Endocrinology, and Diabetes (MEND), Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-2200, United States.
| |
Collapse
|
277
|
New Innovations: Therapies for Genetic Conditions. CURRENT GENETIC MEDICINE REPORTS 2014. [DOI: 10.1007/s40142-014-0043-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
278
|
Liu Z, Chen X, Wang Y, Peng H, Wang Y, Jing Y, Zhang H. PDK4 protein promotes tumorigenesis through activation of cAMP-response element-binding protein (CREB)-Ras homolog enriched in brain (RHEB)-mTORC1 signaling cascade. J Biol Chem 2014; 289:29739-49. [PMID: 25164809 DOI: 10.1074/jbc.m114.584821] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) integrates multiple extracellular and intracellular signals to regulate cell growth and survival. Hyperactivation of mTOR has been observed in various cancers. Regulation of mTOR activity is thus of importance in physiological processes and tumor development. Here, we present pyruvate dehydrogenase kinase 4 (PDK4) as a novel regulator of mTORC1 signaling. mTORC1 activity was augmented with PDK4 overexpression and reduced by PDK4 suppression in various cell lines. Furthermore, PDK4 bound to cAMP-response element-binding protein (CREB) and prevented its degradation. The enhanced CREB consequently transactivated the expression of Ras homolog enriched in brain (RHEB), a direct key activator of mTORC1, independent of AMP-activated protein kinase or tuberous sclerosis complex protein 2. PDK4 potentiated the mTORC1 effectors hypoxia-inducible factor 1α and pyruvate kinase isozymes M2 and promoted aerobic glycolysis (Warburg effect). Knockdown of PDK4 suppressed the tumor development of cancer cells with activated mTORC1. The abundance of PDK4 dictated the responsiveness of cells to the mTOR inhibitor, rapamycin. Combinatory suppression of mTOR and PDK4 exerted synergistic inhibition on cancer cell proliferation. Therefore, PDK4 promotes tumorigenesis through activation of the CREB-RHEB-mTORC1 signaling cascade.
Collapse
Affiliation(s)
- Zhibo Liu
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| | - Xinxin Chen
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| | - Ying Wang
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and the Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Haiyong Peng
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| | - Yanan Wang
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| | - Yanling Jing
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| | - Hongbing Zhang
- From the State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Beijing 100005, China and
| |
Collapse
|
279
|
Byles V, Covarrubias AJ, Ben-Sahra I, Lamming DW, Sabatini DM, Manning BD, Horng T. The TSC-mTOR pathway regulates macrophage polarization. Nat Commun 2014; 4:2834. [PMID: 24280772 PMCID: PMC3876736 DOI: 10.1038/ncomms3834] [Citation(s) in RCA: 457] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/29/2013] [Indexed: 12/27/2022] Open
Abstract
Macrophages are able to polarize to proinflammatory M1 or alternative M2 states with distinct phenotypes and physiological functions. How metabolic status regulates macrophage polarization remains not well understood, and here we examine the role of mTOR (Mechanistic Target of Rapamycin), a central metabolic pathway that couples nutrient sensing to regulation of metabolic processes. Using a mouse model in which myeloid lineage specific deletion of Tsc1 (Tsc1Δ/Δ) leads to constitutive mTOR Complex 1 (mTORC1) activation, we find that Tsc1Δ/Δ macrophages are refractory to IL-4 induced M2 polarization, but produce increased inflammatory responses to proinflammatory stimuli. Moreover, mTORC1-mediated downregulation of Akt signaling critically contributes to defective polarization. These findings highlight a key role for the mTOR pathway in regulating macrophage polarization, and suggest how nutrient sensing and metabolic status could be “hard-wired” to control of macrophage function, with broad implications for regulation of Type 2 immunity, inflammation, and allergy.
Collapse
Affiliation(s)
- Vanessa Byles
- 1] Department of Genetics & Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA [2]
| | | | | | | | | | | | | |
Collapse
|
280
|
Marin-Valencia I, Guerrini R, Gleeson JG. Pathogenetic mechanisms of focal cortical dysplasia. Epilepsia 2014; 55:970-8. [PMID: 24861491 PMCID: PMC4107035 DOI: 10.1111/epi.12650] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2014] [Indexed: 02/01/2023]
Abstract
Focal cortical dysplasias (FCDs) constitute a prevalent cause of intractable epilepsy in children, and is one of the leading conditions requiring epilepsy surgery. Despite recent advances in the cellular and molecular biology of these conditions, the pathogenetic mechanisms of FCDs remain largely unknown. The purpose if this work is to review the molecular underpinnings of FCDs and to highlight potential therapeutic targets. A systematic review of the literature regarding the histologic, molecular, and electrophysiologic aspects of FCDs was conducted. Disruption of the mammalian target of rapamycin (mTOR) signaling comprises a common pathway underlying the structural and electrical disturbances of some FCDs. Other mechanisms such as viral infections, prematurity, head trauma, and brain tumors are also posited. mTOR inhibitors (i.e., rapamycin) have shown positive results on seizure management in animal models and in a small cohort of patients with FCD. Encouraging progress has been achieved on the molecular and electrophysiologic basis of constitutive cells in the dysplastic tissue. Despite the promising results of mTOR inhibitors, large-scale randomized trials are in need to evaluate their efficacy and side effects, along with additional mechanistic studies for the development of novel, molecular-based diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Isaac Marin-Valencia
- Department of Neurology and Neurotherapeutics, and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | | | | |
Collapse
|
281
|
Hazlett LD, Jiang X, McClellan SA. IL-10 function, regulation, and in bacterial keratitis. J Ocul Pharmacol Ther 2014; 30:373-80. [PMID: 24738920 PMCID: PMC4043257 DOI: 10.1089/jop.2014.0018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/15/2014] [Indexed: 01/01/2023] Open
Abstract
The immune system protects the host from pathogenic microbes, but tight regulation of the evoked response is requisite to limit bystander damage. The interleukin (IL)-10 family of cytokines, composed of 9 members: IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, and 3 distantly related members, IL-28A, IL-28B, and IL-29, plays a central role in this regulation. IL-10 family cytokines emerged before the adaptive immune response and elicit diverse host defense mechanisms, especially from epithelial cells during an infection. IL-10 family cytokines are also essential for maintenance and integrity of tissue epithelial layers. These cytokines promote innate immune responses from tissue epithelia that limit the damage caused by both viral and bacterial infections. They also facilitate tissue healing after infection/inflammation. In this regard, IL-10 suppresses pro-inflammatory responses, limiting tissue disruption resulting from an inflammatory response. Thus, a central functional theme of IL-10 family cytokines is their role in tissue protection. This review focuses on IL-10, the founding member of this family of cytokines, and integrates recent data on the function and regulation of IL-10 during bacterial infections. Emphasis is placed on the role of IL-10 in Pseudomonas aeruginosa keratitis and the subsequent infectious/inflammatory processes evoked.
Collapse
Affiliation(s)
- Linda D Hazlett
- Department of Anatomy & Cell Biology, Wayne State University School of Medicine , Detroit, Michigan
| | | | | |
Collapse
|
282
|
Averous J, Lambert-Langlais S, Carraro V, Gourbeyre O, Parry L, B'Chir W, Muranishi Y, Jousse C, Bruhat A, Maurin AC, Proud CG, Fafournoux P. Requirement for lysosomal localization of mTOR for its activation differs between leucine and other amino acids. Cell Signal 2014; 26:1918-27. [PMID: 24793303 DOI: 10.1016/j.cellsig.2014.04.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 04/27/2014] [Indexed: 11/17/2022]
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth and metabolism. It controls many cell functions by integrating nutrient availability and growth factor signals. Amino acids, and in particular leucine, are among the main positive regulators of mTORC1 signaling. The current model for the regulation of mTORC1 by amino acids involves the movement of mTOR to the lysosome mediated by the Rag-GTPases. Here, we have examined the control of mTORC1 signaling and mTOR localization by amino acids and leucine in serum-fed cells, because both serum growth factors (or, e.g., insulin) and amino acids are required for full activation of mTORC1 signaling. We demonstrate that mTORC1 activity does not closely correlate with the lysosomal localization of mTOR. In particular, leucine controls mTORC1 activity without any detectable modification of the lysosomal localization of mTOR, indicating that the signal(s) exerted by leucine is likely distinct from those exerted by other amino acids. In addition, knock-down of the Rag-GTPases attenuated the inhibitory effect of amino acid- or leucine-starvation on the phosphorylation of mTORC1 targets. Furthermore, data from cells where Rag expression has been knocked down revealed that leucine can promote mTORC1 signaling independently of the lysosomal localization of mTOR. Our data complement existing models for the regulation of mTORC1 by amino acids and provide new insights into this important topic.
Collapse
Affiliation(s)
- Julien Averous
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France.
| | - Sarah Lambert-Langlais
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Valérie Carraro
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Ophélie Gourbeyre
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Laurent Parry
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Wafa B'Chir
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Yuki Muranishi
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Céline Jousse
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Alain Bruhat
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Anne-Catherine Maurin
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France
| | - Christopher G Proud
- Centre for Biological Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Pierre Fafournoux
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, F-63122 Saint Genès Champanelle, France; Université Clermont 1, UFR Médecine, UMR 1019 Nutrition Humaine, Clermont-Ferrand, France.
| |
Collapse
|
283
|
Takei N, Nawa H. mTOR signaling and its roles in normal and abnormal brain development. Front Mol Neurosci 2014; 7:28. [PMID: 24795562 PMCID: PMC4005960 DOI: 10.3389/fnmol.2014.00028] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/30/2014] [Indexed: 11/15/2022] Open
Abstract
Target of rapamycin (TOR) was first identified in yeast as a target molecule of rapamycin, an anti-fugal and immunosuppressant macrolide compound. In mammals, its orthologue is called mammalian TOR (mTOR). mTOR is a serine/threonine kinase that converges different extracellular stimuli, such as nutrients and growth factors, and diverges into several biochemical reactions, including translation, autophagy, transcription, and lipid synthesis among others. These biochemical reactions govern cell growth and cause cells to attain an anabolic state. Thus, the disruption of mTOR signaling is implicated in a wide array of diseases such as cancer, diabetes, and obesity. In the central nervous system, the mTOR signaling cascade is activated by nutrients, neurotrophic factors, and neurotransmitters that enhances protein (and possibly lipid) synthesis and suppresses autophagy. These processes contribute to normal neuronal growth by promoting their differentiation, neurite elongation and branching, and synaptic formation during development. Therefore, disruption of mTOR signaling may cause neuronal degeneration and abnormal neural development. While reduced mTOR signaling is associated with neurodegeneration, excess activation of mTOR signaling causes abnormal development of neurons and glia, leading to brain malformation. In this review, we first introduce the current state of molecular knowledge of mTOR complexes and signaling in general. We then describe mTOR activation in neurons, which leads to translational enhancement, and finally discuss the link between mTOR and normal/abnormal neuronal growth during development.
Collapse
Affiliation(s)
- Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University Niigata, Japan
| |
Collapse
|
284
|
Pradhan SA, Rather MI, Tiwari A, Bhat VK, Kumar A. Evidence that TSC2 acts as a transcription factor and binds to and represses the promoter of Epiregulin. Nucleic Acids Res 2014; 42:6243-55. [PMID: 24748662 PMCID: PMC4041451 DOI: 10.1093/nar/gku278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The TSC2 gene, mutated in patients with tuberous sclerosis complex (TSC), encodes a 200 kDa protein TSC2 (tuberin). The importance of TSC2 in the regulation of cell growth and proliferation is irrefutable. TSC2 in complex with TSC1 negatively regulates the mTOR complex 1 (mTORC1) via RHEB in the PI3K-AKT-mTOR pathway and in turn regulates cell proliferation. It shows nuclear as well as cytoplasmic localization. However, its nuclear function remains elusive. In order to identify the nuclear function of TSC2, a whole-genome expression profiling of TSC2 overexpressing cells was performed, and the results showed differential regulation of 266 genes. Interestingly, transcription was found to be the most populated functional category. EREG (Epiregulin), a member of the epidermal growth factor family, was found to be the most downregulated gene in the microarray analysis. Previous reports have documented elevated levels of EREG in TSC lesions, making its regulatory aspects intriguing. Using the luciferase reporter, ChIP and EMSA techniques, we show that TSC2 binds to the EREG promoter between −352 bp and −303 bp and negatively regulates its expression. This is the first evidence for the role of TSC2 as a transcription factor and of TSC2 binding to the promoter of any gene.
Collapse
Affiliation(s)
- Shalmali Avinash Pradhan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Mohammad Iqbal Rather
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Ankana Tiwari
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Vishwanath Kumble Bhat
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
285
|
O'Connor EC, Bariselli S, Bellone C. Synaptic basis of social dysfunction: a focus on postsynaptic proteins linking group-I mGluRs with AMPARs and NMDARs. Eur J Neurosci 2014; 39:1114-29. [DOI: 10.1111/ejn.12510] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/06/2014] [Accepted: 01/10/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Eoin C. O'Connor
- Department of Basic Neurosciences; Medical Faculty; University of Geneva; 1 Rue Michel Servet CH-1211 Geneva Switzerland
| | - Sebastiano Bariselli
- Department of Basic Neurosciences; Medical Faculty; University of Geneva; 1 Rue Michel Servet CH-1211 Geneva Switzerland
| | - Camilla Bellone
- Department of Basic Neurosciences; Medical Faculty; University of Geneva; 1 Rue Michel Servet CH-1211 Geneva Switzerland
| |
Collapse
|
286
|
Matter MS, Decaens T, Andersen JB, Thorgeirsson SS. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J Hepatol 2014; 60:855-65. [PMID: 24308993 PMCID: PMC3960348 DOI: 10.1016/j.jhep.2013.11.031] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/28/2013] [Accepted: 11/27/2013] [Indexed: 02/07/2023]
Abstract
Mechanistic target of rapamycin (mTOR) regulates cell growth, metabolism and aging in response to nutrients, cellular energy stage and growth factors. mTOR is frequently up-regulated in cancer including hepatocellular carcinoma (HCC) and is associated with bad prognosis, poorly differentiated tumors, and earlier recurrence. Blocking mTOR with rapamycin and first generation mTOR inhibitors, called rapalogs, has shown promising reduction of HCC tumor growth in preclinical models. Currently, rapamycin/rapalogs are used in several clinical trials for the treatment of advanced HCC, and as adjuvant therapy in HCC patients after liver transplantation and TACE. A second generation of mTOR pathway inhibitors has been developed recently and is being tested in various clinical trials of solid cancers, and has been used in preclinical HCC models. The results of series of clinical trials using mTOR inhibitors in HCC treatment will emerge in the near future.
Collapse
|
287
|
Picchioni D, Reith RM, Nadel JL, Smith CB. Sleep, plasticity and the pathophysiology of neurodevelopmental disorders: the potential roles of protein synthesis and other cellular processes. Brain Sci 2014; 4:150-201. [PMID: 24839550 PMCID: PMC4020186 DOI: 10.3390/brainsci4010150] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/26/2014] [Accepted: 03/07/2014] [Indexed: 12/28/2022] Open
Abstract
Sleep is important for neural plasticity, and plasticity underlies sleep-dependent memory consolidation. It is widely appreciated that protein synthesis plays an essential role in neural plasticity. Studies of sleep-dependent memory and sleep-dependent plasticity have begun to examine alterations in these functions in populations with neurological and psychiatric disorders. Such an approach acknowledges that disordered sleep may have functional consequences during wakefulness. Although neurodevelopmental disorders are not considered to be sleep disorders per se, recent data has revealed that sleep abnormalities are among the most prevalent and common symptoms and may contribute to the progression of these disorders. The main goal of this review is to highlight the role of disordered sleep in the pathology of neurodevelopmental disorders and to examine some potential mechanisms by which sleep-dependent plasticity may be altered. We will also briefly attempt to extend the same logic to the other end of the developmental spectrum and describe a potential role of disordered sleep in the pathology of neurodegenerative diseases. We conclude by discussing ongoing studies that might provide a more integrative approach to the study of sleep, plasticity, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dante Picchioni
- Behavioral Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; E-Mail:
- Advanced MRI Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - R. Michelle Reith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - Jeffrey L. Nadel
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| | - Carolyn B. Smith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, Bethesda, MD 20892, USA; E-Mails: (R.M.R.); (J.L.N.)
| |
Collapse
|
288
|
|
289
|
Yefidoff-Freedman R, Chen T, Sahoo R, Chen L, Wagner G, Halperin JA, Aktas BH, Chorev M. 3-substituted indazoles as configurationally locked 4EGI-1 mimetics and inhibitors of the eIF4E/eIF4G interaction. Chembiochem 2014; 15:595-611. [PMID: 24458973 DOI: 10.1002/cbic.201300723] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Indexed: 12/12/2022]
Abstract
4EGI-1, the prototypic inhibitor of eIF4E/eIF4G interaction, was identified in a high-throughput screening of small-molecule libraries with the aid of a fluorescence polarization assay that measures inhibition of binding of an eIF4G-derived peptide to recombinant eIF4E. As such, the molecular probe 4EGI-1 has potential for the study of molecular mechanisms involved in human disorders characterized by loss of physiological restraints on translation initiation. A hit-to-lead optimization campaign was carried out to overcome the configurational instability in 4EGI-1, which stems from the E-to-Z isomerization of the hydrazone function. We identified compound 1 a, in which the labile hydrazone was incorporated into a rigid indazole scaffold, as a promising rigidified 4EGI-1 mimetic lead. In a structure-activity relationship study directed towards probing the structural latitude of this new chemotype as an inhibitor of eIF4E/eIF4G interaction and translation initiation we identified 1 d, an indazole-based 4EGI-1 mimetic, as a new and improved lead inhibitor of eIF4E/eIF4G interaction and a promising molecular probe candidate for elucidation of the role of cap-dependent translation initiation in a host of pathophysiological states.
Collapse
Affiliation(s)
- Revital Yefidoff-Freedman
- Laboratory for Translational Research, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115 (USA); Hematology Laboratory for Translational Research, Brigham and Women's Hospital, Harvard Medical School, 20 Shattuck Street, Thorn 7, Boston, MA 02115 (USA)
| | | | | | | | | | | | | | | |
Collapse
|
290
|
Munkley J, Rajan P, Laferty NP, Dalgliesh C, Jackson RM, Robson CN, Leung HY, Elliott DJ. A novel androgen-regulated isoform of the TSC2 tumour suppressor gene increases cell proliferation. Oncotarget 2014; 5:131-9. [PMID: 24318044 PMCID: PMC3960195 DOI: 10.18632/oncotarget.1405] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/19/2013] [Indexed: 01/07/2023] Open
Abstract
TSC2 (Tuberous sclerosis complex 2) is an important tumour suppressor gene, mutations within which are linked to the development of tuberous sclerosis and implicated in multiple tumour types. TSC2 protein complexes with TSC1 and blocks the ability of the Rheb (Ras homolog enriched in brain) GTPase to activate mTOR (mammalian target of rapamycin), a crucial signal transducer which regulates protein synthesis and cell growth. Here, we report the characterisation of a novel isoform of TSC2 which is under direct control of the ligand-activated androgen receptor. TSC2 isoform A (TSC2A) is derived from an internal androgen-regulated alternative promoter and encodes a 508-amino acid cytoplasmic protein corresponding to the C-terminal region of full-length TSC2, lacking the interaction domain for TSC1 and containing an incomplete interaction domain required for Rheb inactivation. Expression of TSC2A is induced in response to androgens and full-length TSC2 is co-ordinately down-regulated, indicating an androgen-driven switch in TSC2 protein isoforms. In contrast to the well-characterised suppressive effect on cell proliferation of full-length TSC2 protein, both LNCaP and HEK293 cells over-expressing TSC2 isoform A proliferate more rapidly (measured by MTT assays) and have increased levels of cells in S-phase (measured by both Edu staining and FACS analysis). Our work indicates, for the first time, a novel role for this well-known tumour suppressor gene, which encodes an activator of cell proliferation in response to androgen stimulation.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Prabhakar Rajan
- Beatson Institute for Cancer Research, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicholas P. Laferty
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Caroline Dalgliesh
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Robert M. Jackson
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Craig N. Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Hing Y. Leung
- Beatson Institute for Cancer Research, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David J. Elliott
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
291
|
Wang P, Drackley J, Stamey-Lanier J, Keisler D, Loor J. Effects of level of nutrient intake and age on mammalian target of rapamycin, insulin, and insulin-like growth factor-1 gene network expression in skeletal muscle of young Holstein calves. J Dairy Sci 2014; 97:383-91. [DOI: 10.3168/jds.2013-7042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/15/2013] [Indexed: 11/19/2022]
|
292
|
Picker JD, Walsh CA. New innovations: therapeutic opportunities for intellectual disabilities. Ann Neurol 2013; 74:382-90. [PMID: 24038210 DOI: 10.1002/ana.24002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/01/2013] [Accepted: 07/29/2013] [Indexed: 12/25/2022]
Abstract
Intellectual disability is common and is associated with significant morbidity. Until the latter half of the 20th century, there were no efficacious treatments. Following initial breakthroughs associated with newborn screening and metabolic corrections, little progress was made until recently. With improved understanding of genetic and cellular mechanisms, novel treatment options are beginning to appear for a number of specific conditions. Fragile X and tuberous sclerosis offer paradigms for the development of targeted therapeutics, but advances in understanding of other disorders such as Down syndrome and Rett syndrome, for example, are also resulting in promising treatment directions. In addition, better understanding of the underlying neurobiology is leading to novel developments in enzyme replacement for storage disorders and adjunctive therapies for metabolic disorders, as well as potentially more generalizable approaches that target dysfunctional cell regulation via RNA and chromatin. Physiologic therapies, including deep brain stimulation and transcranial magnetic stimulation, offer yet another direction to enhance cognitive functioning. Current options and evolving opportunities for the intellectually disabled are reviewed and exemplified.
Collapse
Affiliation(s)
- Jonathan D Picker
- Division of Genetics, Boston Children's Hospital, and Howard Hughes Medical Institute, Boston, MA; Departments of Pediatrics and Neurology,, Harvard Medical School, Boston, MA
| | | |
Collapse
|
293
|
Abstract
The mammalian target of rapamycin (mTOR) pathway is an essential cellular signaling pathway involved in a number of important physiological functions, including cell growth, proliferation, metabolism, protein synthesis, and autophagy. Dysregulation of the mTOR pathway has been implicated in the pathophysiology of a number of neurological diseases. Hyperactivation of the mTOR pathway, leading to increased cell growth and proliferation, has been most convincingly shown to stimulate tumor growth in the brain and other organs in the genetic disorder, tuberous sclerosis complex (TSC). In addition, mTOR may also play a role in promoting epileptogenesis or maintaining seizures in TSC, as well as in acquired epilepsies following brain injury. Finally, the mTOR pathway may also be involved in the pathogenesis of cognitive dysfunction and other neurological deficits in developmental disorders and neurodegenerative diseases. mTOR inhibitors, such as rapamycin and its analogs, may represent novel, rational therapies for a variety of neurological disorders.
Collapse
Affiliation(s)
- Michael Wong
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
294
|
Tanaka M, Wataya-Kaneda M, Nakamura A, Matsumoto S, Katayama I. First left-right comparative study of topical rapamycin vs. vehicle for facial angiofibromas in patients with tuberous sclerosis complex. Br J Dermatol 2013; 169:1314-8. [DOI: 10.1111/bjd.12567] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2013] [Indexed: 11/27/2022]
Affiliation(s)
- M. Tanaka
- Department of Dermatology; Graduate School of Medicine; Osaka University; 2-2 Yamada-oka Suita Osaka 565-0871 Japan
| | - M. Wataya-Kaneda
- Department of Dermatology; Graduate School of Medicine; Osaka University; 2-2 Yamada-oka Suita Osaka 565-0871 Japan
| | - A. Nakamura
- Department of Pharmacy; Osaka University Hospital; Osaka Japan
| | - S. Matsumoto
- Department of Pharmacy; Osaka University Hospital; Osaka Japan
| | - I. Katayama
- Department of Dermatology; Graduate School of Medicine; Osaka University; 2-2 Yamada-oka Suita Osaka 565-0871 Japan
| |
Collapse
|
295
|
Tan SH, Shui G, Zhou J, Shi Y, Huang J, Xia D, Wenk MR, Shen HM. Critical role of SCD1 in autophagy regulation via lipogenesis and lipid rafts-coupled AKT-FOXO1 signaling pathway. Autophagy 2013; 10:226-42. [PMID: 24296537 DOI: 10.4161/auto.27003] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
SCD1 (stearoyl-coenzyme A desaturase 1) is an endoplasmic reticulum-bound enzyme that catalyzes the formation of the first double bond at the cis-Δ9 position of saturated fatty acids (SFA) to form monounsaturated fatty acids (MUFA). Increasing evidence indicates that autophagy plays an important role in regulating lipid metabolism, while little is known about whether key enzymes of lipogenesis like SCD1 can regulate autophagy. In this study, we examined the role of SCD1 in autophagy using the tsc2(-/-) mouse embryonic fibroblasts (MEFs) possessing constitutively active MTORC1 as a cellular model. We found that mRNA and protein levels of SCD1 are significantly elevated in the tsc2(-/-) MEFs compared with Tsc2(+/+) MEFs, resulting in significant increases in levels of various lipid classes. Furthermore, inhibition of SCD1 activity by either a chemical inhibitor or genetic knockdown resulted in an increase of autophagic flux only in the tsc2(-/-) MEFs. Induction of autophagy was independent of MTOR as MTORC1 activity was not suppressed by SCD1 inhibition. Loss of phosphorylation on AKT Ser473 was observed upon SCD1 inhibition and such AKT inactivation was due to disruption of lipid raft formation, without affecting the formation and activity of MTORC2. Increased nuclear translocation of FOXO1 was observed following AKT inactivation, leading to increased transcription of genes involved in the autophagic process. The tsc2(-/-) MEFs were also more susceptible to apoptosis induced by SCD1 inhibition and blockage of autophagy sensitized the cell death response. These results revealed a novel function of SCD1 on regulation of autophagy via lipogenesis and the lipid rafts-AKT-FOXO1 pathway.
Collapse
Affiliation(s)
- Shi-Hao Tan
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Beijing, China
| | - Jing Zhou
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Yin Shi
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Jingxiang Huang
- Department of Pathology; National University Hospital of Singapore; Singapore
| | - Dajing Xia
- Zhejiang University School of Public Health; Hangzhou, Zhejiang, China
| | - Markus R Wenk
- NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore; Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Han-Ming Shen
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore; NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| |
Collapse
|
296
|
Procaccini C, De Rosa V, Galgani M, Carbone F, La Rocca C, Formisano L, Matarese G. Role of adipokines signaling in the modulation of T cells function. Front Immunol 2013; 4:332. [PMID: 24151494 PMCID: PMC3799205 DOI: 10.3389/fimmu.2013.00332] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/01/2013] [Indexed: 12/15/2022] Open
Abstract
The field that links immunity and metabolism is rapidly expanding. Apparently non-immunological disorders such as obesity and type 2 diabetes have been linked to immune dysregulation, suggesting that metabolic alterations can be induced by or be consequence of an altered self-immune tolerance. In this context, adipose tissue produces and releases a variety of pro-inflammatory and anti-inflammatory factors, termed "adipokines," which can be considered as the bridge between obesity-related exogenous factors, such as nutrition and lifestyle, and the molecular events leading to metabolic syndrome, inflammatory, and/or autoimmune conditions. In obesity, increased production of most adipokines impacts on multiple functions such as appetite and energy balance, modulation of immune responses, insulin sensitivity, angiogenesis, blood pressure, lipid metabolism, and so on. This report aims to discuss some of the recent topics of adipocytokine research and their related signaling pathways, that may be of particular importance as could lead to effective therapeutic strategies for obesity-associated diseases.
Collapse
Affiliation(s)
- Claudio Procaccini
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Fortunata Carbone
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Claudia La Rocca
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Luigi Formisano
- Department of Biological, Geological and Environmental Sciences, Division of Pharmacology, University of Sannio, Benevento, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Salerno, Salerno, Italy
- IRCCS MultiMedica, Milano, Italy
| |
Collapse
|
297
|
Mester J, Eng C. When overgrowth bumps into cancer: the PTEN-opathies. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2013; 163C:114-21. [PMID: 23613428 DOI: 10.1002/ajmg.c.31364] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PTEN is a dual-specificity phosphatase and well-known tumor suppressor gene. When functioning properly, it works in its canonical pathway to inhibit AKT/mTOR and MAPK signaling, leading to cell death and growth regulation. PTEN mutations cause dysregulation of these pathways, resulting in cellular proliferation and overgrowth. When germline mutations are present as in patients with PTEN Hamartoma Tumor Syndrome (PHTS), benign and malignant neoplasias occur as well as cerebral overgrowth and neurodevelopmental abnormalities. This review article will summarize recent laboratory and clinical investigations relating to PTEN, highlighting the overgrowth aspects of this syndrome and the molecular drivers behind these key phenotypes. Finally, therapies developed targeted the PI3K/AKT/mTOR pathway for other tumor predisposition syndromes will be discussed.
Collapse
Affiliation(s)
- Jessica Mester
- PTEN/Cowden Multidisciplinary Clinic, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
298
|
Tsun ZY, Bar-Peled L, Chantranupong L, Zoncu R, Wang T, Kim C, Spooner E, Sabatini DM. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol Cell 2013; 52:495-505. [PMID: 24095279 DOI: 10.1016/j.molcel.2013.09.016] [Citation(s) in RCA: 421] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 09/18/2013] [Accepted: 09/18/2013] [Indexed: 10/26/2022]
Abstract
The mTORC1 kinase is a master growth regulator that senses numerous environmental cues, including amino acids. The Rag GTPases interact with mTORC1 and signal amino acid sufficiency by promoting the translocation of mTORC1 to the lysosomal surface, its site of activation. The Rags are unusual GTPases in that they function as obligate heterodimers, which consist of RagA or B bound to RagC or D. While the loading of RagA/B with GTP initiates amino acid signaling to mTORC1, the role of RagC/D is unknown. Here, we show that RagC/D is a key regulator of the interaction of mTORC1 with the Rag heterodimer and that, unexpectedly, RagC/D must be GDP bound for the interaction to occur. We identify FLCN and its binding partners, FNIP1/2, as Rag-interacting proteins with GAP activity for RagC/D, but not RagA/B. Thus, we reveal a role for RagC/D in mTORC1 activation and a molecular function for the FLCN tumor suppressor.
Collapse
Affiliation(s)
- Zhi-Yang Tsun
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Liron Bar-Peled
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Lynne Chantranupong
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Roberto Zoncu
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Tim Wang
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Choah Kim
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Eric Spooner
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, Nine Cambridge Center, Cambridge, MA 02142, USA.,Koch Institute for Integrative for Cancer Research, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
299
|
Lee KM, Hwang SK, Lee JA. Neuronal autophagy and neurodevelopmental disorders. Exp Neurobiol 2013; 22:133-42. [PMID: 24167408 PMCID: PMC3807000 DOI: 10.5607/en.2013.22.3.133] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/12/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022] Open
Abstract
Neurodevelopmental disorders include a wide range of diseases such as autism spectrum disorders and mental retardation. Mutations in several genes that regulate neural development and synapse function have been identified in neurodevelopmental disorders. Interestingly, some affected genes and pathways in these diseases are associated with the autophagy pathway. Autophagy is a complex, bulky degradative process that involves the sequestration of cellular proteins, RNA, lipids, and cellular organelles into lysosomes. Despite recent progress in elucidating the genetics and molecular pathogenesis of these disorders, little is known about the pathogenic mechanisms and autophagy-related pathways involved in common neurodevelopmental disorders. Therefore, in this review, we focus on the current understanding of neuronal autophagy as well as recent findings on genetics and the roles of autophagy pathway in common neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kyung-Min Lee
- Department of Anatomy, Graduate School of Medicine, Kyungpook National University, Daegu 700-422, Korea
| | | | | |
Collapse
|
300
|
Abstract
Neurite growth requires neurite extension and retraction, which are associated with protein degradation. Autophagy is a conserved bulk degradation pathway that regulates several cellular processes. However, little is known about autophagic regulation during early neurite growth. In this study, we investigated whether autophagy was involved in early neurite growth and how it regulated neurite growth in primary cortical neurons. Components of autophagy were expressed and autophagy was activated during early neurite growth. Interestingly, inhibition of autophagy by atg7 small interfering RNA (siRNA) caused elongation of axons, while activation of autophagy by rapamycin suppressed axon growth. Surprisingly, inhibition of autophagy reduced the protein level of RhoA. Moreover, expression of RhoA suppressed axon overelongation mediated by autophagy inhibition, whereas inhibition of the RhoA signaling pathway by Y-27632 recovered rapamycin-mediated suppression of axon growth. Interestingly, hnRNP-Q1, which negatively regulates RhoA, accumulated in autophagy-deficient neurons, while its protein level was reduced by autophagy activation. Overall, our study suggests that autophagy negatively regulates axon extension via the RhoA-ROCK pathway by regulating hnRNP-Q1 in primary cortical neurons. Therefore, autophagy might serve as a fine-tuning mechanism to regulate early axon extension.
Collapse
|