1
|
Lienda M, Mwila M, Sichula C, Kabengele C, Akombwa M, Zulu C, Banda CH, M’hango H. Diagnosis and Management of Tuberous Sclerosis Complex in a Resource-Limited Setting-A Case Report of a 14-Year-Old Female Zambian Adolescent. CLINICAL MEDICINE INSIGHTS-CASE REPORTS 2025; 18:11795476251321268. [PMID: 39974288 PMCID: PMC11837062 DOI: 10.1177/11795476251321268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/31/2025] [Indexed: 02/21/2025]
Abstract
Tuberous sclerosis complex (TSC) is a rare multisystemic neurocutaneous syndrome with a wide spectrum of clinical manifestations. We present a case of a 14-year-old adolescent female who presented with a history of facial angiofibromas since the age of 8 months. Physical examination was remarkable for multiple angiofibromas on the face, and other multiple cutaneous manifestations of TSC. MRI of the head, and abdomen revealed cortical tubers, multiple bilateral periventricular and subependymal nodular lesions, calcifications, and bilateral kidney enlargement with multiple bilateral renal angiomyolipomas of varying sizes in a background of bilateral polycystic kidneys, MRI of the chest was unremarkable. A diagnosis of TSC was made using the clinical diagnostic criteria which consist of major and minor features. A diagnosis using genetic studies could not be made due to a lack of resources. Management was multidisciplinary and regular monitoring every 6 months will be required to monitor disease progression and manage complications as they arise. This case illustrates the multidisciplinary approach needed to address the diverse clinical manifestations of TSC and the diagnostic challenges, treatment limitations, and psychological impact of TSC in low-resource settings like Zambia where access to advanced therapies is limited.
Collapse
Affiliation(s)
| | - Meek Mwila
- University of Zambia School of Medicine, Lusaka, Zambia
| | - Chilala Sichula
- Department of Paediatrics and Child Health, University of Zambia School of Medicine, Lusaka, Zambia
| | | | - Moses Akombwa
- Department of Radiology, University Teaching Hospital—Adult Hospital, Lusaka, Zambia
| | - Christina Zulu
- Department of Paediatrics and Child Health, University of Zambia School of Medicine, Lusaka, Zambia
| | - Chihena Hansini Banda
- Plastic and Reconstructive Surgery Unit, Department of Surgery, University Teaching Hospital, Lusaka, Zambia
| | - Hellen M’hango
- Department of Paediatrics and Child Health, University of Zambia School of Medicine, Lusaka, Zambia
| |
Collapse
|
2
|
Baldi BG, Feitosa PHR, Rubin AS, Amaral AF, Freitas CSG, da Costa CH, Mancuzo EV, do Nascimento ECT, Araujo MS, Rocha MJJ, de Oliveira MR, Galvão TS, Torres PPTES, Carvalho CRR. Brazilian Thoracic Association recommendations for the management of lymphangioleiomyomatosis. J Bras Pneumol 2025; 51:e20240378. [PMID: 39936727 PMCID: PMC11796567 DOI: 10.36416/1806-3756/e20240378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare disease, characterized as a low-grade neoplasm with metastatic potential that mainly affects women of reproductive age, in which there is proliferation of atypical smooth muscle cells (LAM cells) and formation of diffuse pulmonary cysts. It can occur in a sporadic form or in combination with tuberous sclerosis complex. In recent decades, a number of advances have been made in the understanding of the pathophysiology and management of LAM, leading to improvements in its prognosis: identification of the main genetic aspects and the role of the mechanistic target of rapamycin (mTOR) pathway; relationship with hormonal factors, mainly estrogen; characterization of pulmonary and extrapulmonary manifestations in imaging studies; identification and importance in the diagnosis of VEGF-D; a systematic diagnostic approach, often without the need for lung biopsy; use of and indications for the use of mTOR inhibitors, mainly sirolimus, for pulmonary and extrapulmonary manifestations; pulmonary rehabilitation and the management of complications such as pneumothorax and chylothorax; and the role of and indications for lung transplantation. To date, no Brazilian recommendations for a comprehensive approach to the disease have been published. This document is the result of a non-systematic review of the literature, carried out by 12 pulmonologists, a radiologist, and a pathologist, which aims to provide an update of the most important topics related to LAM, mainly to its diagnosis, treatment, and follow-up, including practical and multidisciplinary aspects of its management.
Collapse
Affiliation(s)
- Bruno Guedes Baldi
- . Divisao de Pneumologia, Instituto do Coracao - InCor - Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo - HCFMUSP - São Paulo (SP) Brasil
| | | | - Adalberto Sperb Rubin
- . Serviço de Pneumologia, Pavilhão Pereira Filho, Santa Casa de Porto Alegre, Porto Alegre (RS) Brasil
| | - Alexandre Franco Amaral
- . Divisao de Pneumologia, Instituto do Coracao - InCor - Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo - HCFMUSP - São Paulo (SP) Brasil
| | | | | | - Eliane Viana Mancuzo
- . Serviço de Pneumologia e Cirurgia Torácica, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte (MG) Brasil
| | | | - Mariana Sponholz Araujo
- . Divisão de Pneumologia, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba (PR) Brasil
| | | | - Martina Rodrigues de Oliveira
- . Divisao de Pneumologia, Instituto do Coracao - InCor - Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo - HCFMUSP - São Paulo (SP) Brasil
| | - Tatiana Senna Galvão
- . Hospital Universitário Professor Edgar Santos, Universidade Federal da Bahia, Salvador (BA) Brasil
| | | | - Carlos Roberto Ribeiro Carvalho
- . Divisao de Pneumologia, Instituto do Coracao - InCor - Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo - HCFMUSP - São Paulo (SP) Brasil
| |
Collapse
|
3
|
Pandya PH, Jannu AJ, Bijangi-Vishehsaraei K, Dobrota E, Bailey BJ, Barghi F, Shannon HE, Riyahi N, Damayanti NP, Young C, Malko R, Justice R, Albright E, Sandusky GE, Wurtz LD, Collier CD, Marshall MS, Gallagher RI, Wulfkuhle JD, Petricoin EF, Coy K, Trowbridge M, Sinn AL, Renbarger JL, Ferguson MJ, Huang K, Zhang J, Saadatzadeh MR, Pollok KE. Integrative Multi-OMICs Identifies Therapeutic Response Biomarkers and Confirms Fidelity of Clinically Annotated, Serially Passaged Patient-Derived Xenografts Established from Primary and Metastatic Pediatric and AYA Solid Tumors. Cancers (Basel) 2022; 15:259. [PMID: 36612255 PMCID: PMC9818438 DOI: 10.3390/cancers15010259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Establishment of clinically annotated, molecularly characterized, patient-derived xenografts (PDXs) from treatment-naïve and pretreated patients provides a platform to test precision genomics-guided therapies. An integrated multi-OMICS pipeline was developed to identify cancer-associated pathways and evaluate stability of molecular signatures in a panel of pediatric and AYA PDXs following serial passaging in mice. Original solid tumor samples and their corresponding PDXs were evaluated by whole-genome sequencing, RNA-seq, immunoblotting, pathway enrichment analyses, and the drug−gene interaction database to identify as well as cross-validate actionable targets in patients with sarcomas or Wilms tumors. While some divergence between original tumor and the respective PDX was evident, majority of alterations were not functionally impactful, and oncogenic pathway activation was maintained following serial passaging. CDK4/6 and BETs were prioritized as biomarkers of therapeutic response in osteosarcoma PDXs with pertinent molecular signatures. Inhibition of CDK4/6 or BETs decreased osteosarcoma PDX growth (two-way ANOVA, p < 0.05) confirming mechanistic involvement in growth. Linking patient treatment history with molecular and efficacy data in PDX will provide a strong rationale for targeted therapy and improve our understanding of which therapy is most beneficial in patients at diagnosis and in those already exposed to therapy.
Collapse
Affiliation(s)
- Pankita H. Pandya
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Asha Jacob Jannu
- Department of Biostatistics & Health Data Science Indiana, University School of Medicine, Indianapolis, IN 46202, USA
| | - Khadijeh Bijangi-Vishehsaraei
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Erika Dobrota
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Barbara J. Bailey
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Farinaz Barghi
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harlan E. Shannon
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Niknam Riyahi
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nur P. Damayanti
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Courtney Young
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rada Malko
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ryli Justice
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eric Albright
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George E. Sandusky
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - L. Daniel Wurtz
- Department of Orthopedics Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Christopher D. Collier
- Department of Orthopedics Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark S. Marshall
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rosa I. Gallagher
- Center for Applied Proteomics and Molecular Medicine, Institute for Biomedical Innovation, George Mason University, Manassas, VA 20110, USA
| | - Julia D. Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, Institute for Biomedical Innovation, George Mason University, Manassas, VA 20110, USA
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, Institute for Biomedical Innovation, George Mason University, Manassas, VA 20110, USA
| | - Kathy Coy
- Preclinical Modeling and Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa Trowbridge
- Preclinical Modeling and Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anthony L. Sinn
- Preclinical Modeling and Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jamie L. Renbarger
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael J. Ferguson
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kun Huang
- Department of Biostatistics & Health Data Science Indiana, University School of Medicine, Indianapolis, IN 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - M. Reza Saadatzadeh
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E. Pollok
- Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Grgat D, Dilber D, Hrabak Paar M. Common benign primary pediatric cardiac tumors: a primer for radiologists. Jpn J Radiol 2022; 41:477-487. [PMID: 36495370 DOI: 10.1007/s11604-022-01371-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Cardiac tumors are neoplasms arising from or located in the heart or the pericardium. Although rare, primary cardiac tumors in children require an accurate and timely diagnosis. Most pediatric primary cardiac tumors are benign (around 90%). Echocardiography is the first imaging modality used due to its availability, noninvasiveness, inexpensiveness, and absence of ionizing radiation. Computed tomography (CT) and magnetic resonance imaging (MRI) offer better soft tissue visualization as well as better visualization of extracardiac structures. A great advantage of MRI is the possibility of measuring cardiac function and blood flow, which can be important for obstructing cardiac tumors. In this article, we will offer a brief review of clinical, echocardiographic, CT, and MRI features of cardiac rhabdomyomas, fibromas, teratomas, and lipomas providing their differential diagnosis.
Collapse
Affiliation(s)
- Dora Grgat
- Institute for Emergency Medicine of Zagreb County, Velika Gorica, Croatia
| | - Daniel Dilber
- School of Medicine, Department of Pediatrics, University of Zagreb, University Hospital Center Zagreb, Zagreb, Croatia
| | - Maja Hrabak Paar
- School of Medicine, Department of Diagnostic and Interventional Radiology, University of Zagreb, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia.
| |
Collapse
|
5
|
Gorini F, Santoro M, Pierini A, Mezzasalma L, Baldacci S, Bargagli E, Boncristiano A, Brunetto MR, Cameli P, Cappelli F, Castaman G, Coco B, Donati MA, Guerrini R, Linari S, Murro V, Olivotto I, Parronchi P, Pochiero F, Rossi O, Scappini B, Sodi A, Vannucchi AM, Coi A. Orphan Drug Use in Patients With Rare Diseases: A Population-Based Cohort Study. Front Pharmacol 2022; 13:869842. [PMID: 35652051 PMCID: PMC9148958 DOI: 10.3389/fphar.2022.869842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Orphan drugs are used for the diagnosis, prevention and treatment of rare diseases that, in the European Union, are defined as disorders affecting no more than 5 persons in 10,000. So far, a total of around 800 orphan medicinal products have been approved by the European Medicines Agency, however the utilization profile of orphan drugs has yet to be explored. This study aimed at assessing the utilization profile of orphan drugs authorized for marketing by the Italian Medicines Agency using population-based data. Methods: A total of 21 orphan drugs used in outpatient settings, approved in the European Union before or during the 2008-2018 period and involving 15 rare diseases, were included in the study. The monitored population included patients with one of the conditions surveilled by the population-based Tuscany Registry of Rare Diseases and diagnosed between 2000-2018. A multi-database approach was applied, by linking data from the registry with information collected in drug prescriptions databases. The prevalence and intensity of use were estimated for the selected orphan drugs and other non-orphan medications, used to treat the same rare disease and for which a change in the prevalence of use was hypothesized after authorization of the orphan drug. Results: For some diseases (acquired aplastic anemia, tuberous sclerosis complex, most metabolic diseases) a low prevalence of orphan drugs use was observed (range between 1.1-12.5%). Conversely, orphan drugs were frequently used in hemophilia B, Wilson disease and idiopathic pulmonary fibrosis (maximum of 78.3, 47.6 and 41.8%, respectively). For hemophilia B and Leber's hereditary optic neuropathy, there are currently no other medications used in clinical practice in addition to orphan drugs. Six orphan drugs were used for the treatment of pulmonary arterial hypertension, appearing the elective therapy for this disease, albeit with different utilization profiles (range of prevalence 1.7-55.6%). Conclusion: To the best of our knowledge, this is the first study investigating the utilization profile of orphan drugs prescribed in a defined geographical area, and providing relevant information to monitor over time potential changes in the prevalence of these medications as well as in the health care decision making.
Collapse
Affiliation(s)
- Francesca Gorini
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Michele Santoro
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Anna Pierini
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Lorena Mezzasalma
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Silvia Baldacci
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Siena, Italy
| | | | | | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Siena, Italy
| | - Francesco Cappelli
- Cardiomyopathy Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Giancarlo Castaman
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Barbara Coco
- Hepatology Unit, University Hospital of Pisa, Pisa, Italy
| | - Maria Alice Donati
- Metabolic and Muscular Unit, A. Meyer Children Hospital, Florence, Italy
| | - Renzo Guerrini
- Neuroscience Department, A. Meyer Children Hospital-University of Florence, Florence, Italy
| | - Silvia Linari
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Vittoria Murro
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Paola Parronchi
- Department of Experimental and Clinical Medicine, SOD Immunologia e Terapie Cellulari, Careggi University Hospital, University of Florence, Florence, Italy
| | - Francesca Pochiero
- Metabolic and Muscular Unit, A. Meyer Children Hospital, Florence, Italy
| | - Oliviero Rossi
- Immunuallergology Unit, SOD Immunoallergologia, Careggi University Hospital, Florence, Italy
| | | | - Andrea Sodi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Alessandro Maria Vannucchi
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, Careggi University Hospital, University of Florence, Florence, Italy
| | - Alessio Coi
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| |
Collapse
|
6
|
Huang Q, Li F, Hu H, Fang Z, Gao Z, Xia G, Ng WL, Khodadadi-Jamayran A, Chen T, Deng J, Zhang H, Almonte C, Labbe K, Han H, Geng K, Tang S, Freeman GJ, Li Y, Chen H, Wong KK. Loss of TSC1/TSC2 sensitizes immune checkpoint blockade in non-small cell lung cancer. SCIENCE ADVANCES 2022; 8:eabi9533. [PMID: 35119931 PMCID: PMC8816329 DOI: 10.1126/sciadv.abi9533] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Tuberous sclerosis complex subunit 1 (TSC1) and 2 (TSC2) are frequently mutated in non-small cell lung cancer (NSCLC), however, their effects on antitumor immunity remained unexplored. A CRISPR screening in murine KrasG12D/Trp53-/- (KP) model identified Tsc1 and Tsc2 as potent regulators of programmed cell death ligand 1 (Pd-l1) expression in vitro and sensitivity to anti-programmed cell death receptor 1 (PD-1) treatment in vivo. TSC1 or TSC2 knockout (KO) promoted the transcriptional and membrane expression of PD-L1 in cell lines. TSC2-deficient tumors manifested an inflamed microenvironment in patient samples and The Cancer Genome Atlas dataset. In syngeneic murine models, KP-Tsc2-KO tumors showed notable response to anti-PD-1 antibody treatment, but Tsc2-wild-type tumors did not. Patients with TSC1/TSC2-mutant NSCLC receiving immune checkpoint blockade (ICB) had increased durable clinical benefit and survival. Collectively, TSC1/TSC2 loss defines a distinct subtype of NSCLC characterized as inflamed tumor microenvironment and superior sensitivity to ICB.
Collapse
Affiliation(s)
- Qingyuan Huang
- Department of Thoracic Surgery and State Key
Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center,
Shanghai, China
- Institute of Thoracic Oncology, Fudan University,
Shanghai, China
| | - Fei Li
- Department of Pathology, School of Basic Medical
Sciences, Fudan University, Shanghai, China
- Corresponding author. (H.C.); (K.-K.W.);
(F.L.)
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Zhaoyuan Fang
- State Key Laboratory of Cell Biology, Innovation
Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell
Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of
Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhendong Gao
- Department of Thoracic Surgery and State Key
Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center,
Shanghai, China
- Institute of Thoracic Oncology, Fudan University,
Shanghai, China
| | - Guozhan Xia
- Department of Thoracic Surgery and State Key
Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center,
Shanghai, China
- Institute of Thoracic Oncology, Fudan University,
Shanghai, China
| | - Wai-Lung Ng
- School of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories and Genome
Technology Center, Division of Advanced Research Technologies, New York
University Langone Medical Center, New York, NY, USA
| | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Jiehui Deng
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Hua Zhang
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Christina Almonte
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Kristen Labbe
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Han Han
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Ke Geng
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Sittinon Tang
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer
Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuan Li
- Department of Pathology, Fudan University Shanghai
Cancer Center, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery and State Key
Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center,
Shanghai, China
- Institute of Thoracic Oncology, Fudan University,
Shanghai, China
- Corresponding author. (H.C.); (K.-K.W.);
(F.L.)
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York
University Grossman School of Medicine, NYU Langone Health, New York, NY,
USA
- Corresponding author. (H.C.); (K.-K.W.);
(F.L.)
| |
Collapse
|
7
|
Fidalgo da Silva E, Fong J, Roye-Azar A, Nadi A, Drouillard C, Pillon A, Porter LA. Beyond Protein Synthesis; The Multifaceted Roles of Tuberin in Cell Cycle Regulation. Front Cell Dev Biol 2022; 9:806521. [PMID: 35096832 PMCID: PMC8795880 DOI: 10.3389/fcell.2021.806521] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of cells to sense diverse environmental signals, including nutrient availability and conditions of stress, is critical for both prokaryotes and eukaryotes to mount an appropriate physiological response. While there is a great deal known about the different biochemical pathways that can detect and relay information from the environment, how these signals are integrated to control progression through the cell cycle is still an expanding area of research. Over the past three decades the proteins Tuberin, Hamartin and TBC1D7 have emerged as a large protein complex called the Tuberous Sclerosis Complex. This complex can integrate a wide variety of environmental signals to control a host of cell biology events including protein synthesis, cell cycle, protein transport, cell adhesion, autophagy, and cell growth. Worldwide efforts have revealed many molecular pathways which alter Tuberin post-translationally to convey messages to these important pathways, with most of the focus being on the regulation over protein synthesis. Herein we review the literature supporting that the Tuberous Sclerosis Complex plays a critical role in integrating environmental signals with the core cell cycle machinery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - L. A. Porter
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
8
|
Sobiborowicz A, Świtaj T, Teterycz P, Spałek MJ, Szumera-Ciećkiewicz A, Wągrodzki M, Zdzienicki M, Czarnecka AM, Rutkowski P. Feasibility and Long-Term Efficacy of PEComa Treatment-20 Years of Experience. J Clin Med 2021; 10:jcm10102200. [PMID: 34069629 PMCID: PMC8160690 DOI: 10.3390/jcm10102200] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Perivascular epithelioid cell tumors (PEComas) represent a family of rare mesenchymal neoplasms, some of which are malignant. There are no specific management guidelines for PEComas, and factors correlating with the disease course are not well defined. This analysis aimed to describe the outcomes of PEComa patients treated radically, including those treated exclusively in the national reference sarcoma center. The secondary aim of the study was to analyze factors associated with PEComa treatment efficacy. We performed an analysis of 27 patients subsequently treated radically for PEComa between 1999 and 2019 who were in follow-up in the national sarcoma reference center. The proportional-hazards model was used to compare the risk of death. The median age at diagnosis was 45 (21–67) years, and 67% of patients were female. The median follow-up period was 68 months (95% CI: 39–101). At the time of analysis, eleven patients (40.7%) experienced progression of the disease and four (14.8%) died. Surgery in the reference sarcoma center was associated with a longer disease control (log-rank p < 0.001). The 5-year-OS rate was 88% (95% CI: 74–100) for the whole analyzed group. We concluded that PEComa treatment should be managed in reference sarcoma centers by a multidisciplinary tumor board with an experienced surgical team. Microscopically radical resection is associated with a longer disease-free survival. Patients requiring long-term follow-ups as late recurrence may be expected.
Collapse
Affiliation(s)
- Aleksandra Sobiborowicz
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
- Medical Faculty, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Tomasz Świtaj
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
| | - Paweł Teterycz
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
- Departament of Computional Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Mateusz J. Spałek
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology and Laboratory Diagnostics, Maria Skłodowska-Curie Institute—Oncology Center, 02-781 Warsaw, Poland or (A.S.-C.); (M.W.)
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 00-791 Warsaw, Poland
| | - Michał Wągrodzki
- Department of Pathology and Laboratory Diagnostics, Maria Skłodowska-Curie Institute—Oncology Center, 02-781 Warsaw, Poland or (A.S.-C.); (M.W.)
| | - Marcin Zdzienicki
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
- Correspondence: or ; Tel.: +48-225-462-455
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone, Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.S.); (T.Ś.); (P.T.); (M.J.S.); (M.Z.); (P.R.)
| |
Collapse
|
9
|
Clements D, Miller S, Johnson SR. Pulmonary Lymphangioleiomyomatosis originates in the pleural mesothelial cell population. Med Hypotheses 2020; 141:109703. [PMID: 32276237 DOI: 10.1016/j.mehy.2020.109703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a cystic lung disease mainly affecting women, in which degradation of the lung parenchyma is associated with a cell of unknown provenance, known as a LAM cell. LAM cells carry TSC2 mutations and can be identified in the lung parenchyma by their expression of both smooth muscle actin and antigens characteristic of melanocytes and melanocytic tumors. The nature of the cell-of-origin of LAM is controversial, and despite continued research effort remains elusive. Further, it has not been possible to culture pulmonary LAM cells in vitro, and current research relies on cells and animal models which may not recapitulate all features of the disease. We noted aberrant expression of melanoma antigens in pleural mesothelial cells in lung tissue from LAM patients, indicating that these cells could be the precursors of parenchymal LAM cells. We hypothesise that loss of tuberin function following TSC2 mutation in the mesothelial cell lineage gives rise to the cell-of-origin of pulmonary LAM (P-LAM), and of other associated conditions commonly noted in LAM patients. The unique properties of mesothelial cells provide a straightforward explanation of the diverse presentation of LAM.
Collapse
Affiliation(s)
- D Clements
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK.
| | - S Miller
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK
| | - S R Johnson
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; National Centre for Lymphangioleiomyomatosis, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
10
|
Rankovic M, Zweckstetter M. Upregulated levels and pathological aggregation of abnormally phosphorylated Tau-protein in children with neurodevelopmental disorders. Neurosci Biobehav Rev 2019; 98:1-9. [DOI: 10.1016/j.neubiorev.2018.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
|
11
|
Blaz J, Barrera-Redondo J, Vázquez-Rosas-Landa M, Canedo-Téxon A, Aguirre von Wobeser E, Carrillo D, Stouthamer R, Eskalen A, Villafán E, Alonso-Sánchez A, Lamelas A, Ibarra-Juarez LA, Pérez-Torres CA, Ibarra-Laclette E. Genomic Signals of Adaptation towards Mutualism and Sociality in Two Ambrosia Beetle Complexes. Life (Basel) 2018; 9:E2. [PMID: 30583535 PMCID: PMC6463014 DOI: 10.3390/life9010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/08/2018] [Accepted: 12/20/2018] [Indexed: 01/03/2023] Open
Abstract
Mutualistic symbiosis and eusociality have developed through gradual evolutionary processes at different times in specific lineages. Like some species of termites and ants, ambrosia beetles have independently evolved a mutualistic nutritional symbiosis with fungi, which has been associated with the evolution of complex social behaviors in some members of this group. We sequenced the transcriptomes of two ambrosia complexes (Euwallacea sp. near fornicatus⁻Fusarium euwallaceae and Xyleborus glabratus⁻Raffaelea lauricola) to find evolutionary signatures associated with mutualism and behavior evolution. We identified signatures of positive selection in genes related to nutrient homeostasis; regulation of gene expression; development and function of the nervous system, which may be involved in diet specialization; behavioral changes; and social evolution in this lineage. Finally, we found convergent changes in evolutionary rates of proteins across lineages with phylogenetically independent origins of sociality and mutualism, suggesting a constrained evolution of conserved genes in social species, and an evolutionary rate acceleration related to changes in selective pressures in mutualistic lineages.
Collapse
Affiliation(s)
- Jazmín Blaz
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| | - Josué Barrera-Redondo
- Departamento de Ecología Evolutiva, Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México 04500, Mexico.
| | | | - Anahí Canedo-Téxon
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| | | | - Daniel Carrillo
- Tropical Research and Education Center, University of Florida, Homestead, FL 33031, USA.
| | - Richard Stouthamer
- Department of Plant Pathology, University of California⁻Riverside, Riverside, CA 92521, USA.
| | - Akif Eskalen
- Department of Plant Pathology, University of California, Davis, CA 95616-8751, USA.
| | - Emanuel Villafán
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| | - Alexandro Alonso-Sánchez
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| | - Araceli Lamelas
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| | - Luis Arturo Ibarra-Juarez
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
- Cátedras CONACyT/Instituto de Ecología A.C., Xalapa, Veracruz 91070, Mexico.
| | - Claudia Anahí Pérez-Torres
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
- Cátedras CONACyT/Instituto de Ecología A.C., Xalapa, Veracruz 91070, Mexico.
| | - Enrique Ibarra-Laclette
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C, Xalapa, Veracruz 91070, Mexico.
| |
Collapse
|
12
|
Seiler M, Peng S, Agrawal AA, Palacino J, Teng T, Zhu P, Smith PG, Buonamici S, Yu L. Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences across 33 Cancer Types. Cell Rep 2018; 23:282-296.e4. [PMID: 29617667 PMCID: PMC5933844 DOI: 10.1016/j.celrep.2018.01.088] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/12/2017] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
Hotspot mutations in splicing factor genes have been recently reported at high frequency in hematological malignancies, suggesting the importance of RNA splicing in cancer. We analyzed whole-exome sequencing data across 33 tumor types in The Cancer Genome Atlas (TCGA), and we identified 119 splicing factor genes with significant non-silent mutation patterns, including mutation over-representation, recurrent loss of function (tumor suppressor-like), or hotspot mutation profile (oncogene-like). Furthermore, RNA sequencing analysis revealed altered splicing events associated with selected splicing factor mutations. In addition, we were able to identify common gene pathway profiles associated with the presence of these mutations. Our analysis suggests that somatic alteration of genes involved in the RNA-splicing process is common in cancer and may represent an underappreciated hallmark of tumorigenesis.
Collapse
Affiliation(s)
- Michael Seiler
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Shouyong Peng
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Anant A Agrawal
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - James Palacino
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Teng Teng
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Ping Zhu
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Peter G Smith
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Silvia Buonamici
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA.
| | - Lihua Yu
- H3 Biomedicine, Inc., 300 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
Stepanova V, Dergilev KV, Holman KR, Parfyonova YV, Tsokolaeva ZI, Teter M, Atochina-Vasserman EN, Volgina A, Zaitsev SV, Lewis SP, Zabozlaev FG, Obraztsova K, Krymskaya VP, Cines DB. Urokinase-type plasminogen activator (uPA) is critical for progression of tuberous sclerosis complex 2 (TSC2)-deficient tumors. J Biol Chem 2017; 292:20528-20543. [PMID: 28972182 DOI: 10.1074/jbc.m117.799593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a fatal lung disease associated with germline or somatic inactivating mutations in tuberous sclerosis complex genes (TSC1 or TSC2). LAM is characterized by neoplastic growth of smooth muscle-α-actin-positive cells that destroy lung parenchyma and by the formation of benign renal neoplasms called angiolipomas. The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin slows progression of these diseases but is not curative and associated with notable toxicity at clinically effective doses, highlighting the need for better understanding LAM's molecular etiology. We report here that LAM lesions and angiomyolipomas overexpress urokinase-type plasminogen activator (uPA). Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts expressed higher uPA levels than their WT counterparts, resulting from the TSC inactivation. Inhibition of uPA expression in Tsc2-null cells reduced the growth and invasiveness and increased susceptibility to apoptosis. However, rapamycin further increased uPA expression in TSC2-null tumor cells and immortalized TSC2-null angiomyolipoma cells, but not in cells with intact TSC. Induction of glucocorticoid receptor signaling or forkhead box (FOXO) 1/3 inhibition abolished the rapamycin-induced uPA expression in TSC-compromised cells. Moreover, rapamycin-enhanced migration of TSC2-null cells was inhibited by the uPA inhibitor UK122, dexamethasone, and a FOXO inhibitor. uPA-knock-out mice developed fewer and smaller TSC2-null lung tumors, and introduction of uPA shRNA in tumor cells or amiloride-induced uPA inhibition reduced tumorigenesis in vivo These findings suggest that interference with the uPA-dependent pathway, when used along with rapamycin, might attenuate LAM progression and potentially other TSC-related disorders.
Collapse
Affiliation(s)
| | - Konstantin V Dergilev
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Kelci R Holman
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Yelena V Parfyonova
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Zoya I Tsokolaeva
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Mimi Teter
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Elena N Atochina-Vasserman
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Alla Volgina
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | - Shane P Lewis
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Fedor G Zabozlaev
- the Department of Pathology, Federal Research Clinical Center Federal Medical and Biological Agency of Russia, Moscow 115682, Russia
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Vera P Krymskaya
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Douglas B Cines
- From the Department of Pathology and Laboratory Medicine and
| |
Collapse
|
14
|
Furlano M, Barreiro Y, Martí T, Facundo C, Ruiz-García C, DaSilva I, Ayasreh N, Cabrera-López C, Ballarín J, Ars E, Torra R. Renal angiomyolipoma bleeding in a patient with TSC2/PKD1 contiguous gene syndrome after 17 years of renal replacement therapy. Nefrologia 2016; 37:87-92. [PMID: 27595512 DOI: 10.1016/j.nefro.2016.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/17/2016] [Accepted: 04/19/2016] [Indexed: 12/15/2022] Open
Abstract
We report the case of a 32-year-old male diagnosed with TSC2/PKD1 contiguous gene syndrome, presenting with tuberous sclerosis (TS) and autosomal dominant polycystic kidney disease simultaneously. He progressed to end-stage renal disease and received a kidney transplant at the age of 12. The native kidneys presented angiomyolipomas (AML), which are common benign tumours in patients with TS. Seventeen years after transplantation, he presented with abdominal pain, anaemia and a retroperitoneal haematoma, the latter caused by renal AML bleeding. Selective embolisation was performed. Our patient could have benefited from the administration of mTOR inhibitors at transplant. This therapy is immunosuppressive and reduces the size of benign tumours in TS as well as the risk of rupture and bleeding. This patient did not receive mTOR inhibitors at the time of the transplant because the relationship between mTOR inhibitors and TS was unknown at that time. This case confirms the persistent risk of renal AML bleeding for both transplanted patients and patients on dialysis. As a result, we would recommend routine check-ups of native kidneys and nephrectomy assessment.
Collapse
Affiliation(s)
- Mónica Furlano
- Enfermedades Renales Hereditarias, Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España
| | - Yaima Barreiro
- Servicio de Hemodiálisis, Avericum S.L., Las Palmas de Gran Canaria, España
| | - Teresa Martí
- Servicio de Radiología, Fundació Puigvert, Barcelona, España
| | - Carme Facundo
- Unidad de Trasplante Renal, Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España
| | - César Ruiz-García
- Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España
| | - Iara DaSilva
- Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España
| | - Nadia Ayasreh
- Enfermedades Renales Hereditarias, Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España
| | - Cristina Cabrera-López
- Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España; Servicio de Nefrología, Hospital Moisés Broggi, Sant Joan Despí, Barcelona
| | - José Ballarín
- Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España
| | - Elisabet Ars
- Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España; Laboratorio de Biología Molecular, Fundació Puigvert, Barcelona, España
| | - Roser Torra
- Enfermedades Renales Hereditarias, Servicio de Nefrología, Fundació Puigvert, Barcelona, España; Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Barcelona, España; Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, España.
| |
Collapse
|
15
|
Sharma M, Kumari K, Kakkar A, Malgulwar P, Pathak P, Suri V, Sarkar C, Chandra S, Faruq M, Gupta R, Saran R. Role of mTOR signaling pathway in the pathogenesis of subependymal giant cell astrocytoma – A study of 28 cases. Neurol India 2016; 64:988-94. [DOI: 10.4103/0028-3886.190274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
16
|
Abstract
Human papillomaviruses (HPVs) infect the epidermis as well as mucous membranes of humans. They are the causative agents of anogenital tract and some oropharyngeal cancers. Infections begin in the basal epithelia, where the viral genome replicates slowly along with its host cell. As infected cells begin to differentiate and progress toward the periphery, the virus drives proliferation in cells that would otherwise be quiescent. To uncouple differentiation from continued cellular propagation, HPVs express two oncoproteins, HPV E6 and E7. This review focuses on high-risk α-HPV E6, which in addition to supporting viral replication has transforming properties. HPV E6 promotes p53 degradation and activates telomerase, but the multifaceted oncoprotein has numerous other functions that are highlighted here.
Collapse
Affiliation(s)
- Nicholas A Wallace
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109;
| | - Denise A Galloway
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109;
| |
Collapse
|
17
|
Frost M, Hulbert J. Clinical management of tuberous sclerosis complex over the lifetime of a patient. PEDIATRIC HEALTH MEDICINE AND THERAPEUTICS 2015; 6:139-146. [PMID: 29388579 PMCID: PMC5683262 DOI: 10.2147/phmt.s67342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tuberous sclerosis complex (TSC) is a genetic disorder characterized by nonmalignant tumors (hamartomas) that can occur in various organ systems, including the brain, kidneys, lungs, skin, eyes, and heart. Clinical manifestations of TSC can occur at any age, thereby making the diagnosis difficult. No typical disease presentation is known, and the clinical presentation usually differs between pediatric and adult patients. Furthermore, variable penetrance of the genetic mutation causes a range of disease severity from very mild to severe, and affected individuals can go undetected for years because many of the clinical manifestations of TSC lack specificity. Once a diagnosis is made, TSC management strategies should be tailored to address the symptoms and risks most relevant to the age of the patient. Improved understanding of the genetic basis of TSC and of the central issue of mTOR overactivation has led to use of pharmacotherapies such as the mTOR inhibitors everolimus and sirolimus in the treatment of TSC disease. In Phase II and III studies, everolimus has demonstrated efficacy and safety in the treatment of both brain (subependymal giant cell astrocytoma) and renal (angiomyolipoma) manifestations associated with TSC. It is important to bear in mind that TSC is a lifelong condition, and for those diagnosed as children, a continuum of care will be needed as they transition from pediatric to adult health services. Clearly identifying the likely differences among diagnosis, monitoring, and management of pediatric and adult patients with TSC is an important step in enabling efficiencies to be maximized without compromising the care provided to patients.
Collapse
|
18
|
Luo R, Cai Q, Mu D. A Chinese tuberous sclerosis complex family and a novel tuberous sclerosis complex-2 mutation. Chin Med J (Engl) 2015; 128:128-30. [PMID: 25563326 PMCID: PMC4837808 DOI: 10.4103/0366-6999.147860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
| | | | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital; Key Laboratory of Obstetric and Gynecologic and Pediatric Dieases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
19
|
Park IJ, Yang WK, Nam SH, Hong J, Yang KR, Kim J, Kim SS, Choe W, Kang I, Ha J. Cryptotanshinone induces G1 cell cycle arrest and autophagic cell death by activating the AMP-activated protein kinase signal pathway in HepG2 hepatoma. Apoptosis 2015; 19:615-28. [PMID: 24173372 DOI: 10.1007/s10495-013-0929-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AMP-activated protein kinase (AMPK) performs a pivotal function in energy homeostasis via the monitoring of intracellular energy status. Once activated under the various metabolic stress conditions, AMPK regulates a multitude of metabolic pathways to balance cellular energy. In addition, AMPK also induces cell cycle arrest or apoptosis through several tumor suppressors including LKB1, TSC2, and p53. LKB1 is a direct upstream kinase of AMPK, while TSC2 and p53 are direct substrates of AMPK. Therefore, it is expected that activators of AMPK signal pathway might be useful for treatment or prevention of cancer. In the present study, we report that cryptotanshinone, a natural compound isolated from Salvia miltiorrhiza, robustly activated AMPK signaling pathway, including LKB1, p53, TSC2, thereby leading to suppression of mTORC1 in a number of LKB1-expressing cancer cells including HepG2 human hepatoma, but not in LKB1-deficient cancer cells. Cryptotanshinone induced HepG2 cell cycle arrest at the G1 phase in an AMPK-dependent manner, and a portion of cells underwent apoptosis as a result of long-term treatment. It also induced autophagic HepG2 cell death in an AMPK-dependent manner. Cryptotanshinone significantly attenuated tumor growth in an HCT116 cancer xenograft in vivo model, with a substantial activation of AMPK signal pathways. Collectively, we demonstrate for the first time that cryptotanshinone harbors the therapeutic potential for the treatment of cancer through AMPK activation.
Collapse
Affiliation(s)
- In-Ja Park
- Department of Biochemistry and Molecular Biology, Medical Research Center for Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Camacho L, Guerrero P, Marchetti D. MicroRNA and protein profiling of brain metastasis competent cell-derived exosomes. PLoS One 2013; 8:e73790. [PMID: 24066071 PMCID: PMC3774795 DOI: 10.1371/journal.pone.0073790] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/25/2013] [Indexed: 01/07/2023] Open
Abstract
Exosomes are small membrane vesicles released by most cell types including tumor cells. The intercellular exchange of proteins and genetic material via exosomes is a potentially effective approach for cell-to-cell communication and it may perform multiple functions aiding to tumor survival and metastasis. We investigated microRNA and protein profiles of brain metastatic (BM) versus non-brain metastatic (non-BM) cell-derived exosomes. We studied the cargo of exosomes isolated from brain-tropic 70W, MDA-MB-231BR, and circulating tumor cell brain metastasis-selected markers (CTC1BMSM) variants, and compared them with parental non-BM MeWo, MDA-MB-231P and CTC1P cells, respectively. By performing microRNA PCR array we identified one up-regulated (miR-210) and two down-regulated miRNAs (miR-19a and miR-29c) in BM versus non-BM exosomes. Second, we analyzed the proteomic content of cells and exosomes isolated from these six cell lines, and detected high expression of proteins implicated in cell communication, cell cycle, and in key cancer invasion and metastasis pathways. Third, we show that BM cell-derived exosomes can be internalized by non-BM cells and that they effectively transport their cargo into cells, resulting in increased cell adhesive and invasive potencies. These results provide a strong rationale for additional investigations of exosomal proteins and miRNAs towards more profound understandings of exosome roles in brain metastasis biogenesis, and for the discovery and application of non-invasive biomarkers for new therapies combating brain metastasis.
Collapse
Affiliation(s)
- Laura Camacho
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Paola Guerrero
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dario Marchetti
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Departments of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
21
|
You J, Liu H, Fu X, Chen M, Niu G, Tian H, Zhang F. Two novel TSC2 mutations in Chinese patients with tuberous sclerosis complex. Indian J Dermatol Venereol Leprol 2013; 79:104-5. [PMID: 23254740 DOI: 10.4103/0378-6323.104680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
22
|
Wang GX, Wang DW, Yi CY, Qu JS, Wang YL. Mutational analyses of the TSC1 and TSC2 genes in cases of tuberous sclerosis complex in Chinese Han children. GENETICS AND MOLECULAR RESEARCH 2013; 12:1168-75. [PMID: 23661441 DOI: 10.4238/2013.april.12.3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant neurogenetic disorder characterized by hamartomas in multiple organs and is caused by a wide spectrum of mutations in 1 of 2 causative genes (TSC1 or TSC2). Here, we present mutational analyses of the TSC1 and TSC2 genes in 4 cases of TSC in Chinese Han children, including 2 familial and 2 sporadic cases, using PCR and DNA sequencing of the entire coding region as well as exon-intron boundaries of these genes. Three mutations were identified in the TSC2 gene. Of these mutations, 2 mutations (c.3312-3313delGA and c.45delT) were novel, and the 3rd mutation (c.5238-5255del) was previously reported in Chinese Han and other populations. These mutations were not present in healthy family members or in 100 unrelated normal controls. The identification of these mutations in this study further expands the spectrum of known TSC2 gene mutations and contributes to prenatal molecular diagnosis and preimplantation genetic testing of TSC.
Collapse
Affiliation(s)
- G-X Wang
- Department of Paediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | | | | | | | | |
Collapse
|
23
|
Abstract
Cardiac tumours in infancy are rare and are mostly benign with rhabdomyomas, fibromas and teratomas accounting for the majority. The presentation depends on size and location of the mass as they tend to cause cavity obstruction or arrhythmias. Most rhabdomyomas tend to regress spontaneously but fibromas and teratomas generally require surgical intervention for severe haemodynamic or arrhythmic complications. Other relatively rare cardiac tumours too are discussed along with an Indian perspective.
Collapse
Affiliation(s)
- O P Yadava
- National Heart Institute, New Delhi, India.
| |
Collapse
|
24
|
Liu F, Lunsford EP, Tong J, Ashitate Y, Gibbs SL, Yu J, Choi HS, Henske EP, Frangioni JV. Real-time monitoring of tumorigenesis, dissemination, & drug response in a preclinical model of lymphangioleiomyomatosis/tuberous sclerosis complex. PLoS One 2012; 7:e38589. [PMID: 22719903 PMCID: PMC3376142 DOI: 10.1371/journal.pone.0038589] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 05/09/2012] [Indexed: 12/13/2022] Open
Abstract
Background TSC2-deficient cells can proliferate in the lungs, kidneys, and other organs causing devastating progressive multisystem disorders such as lymphangioleiomyomatosis (LAM) and tuberous sclerosis complex (TSC). Preclinical models utilizing LAM patient-derived cells have been difficult to establish. We developed a novel animal model system to study the molecular mechanisms of TSC/LAM pathogenesis and tumorigenesis and provide a platform for drug testing. Methods and Findings TSC2-deficient human cells, derived from the angiomyolipoma of a LAM patient, were engineered to co-express both sodium-iodide symporter (NIS) and green fluorescent protein (GFP). Cells were inoculated intraparenchymally, intravenously, or intratracheally into athymic NCr nu/nu mice and cells were tracked and quantified using single photon emission computed tomography (SPECT) and computed tomography (CT). Surprisingly, TSC2-deficient cells administered intratracheally resulted in rapid dissemination to lymph node basins throughout the body, and histopathological changes in the lung consistent with LAM. Estrogen was found to be permissive for tumor growth and dissemination. Rapamycin inhibited tumor growth, but tumors regrew after the drug treatment was withdrawn. Conclusions We generated homogeneous NIS/GFP co-expressing TSC2-deficient, patient-derived cells that can proliferate and migrate in vivo after intratracheal instillation. Although the animal model we describe has some limitations, we demonstrate that systemic tumors formed from TSC2-deficient cells can be monitored and quantified noninvasively over time using SPECT/CT, thus providing a much needed model system for in vivo drug testing and mechanistic studies of TSC2-deficient cells and their related clinical syndromes.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Blotting, Western
- Cell Transformation, Neoplastic
- Disease Models, Animal
- Green Fluorescent Proteins/genetics
- Humans
- Lymphangioleiomyomatosis/drug therapy
- Lymphangioleiomyomatosis/pathology
- Mice
- Mice, Nude
- Microscopy, Fluorescence
- Monitoring, Physiologic/methods
- Tomography, Emission-Computed, Single-Photon
- Tomography, X-Ray Computed
- Tuberous Sclerosis/drug therapy
- Tuberous Sclerosis/pathology
- Tuberous Sclerosis Complex 2 Protein
- Tumor Cells, Cultured
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Fangbing Liu
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elaine P. Lunsford
- Longwood Small Animal Imaging Facility, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jingli Tong
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yoshitomo Ashitate
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Summer L. Gibbs
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jane Yu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hak Soo Choi
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elizabeth P. Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - John V. Frangioni
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
25
|
Chévere-Torres I, Kaphzan H, Bhattacharya A, Kang A, Maki JM, Gambello MJ, Arbiser JL, Santini E, Klann E. Metabotropic glutamate receptor-dependent long-term depression is impaired due to elevated ERK signaling in the ΔRG mouse model of tuberous sclerosis complex. Neurobiol Dis 2011; 45:1101-10. [PMID: 22198573 DOI: 10.1016/j.nbd.2011.12.028] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 11/08/2011] [Accepted: 12/08/2011] [Indexed: 11/28/2022] Open
Abstract
Tuberous sclerosis complex (TSC) and fragile X syndrome (FXS) are caused by mutations in negative regulators of translation. FXS model mice exhibit enhanced metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). Therefore, we hypothesized that a mouse model of TSC, ΔRG transgenic mice, also would exhibit enhanced mGluR-LTD. We measured the impact of TSC2-GAP mutations on the mTORC1 and ERK signaling pathways and protein synthesis-dependent hippocampal synaptic plasticity in ΔRG transgenic mice. These mice express a dominant/negative TSC2 that binds to TSC1, but has a deletion and substitution mutation in its GAP-domain, resulting in inactivation of the complex. Consistent with previous studies of several other lines of TSC model mice, we observed elevated S6 phosphorylation in the brains of ΔRG mice, suggesting upregulated translation. Surprisingly, mGluR-LTD was not enhanced, but rather was impaired in the ΔRG transgenic mice, indicating that TSC and FXS have divergent synaptic plasticity phenotypes. Similar to patients with TSC, the ΔRG transgenic mice exhibit elevated ERK signaling. Moreover, the mGluR-LTD impairment displayed by the ΔRG transgenic mice was rescued with the MEK-ERK inhibitor U0126. Our results suggest that the mGluR-LTD impairment observed in ΔRG mice involves aberrant TSC1/2-ERK signaling.
Collapse
|
26
|
Cohen JD, Gard JMC, Nagle RB, Dietrich JD, Monks TJ, Lau SS. ERK crosstalks with 4EBP1 to activate cyclin D1 translation during quinol-thioether-induced tuberous sclerosis renal cell carcinoma. Toxicol Sci 2011; 124:75-87. [PMID: 21813464 DOI: 10.1093/toxsci/kfr203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase signaling cascades have been implicated in a number of human cancers. The tumor suppressor gene tuberous sclerosis-2 (Tsc-2) functions as a negative regulator of mTOR. Critical proteins in both pathways are activated following treatment of Eker rats (Tsc-2(EK/+)) with the nephrocarcinogen 2,3,5-tris-(glutathion-S-yl)hydroquinone (TGHQ), which also results in loss of the wild-type allele of Tsc-2 in renal preneoplastic lesions and tumors. Western blot analysis of kidney tumors formed following treatment of Tsc-2(EK/+) rats with TGHQ for 8 months revealed increases in B-Raf, Raf-1, pERK, cyclin D1, 4EBP1, and p-4EBP1-Ser65, -Thr70, and -Thr37/46 expression. Similar changes are observed following TGHQ-mediated transformation of primary renal epithelial cells derived from Tsc-2(EK/+) rats (quinol-thioether rat renal epithelial [QTRRE] cells) that are also null for tuberin. These cells exhibit high ERK, B-Raf, and Raf-1 kinase activity and increased expression of all p-4EBP1s and cyclin D1. Treatment of the QTRRE cells with the Raf kinase inhibitor, sorafenib, or the MEK1/2 kinase inhibitor, PD 98059, produced a significant decrease in the protein expression of all p-4EBP1s and cyclin D1. Following siRNA knockdown of Raf-1, Western blot analysis revealed a significant decrease in Raf-1, cyclin D1, and all p-4EBP1 forms noted above. In contrast, siRNA knockdown of B-Raf resulted in a nominal change in these proteins. The data indicate that Raf-1/MEK/ERK participates in crosstalk with 4EBP1, which represents a novel pathway interaction leading to increased protein synthesis, cell growth, and kidney tumor formation.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Southwest Environmental Health Sciences Center, Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA
| | | | | | | | | | | |
Collapse
|
27
|
Impaired social interactions and motor learning skills in tuberous sclerosis complex model mice expressing a dominant/negative form of tuberin. Neurobiol Dis 2011; 45:156-64. [PMID: 21827857 DOI: 10.1016/j.nbd.2011.07.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 06/23/2011] [Accepted: 07/23/2011] [Indexed: 11/20/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disorder characterized by the development of hamartomas in multiple organs. Neurological manifestation includes cortical dysplasia, epilepsy, and cognitive deficits such as mental impairment and autism. We measured the impact of TSC2-GAP mutations on cognitive processes and behavior in, ΔRG transgenic mice that express a dominant/negative TSC2 that binds to TSC1, but has mutations affecting its GAP domain and its rabaptin-5 binding motif, resulting in inactivation of the TSC1/2 complex. We performed a behavioral characterization of the ΔRG transgenic mice and found that they display mild, but significant impairments in social behavior and rotarod motor learning. These findings suggest that the ΔRG transgenic mice recapitulate some behavioral abnormalities observed in human TSC patients.
Collapse
|
28
|
Goncharova EA, Lim PN, Chisolm A, Fogle HW, Taylor JH, Goncharov DA, Eszterhas A, Panettieri RA, Krymskaya VP. Interferons modulate mitogen-induced protein synthesis in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2010; 299:L25-35. [PMID: 20382746 PMCID: PMC2904093 DOI: 10.1152/ajplung.00228.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 04/06/2010] [Indexed: 01/10/2023] Open
Abstract
Severe asthma is characterized by increased airway smooth muscle (ASM) mass due, in part, to ASM cell growth and contractile protein expression associated with increased protein synthesis. Little is known regarding the combined effects of mitogens and interferons on ASM cytosolic protein synthesis. We demonstrate that human ASM mitogens including PDGF, EGF, and thrombin stimulate protein synthesis. Surprisingly, pleiotropic cytokines IFN-beta and IFN-gamma, which inhibit ASM proliferation, also increased cytosolic protein content in ASM cells. Thus IFN-beta alone significantly increased protein synthesis by 1.62 +/- 0.09-fold that was further enhanced by EGF to 2.52 +/- 0.17-fold. IFN-gamma alone also stimulated protein synthesis by 1.91 +/- 0.15-fold; treatment of cells with PDGF, EGF, and thrombin in the presence of IFN-gamma stimulated protein synthesis by 2.24 +/- 0.3-, 1.25 +/- 0.17-, and 2.67 +/- 0.34-fold, respectively, compared with growth factors alone. The mammalian target of rapamycin (mTOR)/S6 kinase 1 (S6K1) inhibition with rapamycin inhibited IFN- and EGF-induced protein synthesis, suggesting that IFN-induced protein synthesis is modulated by mTOR/S6K1 activation. Furthermore, overexpression of tumor suppressor protein tuberous sclerosis complex 2 (TSC2), which is an upstream negative regulator of mTOR/S6K1 signaling, also inhibited mitogen-induced protein synthesis in ASM cells. IFN-beta and IFN-gamma stimulated miR143/145 microRNA expression and increased SM alpha-actin accumulation but had little effect on ASM cell size. In contrast, EGF increased ASM cell size but had little effect on miR143/145 expression. Our data demonstrate that both IFNs and mitogens stimulate protein synthesis but have differential effects on cell size and contractile protein expression and suggest that combined effects of IFNs and mitogens may contribute to ASM cell growth, contractile protein expression, and ASM remodeling in asthma.
Collapse
Affiliation(s)
- Elena A Goncharova
- Pulmonary, Allergy, and Critical Care Division, Airway Biology Initiative, Department of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pilot-Storck F, Chopin E, Rual JF, Baudot A, Dobrokhotov P, Robinson-Rechavi M, Brun C, Cusick ME, Hill DE, Schaeffer L, Vidal M, Goillot E. Interactome mapping of the phosphatidylinositol 3-kinase-mammalian target of rapamycin pathway identifies deformed epidermal autoregulatory factor-1 as a new glycogen synthase kinase-3 interactor. Mol Cell Proteomics 2010; 9:1578-93. [PMID: 20368287 DOI: 10.1074/mcp.m900568-mcp200] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The phosphatidylinositol 3-kinase-mammalian target of rapamycin (PI3K-mTOR) pathway plays pivotal roles in cell survival, growth, and proliferation downstream of growth factors. Its perturbations are associated with cancer progression, type 2 diabetes, and neurological disorders. To better understand the mechanisms of action and regulation of this pathway, we initiated a large scale yeast two-hybrid screen for 33 components of the PI3K-mTOR pathway. Identification of 67 new interactions was followed by validation by co-affinity purification and exhaustive literature curation of existing information. We provide a nearly complete, functionally annotated interactome of 802 interactions for the PI3K-mTOR pathway. Our screen revealed a predominant place for glycogen synthase kinase-3 (GSK3) A and B and the AMP-activated protein kinase. In particular, we identified the deformed epidermal autoregulatory factor-1 (DEAF1) transcription factor as an interactor and in vitro substrate of GSK3A and GSK3B. Moreover, GSK3 inhibitors increased DEAF1 transcriptional activity on the 5-HT1A serotonin receptor promoter. We propose that DEAF1 may represent a therapeutic target of lithium and other GSK3 inhibitors used in bipolar disease and depression.
Collapse
Affiliation(s)
- Fanny Pilot-Storck
- UMR5239 Laboratoire de Biologie Moléculaire de la Cellule, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Guo L, Ying W, Zhang J, Yuan Y, Qian X, Wang J, Yang X, He F. Tandem affinity purification and identification of the human TSC1 protein complex. Acta Biochim Biophys Sin (Shanghai) 2010; 42:266-73. [PMID: 20383465 DOI: 10.1093/abbs/gmq014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mutations in the TSC1 and TSC2 genes lead to tuberous sclerosis complex (TSC), which is characterized clinically by mental retardation, epilepsy, and benign tumors affecting multiple tissues. Numerous components of the TSC protein complex remain uncharacterized. Here we report the purification of the TSC1 complex under physiological conditions using a proteomic strategy. We purified the TSC1 protein complex using a tandem affinity purification method and identified a protein complex containing 139 components. Two known binding proteins of TSC1 (TSC2 and DOCK7) were identified along with other new potential partners, which cover reported and novel TSC1 functional categories. Bioinformatics and biochemical methods were used to evaluate the observed protein-protein interactions. A comparative analysis with a published expression proteomics/genomics study of TSC1 revealed more than 20 common candidates that might be functionally relevant. The data set provides new directions in which to expand our knowledge of the functions of TSC1 and the mechanisms of TSC. The results are highly reliable, which is reflected by the identification of a few reported partners of TSC1 and many TSC1/2-regulated proteins. Interestingly, many new functional categories were identified, such as DNA repair, which provide novel hints to the function of TSC1. Moreover, a few neuronal disease-related proteins that might regulate the normal functions of neurons were identified. Thus, the results suggest that many of the new interactions should be biologically significance. It will be interesting to further investigate the regulatory mechanisms of these components.
Collapse
Affiliation(s)
- Longhua Guo
- Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Abstract
Lymphangioleiomyomatosis is a rare pulmonary disease encountered almost exclusively in women of reproductive age. Pulmonary involvement is characterized by multiple thin-walled cysts in the lungs, recurrent pneumothorax, obstructive lung disorders, and progression to chronic respiratory failure over a mean period of 10 years. Certainty of diagnosis requires a lung biopsy, but international criteria have been proposed for a diagnosis without such a biopsy. International recommendations were recently issued for the diagnosis and treatment of lymphangioleiomyomatosis. Treatment is principally symptomatic and relies on the management of bronchial obstruction by bronchodilators; of hypoxemia by oxygen therapy; of pleural complications by pleurodesis, most often surgical; and of renal angiomyolipomas by percutaneous embolization in cases of hemorrhagic risk. Hormone treatment is not recommended. Hopes are high for mTor inhibitors (sirolimus and everolimus) and treatment trials are currently underway. Lung transplantation must be considered when chronic respiratory failure occurs in patients younger than 60 years.
Collapse
|
33
|
Morel M, Couturier J, Pontcharraud R, Gil R, Fauconneau B, Paccalin M, Page G. Evidence of molecular links between PKR and mTOR signalling pathways in Abeta neurotoxicity: role of p53, Redd1 and TSC2. Neurobiol Dis 2009; 36:151-61. [PMID: 19631745 DOI: 10.1016/j.nbd.2009.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/03/2009] [Accepted: 07/13/2009] [Indexed: 01/13/2023] Open
Abstract
The control of translation is disturbed in Alzheimer's disease (AD). This study analysed the crosslink between the up regulation of double-stranded RNA-dependent-protein kinase (PKR) and the down regulation of mammalian target of rapamycin (mTOR) signalling pathways via p53, the protein Regulated in the Development and DNA damage response 1 (Redd1) and the tuberous sclerosis complex (TSC2) factors in two beta-amyloid peptide (Abeta) neurotoxicity models. In SH-SY5Y cells, Abeta42 induced an increase of P(T451)-PKR and of the ratio p66/(p66+p53) in nuclei and a physical interaction between these proteins. Redd1 gene levels increased and P(T1462)-TSC2 decreased. These disturbances were earlier in rat primary neurons with nuclear co-localization of Redd1 and PKR. The PKR gene silencing in SH-SY5Y cells prevented these alterations. p53, Redd1 and TSC2 could represent the molecular links between PKR and mTOR in Abeta neurotoxicity. PKR could be a critical target in a therapeutic program of AD.
Collapse
Affiliation(s)
- Milena Morel
- Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie BP 199, 86034 Poitiers Cedex, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Morel M, Couturier J, Lafay-Chebassier C, Paccalin M, Page G. PKR, the double stranded RNA-dependent protein kinase as a critical target in Alzheimer's disease. J Cell Mol Med 2009; 13:1476-88. [PMID: 19602051 PMCID: PMC3828860 DOI: 10.1111/j.1582-4934.2009.00849.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Amyloid β-peptide (Aβ) deposits and neurofibrillary tangles are key hallmarks in Alzheimer's disease (AD). Aβ stimulates many signal transducers involved in the neuronal death. However, many mechanisms remain to be elucidated because no definitive therapy of AD exists. Some studies have focused on the control of translation which involves eIF2 and eIF4E, main eukaryotic factors of initiation. The availability of these factors depends on the activation of the double-stranded RNA-dependent protein kinase (PKR) and the mammalian target of rapamycin (mTOR), respectively. mTOR positively regulates the translation while PKR results in a protein synthesis shutdown. Many studies demonstrated that the PKR signalling pathway is up-regulated in cellular and animal models of AD and in the brain of AD patients. Interestingly, our results showed that phosphorylated PKR and eIF2α levels were significantly increased in lymphocytes of AD patients. These modifications were significantly correlated with cognitive and memory test scores performed in AD patients. On the contrary, the mTOR signalling pathway is down-regulated in cellular and animal models of AD. Recently, we showed that p53, regulated protein in development and DNA damage response 1 and tuberous sclerosis complex 2 could represent molecular links between PKR and mTOR signalling pathways. PKR could be an early biomarker of the neuronal death and a critical target for a therapeutic programme in AD.
Collapse
Affiliation(s)
- Milena Morel
- Research Group on Brain Aging (EA 3808) University of Poitiers, Poitiers Cedex, France
| | | | | | | | | |
Collapse
|
35
|
Buller CL, Loberg RD, Fan MH, Zhu Q, Park JL, Vesely E, Inoki K, Guan KL, Brosius FC. A GSK-3/TSC2/mTOR pathway regulates glucose uptake and GLUT1 glucose transporter expression. Am J Physiol Cell Physiol 2008; 295:C836-43. [PMID: 18650261 PMCID: PMC2544442 DOI: 10.1152/ajpcell.00554.2007] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glucose transport is a highly regulated process and is dependent on a variety of signaling events. Glycogen synthase kinase-3 (GSK-3) has been implicated in various aspects of the regulation of glucose transport, but the mechanisms by which GSK-3 activity affects glucose uptake have not been well defined. We report that basal glycogen synthase kinase-3 (GSK-3) activity regulates glucose transport in several cell types. Chronic inhibition of basal GSK-3 activity (8-24 h) in several cell types, including vascular smooth muscle cells, resulted in an approximately twofold increase in glucose uptake due to a similar increase in protein expression of the facilitative glucose transporter 1 (GLUT1). Conversely, expression of a constitutively active form of GSK-3beta resulted in at least a twofold decrease in GLUT1 expression and glucose uptake. Since GSK-3 can inhibit mammalian target of rapamycin (mTOR) signaling via phosphorylation of the tuberous sclerosis complex subunit 2 (TSC2) tumor suppressor, we investigated whether chronic GSK-3 effects on glucose uptake and GLUT1 expression depended on TSC2 phosphorylation and TSC inhibition of mTOR. We found that absence of functional TSC2 resulted in a 1.5-to 3-fold increase in glucose uptake and GLUT1 expression in multiple cell types. These increases in glucose uptake and GLUT1 levels were prevented by inhibition of mTOR with rapamycin. GSK-3 inhibition had no effect on glucose uptake or GLUT1 expression in TSC2 mutant cells, indicating that GSK-3 effects on GLUT1 and glucose uptake were mediated by a TSC2/mTOR-dependent pathway. The effect of GSK-3 inhibition on GLUT1 expression and glucose uptake was restored in TSC2 mutant cells by transfection of a wild-type TSC2 vector, but not by a TSC2 construct with mutated GSK-3 phosphorylation sites. Thus, TSC2 and rapamycin-sensitive mTOR function downstream of GSK-3 to modulate effects of GSK-3 on glucose uptake and GLUT1 expression. GSK-3 therefore suppresses glucose uptake via TSC2 and mTOR and may serve to match energy substrate utilization to cellular growth.
Collapse
Affiliation(s)
- Carolyn L. Buller
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Robert D. Loberg
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ming-Hui Fan
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Qihong Zhu
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - James L. Park
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Eileen Vesely
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ken Inoki
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kun-Liang Guan
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| | - Frank C. Brosius
- Departments of Internal Medicine, Physiology, and Biochemistry; and Life Sciences Institute, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
36
|
Hong F, Larrea MD, Doughty C, Kwiatkowski DJ, Squillace R, Slingerland JM. mTOR-raptor binds and activates SGK1 to regulate p27 phosphorylation. Mol Cell 2008; 30:701-11. [PMID: 18570873 DOI: 10.1016/j.molcel.2008.04.027] [Citation(s) in RCA: 201] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 12/20/2007] [Accepted: 04/28/2008] [Indexed: 11/24/2022]
Abstract
The cell-cycle effects of mTORC1 are not fully understood. We provide evidence that mTOR-raptor phosphorylates SGK1 to modulate p27 function. Cellular mTOR activation, by refeeding of amino acid-deprived cells or by TSC2 shRNA, activated SGK1 and p27 phosphorylation at T157, and both were inhibited by short-term rapamycin treatment and by SGK1 shRNA. mTOR overexpression activated both Akt and SGK1, causing TGF-beta resistance through impaired nuclear import and cytoplasmic accumulation of p27. Rapamycin or raptor shRNA impaired mTOR-driven p70 and SGK1 activation, but not that of Akt, and decreased cytoplasmic p27. mTOR/raptor/SGK1 complexes were detected in cells. mTOR phosphorylated SGK1, but not SGK1-S422A, in vitro. SGK1 phosphorylated p27 in vitro. These data implicate SGK1 as an mTORC1 (mTOR-raptor) substrate. mTOR may promote G1 progression in part through SGK1 activation and deregulate the cell cycle in cancers through both Akt- and SGK-mediated p27 T157 phosphorylation and cytoplasmic p27 mislocalization.
Collapse
Affiliation(s)
- Feng Hong
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
37
|
Goncharova EA, Krymskaya VP. Pulmonary lymphangioleiomyomatosis (LAM): progress and current challenges. J Cell Biochem 2008; 103:369-82. [PMID: 17541983 DOI: 10.1002/jcb.21419] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lymphangioleiomyomatosis (LAM), a rare lung disease, is characterized by the progressive proliferation, migration, and differentiation of smooth muscle (SM)-like LAM cells, which lead to the cystic destruction of the lung parenchyma, obstruction of airways and lymphatics, and loss of pulmonary function. LAM is a disease predominantly affecting women and is exacerbated by pregnancy; only a lung transplant can save the life of a patient. It has been discovered that in LAM, somatic or genetic mutations of tumor suppressor genes tuberous sclerosis complex 1 (TSC1) or TSC2 occur and the TSC1/TSC2 protein complex functions as a negative regulator of the mTOR/S6K1 signaling pathway. These two pivotal observations paved the way for the first rapamycin clinical trial for LAM. The recent discoveries that TSC1/TSC2 complex functions as an integrator of signaling networks regulated by growth factors, insulin, nutrients, and energy heightened the interest regarding this rare disease because the elucidation of disease-relevant mechanisms of LAM will promote a better understanding of other metabolic diseases such as diabetes, cancer, and cardiovascular diseases. In this review, we will summarize the progress made in our understanding of TSC1/TSC2 cellular signaling and the molecular mechanisms of LAM; we will also highlight some of the lesser explored directions and challenges in LAM research.
Collapse
|
38
|
Buchkovich NJ, Yu Y, Zampieri CA, Alwine JC. The TORrid affairs of viruses: effects of mammalian DNA viruses on the PI3K-Akt-mTOR signalling pathway. Nat Rev Microbiol 2008; 6:266-75. [PMID: 18311165 DOI: 10.1038/nrmicro1855] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The successful replication of mammalian DNA viruses requires that they gain control of key cellular signalling pathways that affect broad aspects of cellular macromolecular synthesis, metabolism, growth and survival. The phosphatidylinositol 3'-kinase-Akt-mammalian target of rapamycin (PI3K-Akt-mTOR) pathway is one such pathway. Mammalian DNA viruses have evolved various mechanisms to activate this pathway to obtain the benefits of Akt activation, including the maintenance of translation through the activation of mTOR. In addition, viruses must overcome the inhibition of this pathway that results from the activation of cellular stress responses during viral infection. This Review will discuss the range of mechanisms that mammalian DNA viruses use to activate this pathway, as well as the multiple mechanisms these viruses have evolved to circumvent inhibitory stress signalling.
Collapse
Affiliation(s)
- Nicholas J Buchkovich
- Department of Cancer Biology and Abramson Family Cancer Research Institute, University of Pennsylvania, 314 Biomedical Research Building, 421 Curie Blvd, Philadelphia, 19104-6142 Pennsylvania, USA
| | | | | | | |
Collapse
|
39
|
Goncharova EA, Goncharov DA, Chisolm A, Spaits MS, Lim PN, Cesarone G, Khavin I, Tliba O, Amrani Y, Panettieri RA, Krymskaya VP. Interferon beta augments tuberous sclerosis complex 2 (TSC2)-dependent inhibition of TSC2-null ELT3 and human lymphangioleiomyomatosis-derived cell proliferation. Mol Pharmacol 2008; 73:778-88. [PMID: 18094073 DOI: 10.1124/mol.107.040824] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM), a rare pulmonary disorder, manifests as an abnormal neoplastic growth of smooth muscle-like cells within the lungs. Mutational inactivation of tumor suppressor tuberous sclerosis complex 2 (TSC2) in LAM constitutively activates the mammalian target of rapamycin (mTOR)/p70 S6 kinase 1 (S6K1) signaling pathway and promotes neoplastic growth of LAM cells. In many cell types, type I interferon beta (IFNbeta) inhibits proliferation and induces apoptosis through signal transducers and activators of transcription (STAT)-dependent and STAT-independent signaling pathways, one of which is the mTOR/S6K1 signaling pathway. Our study shows that IFNbeta is expressed in LAM tissues and LAM-derived cell cultures; however, IFNbeta attenuates LAM-derived cell proliferation only at high concentrations, 100 and 1000 U/ml (IC(50) value for IFNbeta is 20 U/ml compared with 1 U/ml for normal human mesenchymal cells, human bronchus fibroblasts and human airway smooth muscle cells). Likewise, IFNbeta only attenuates proliferation of smooth muscle TSC2-null ELT3 cells. Analysis of IFNbeta signaling in LAM cells showed expression of IFNbeta receptor alpha (IFNbetaRalpha) and IFNbetaRbeta, activation and nuclear translocation of STAT1, and phosphorylation of STAT3 and p38 mitogen-activated protein kinase (MAPK), but IFNbeta had little effect on S6K1 activity. However, the re-expression of TSC2 or inhibition of mTOR/S6K1 with rapamycin (sirolimus) augmented antiproliferative effects of IFNbeta in LAM and TSC2-null ELT3 cells. Our study demonstrates that IFNbeta-dependent activation of STATs and p38 MAPK is not sufficient to fully inhibit proliferation of cells with TSC2 dysfunction and that TSC2-dependent inhibition of mTOR/S6K1 cooperates with IFNbeta in inhibiting human LAM and TSC2-null ELT3 cell proliferation.
Collapse
Affiliation(s)
- Elena A Goncharova
- Department of Medicine, University of Pennsylvania, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Abstract
Newborn humans and animals grow at very rapid rates because they use the protein that they eat very efficiency to increase body protein mass. This high efficiency of protein deposition in neonates is largely due to their ability to markedly increase the amount of protein synthesized in their muscles when they eat. This enhanced stimulation of muscle protein synthesis after eating is mediated by the rise in the hormone, insulin, and the rise in amino acids, which are the building blocks of protein. Intracellular signaling components that respond to insulin and amino acids have been identified and these have been shown to be involved in the feeding-induced stimulation of protein synthesis in skeletal muscle of the neonate. The enhanced activation of these intracellular signaling components in neonatal muscle contributes to the high rate of muscle protein synthesis and rapid gain in skeletal muscle mass in newborns.
Collapse
|
42
|
Krymskaya VP. Smooth muscle-like cells in pulmonary lymphangioleiomyomatosis. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2008; 5:119-26. [PMID: 18094094 PMCID: PMC2645298 DOI: 10.1513/pats.200705-061vs] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 06/13/2007] [Indexed: 11/20/2022]
Abstract
Proliferation, migration, and differentiation of smooth muscle (SM)-like lymphangioleiomyomatosis (LAM) cells in the lungs are pathologic manifestations of pulmonary LAM, a rare lung disease predominantly afflicting women and exacerbated by pregnancy. LAM cells form nodules throughout the lung without any predominant localization, but can also form small cell clusters dispersed within lung parenchyma. LAM cells have the appearance of "immature" SM-like cells, irregularly distributed within the nodule in contrast to organized SM cell layers in airways and vasculature. Progressive growth of LAM cells leads to the cystic destruction of the lung parenchyma, obstruction of airways and lymphatics, and loss of pulmonary function. Pathogenetically, LAM occurs from somatic or genetic mutations of tumor suppressor genes tuberous sclerosis complex 1 (TSC1) or TSC2. The TSC1/TSC2 protein complex is an integrator of signaling networks regulated by growth factors, insulin, nutrients, and energy. The observation that the TSC1/TSC2 functions as a negative regulator of the mammalian target of rapamycin (mTOR)/p70 S6 kinase (S6K1) signaling pathway yielded the first rapamycin clinical trial for LAM. Although LAM is a rare lung disease, the elucidation of disease-relevant mechanisms of LAM will provide a better understanding of not only SM-like cell growth, migration, and differentiation in LAM but may also offer insights into other metabolic diseases such as cardiovascular diseases, diabetes, and cancer. In this article, we will summarize the progress made in our understanding of LAM, and we will focus on how dysregulation of TSC1/TSC2 signaling results in abnormal proliferation and migration of SM-like LAM cells.
Collapse
Affiliation(s)
- Vera P Krymskaya
- Pulmonary Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104-3403, USA.
| |
Collapse
|
43
|
Yuan CD, Chang XL, Gao M, Xiao FL, Wu YQ, Liu Q, Shen SK, Liu JL, Du WH, Liu JJ, Yang S, Zhang XJ, Zhou FS, Fang QY. A novel mutation (insTCCG) in the TSC2 gene in a Chinese patient with Tuberous sclerosis complex. J Dermatol Sci 2008; 49:92-4. [PMID: 17888633 DOI: 10.1016/j.jdermsci.2007.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2007] [Revised: 07/24/2007] [Accepted: 08/01/2007] [Indexed: 10/22/2022]
|
44
|
Suryawan A, Orellana RA, Nguyen HV, Jeyapalan AS, Fleming JR, Davis TA. Activation by insulin and amino acids of signaling components leading to translation initiation in skeletal muscle of neonatal pigs is developmentally regulated. Am J Physiol Endocrinol Metab 2007; 293:E1597-605. [PMID: 17878222 PMCID: PMC2714663 DOI: 10.1152/ajpendo.00307.2007] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Insulin and amino acids act independently to stimulate protein synthesis in skeletal muscle of neonatal pigs, and the responses decrease with development. The purpose of this study was to compare the separate effects of fed levels of INS and AA on the activation of signaling components leading to translation initiation and how these responses change with development. Overnight-fasted 6- (n = 4/group) and 26-day-old (n = 6/ group) pigs were studied during 1) euinsulinemic-euglycemiceuaminoacidemic conditions (controls), 2) euinsulinemic-euglycemichyperaminoacidemic clamps (AA), and 3) hyperinsulinemic-euglycemic-euaminoacidemic clamps (INS). INS, but not AA, increased the phosphorylation of protein kinase B (PKB) and tuberous sclerosis 2 (TSC2). Both INS and AA increased protein synthesis and the phosphorylation of mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase-1, and eukaryotic initiation factor (eIF)4E-binding protein 1 (4E-BP1), and these responses were higher in 6-day-old compared with 26-day-old pigs. Both INS and AA decreased the binding of 4E-BP1 to eIF4E and increased eIF4E binding to eIF4G; these effects were greater in 6-day-old than in 26-day-old pigs. Neither INS nor AA altered the composition of mTORC1 (raptor, mTOR, and GbetaL) or mTORC2 (rictor, mTOR, and GbetaL) complexes. Furthermore, neither INS, AA, nor age had any effect on the abundance of Rheb and the phosphorylation of AMP-activated protein kinase and eukaryotic elongation factor 2. Our results suggest that the activation by insulin and amino acids of signaling components leading to translation initiation is developmentally regulated and parallels the developmental decline in protein synthesis in skeletal muscle of neonatal pigs.
Collapse
Affiliation(s)
- Agus Suryawan
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, 1100 Bates St., Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
45
|
Goncharova EA, Goncharov DA, Krymskaya VP. Assays for in vitro monitoring of human airway smooth muscle (ASM) and human pulmonary arterial vascular smooth muscle (VSM) cell migration. Nat Protoc 2007; 1:2933-9. [PMID: 17406553 DOI: 10.1038/nprot.2006.434] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Migration of human pulmonary vascular smooth muscle (VSM) cells contributes to vascular remodeling in pulmonary arterial hypertension and atherosclerosis. Evidence also indicates that, in part, migration of airway smooth muscle (ASM) cells may contribute to airway remodeling associated with asthma. Here we describe migration of VSM and ASM cells in vitro using Transwell or Boyden chamber assays. Because dissecting signaling mechanisms regulating cell migration requires molecular approaches, our protocol also describes how to assess migration of transfected VSM and ASM cells. Transwell or Boyden chamber assays can be completed in approximately 8 h and include plating of serum-deprived VSM or ASM cell suspension on membrane precoated with collagen, migration of cells toward chemotactic gradient and visual (Transwell) or digital (Boyden chamber) analysis of membrane. Although the Transwell assay is easy, the Boyden chamber assay requires hands-on experience; however, both assays are reliable cell-based approaches providing valuable information on how chemotactic and inflammatory factors modulate VSM and ASM migration.
Collapse
Affiliation(s)
- Elena A Goncharova
- Department of Medicine, Pulmonary, Allergy & Critical Care Division, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
46
|
Davis TA, Suryawan A, Orellana RA, Nguyen HV, Fiorotto ML. Postnatal ontogeny of skeletal muscle protein synthesis in pigs. J Anim Sci 2007; 86:E13-8. [PMID: 17785597 PMCID: PMC2640319 DOI: 10.2527/jas.2007-0419] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The neonatal period is characterized by rapid growth and elevated rates of synthesis and accretion of skeletal muscle proteins. The fractional rate of muscle protein synthesis is very high at birth and declines rapidly with age. The elevated capacity for muscle protein synthesis in the neonatal pig is driven by the high ribosome content and, together with an increased efficiency of the translation process, promotes accelerated protein synthesis rates. Feeding profoundly stimulates muscle protein synthesis in neonatal pigs and the response decreases with age. The feeding-induced stimulation of muscle protein synthesis is modulated by an enhanced sensitivity to the postprandial increase in insulin and amino acids. The developmental decline in the response to insulin and amino acids parallels a marked decrease in the feeding-induced activation of translation initiation factors that regulate the binding of mRNA to the 40S ribosomal complex. The abundance and activation of many known positive regulators of the nutrient- and insulin-signaling pathways that are involved in translation initiation are high, whereas those of many negative regulators are low in skeletal muscle of younger pigs. Thus, the activation and(or) abundance of the positive regulators, such as the insulin receptor, insulin receptor-substrate-1, phosphoinositide-3 kinase, phosphoinositide-dependent kinase-1, protein kinase B, mammalian target of rapamycin, raptor, ribosomal protein S6 kinase-1, eukaryotic initiation factor (eIF) 4E-binding protein 1, and eIF4E associated with eIF4G, are greater in 7-d-old pigs than in 26-d-old pigs. The activation of negative regulators, including protein tyrosine phosphatase-1B, phosphatase and tensin homologue deleted on chromosome 10, protein phosphatase 2A, and tuberous sclerosis complex 1/2, are lower in 7-d-old pigs than in 26-d-old pigs. Thus, the developmental decline in the stimulation of skeletal muscle protein synthesis by insulin and amino acids is due in part to the developmentally related decrease in the activation of the signaling pathways that lead to translation initiation.
Collapse
Affiliation(s)
- T A Davis
- United States Department of Agriculture, Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston TX 77030, USA.
| | | | | | | | | |
Collapse
|
47
|
Krymskaya VP. Targeting the phosphatidylinositol 3-kinase pathway in airway smooth muscle: rationale and promise. BioDrugs 2007; 21:85-95. [PMID: 17402792 DOI: 10.2165/00063030-200721020-00003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway plays a critical role in regulating cell growth, proliferation, survival, and motility. Structural alterations, e.g. airway remodeling, in asthma and chronic obstructive pulmonary disease (COPD) are associated with increased airway smooth muscle (ASM) cell growth and proliferation due to the frequent stimulation of ASM by inflammatory mediators, contractile agonists, and growth factors. The critical role of the PI3K signaling pathway in regulating ASM cell growth and proliferation is well established. However, recent discovery of the tumor suppressor proteins tuberous sclerosis complex 1 (TSC1) and TSC2, also known as hamartin and tuberin, as downstream effectors of PI3K and upstream regulators of the mammalian target of rapamycin (mTOR) and S6 kinase 1(S6K1) shed a new light on the PI3K signaling cascade in regulating cell growth and proliferation. The activity of TSC1/TSC2 is regulated by growth factors, nutrients, and energy; thus, TSC1/TSC2 serves as a signaling module for protein translational regulation, cell cycle progression, and cell size, which are key events controlling cell growth and proliferation. This article highlights the potential contribution of the PI3K-TSC1/TSC2-mTOR/S6K1 pathway in smooth muscle remodeling. Pharmacologic manipulation of this signaling pathway could have a major impact on treatment of asthma and COPD.
Collapse
Affiliation(s)
- Vera P Krymskaya
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-3403, USA.
| |
Collapse
|
48
|
Uzun O, Wilson DG, Vujanic GM, Parsons JM, De Giovanni JV. Cardiac tumours in children. Orphanet J Rare Dis 2007; 2:11. [PMID: 17331235 PMCID: PMC3225855 DOI: 10.1186/1750-1172-2-11] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 03/01/2007] [Indexed: 01/12/2023] Open
Abstract
Cardiac tumours are benign or malignant neoplasms arising primarily in the inner lining, muscle layer, or the surrounding pericardium of the heart. They can be primary or metastatic. Primary cardiac tumours are rare in paediatric practice with a prevalence of 0.0017 to 0.28 in autopsy series. In contrast, the incidence of cardiac tumours during foetal life has been reported to be approximately 0.14%. The vast majority of primary cardiac tumours in children are benign, whilst approximately 10% are malignant. Secondary malignant tumours are 10-20 times more prevalent than primary malignant tumours. Rhabdomyoma is the most common cardiac tumour during foetal life and childhood. It accounts for more than 60% of all primary cardiac tumours. The frequency and type of cardiac tumours in adults differ from those in children with 75% being benign and 25% being malignant. Myxomas are the most common primary tumours in adults constituting 40% of benign tumours. Sarcomas make up 75% of malignant cardiac masses. Echocardiography, Computing Tomography (CT) and Magnetic Resonance Imaging (MRI) of the heart are the main non-invasive diagnostic tools. Cardiac catheterisation is seldom necessary. Tumour biopsy with histological assessment remains the gold standard for confirmation of the diagnosis. Surgical resection of primary cardiac tumours should be considered to relieve symptoms and mechanical obstruction to blood flow. The outcome of surgical resection in symptomatic, non-myxomatous benign cardiac tumours is favourable. Patients with primary cardiac malignancies may benefit from palliative surgery but this approach should not be recommended for patients with metastatic cardiac tumours. Surgery, chemotherapy and radiotherapy may prolong survival. The prognosis for malignant primary cardiac tumours is generally extremely poor.
Collapse
Affiliation(s)
- Orhan Uzun
- Consultant Paediatric Cardiologist, Department of Paediatric Cardiology, University Hospital Of Wales, Heath Park Cardiff, CF14 4XW, Wales, UK
| | - Dirk G Wilson
- Consultant Paediatric Cardiologist, Department of Paediatric Cardiology, University Hospital Of Wales, Heath Park Cardiff, CF14 4XW, Wales, UK
| | - Gordon M Vujanic
- Consultant Senior Lecturer in Paediatric Pathology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XW, Wales, UK
| | - Jonathan M Parsons
- Consultant Paediatric Cardiologist, Yorkshire Heart Centre, Department Of Paediatric Cardiology, Leeds, UK
| | - Joseph V De Giovanni
- Consultant Paediatric Cardiologist. Birmingham Children's Hospital, Birmingham, UK
| |
Collapse
|
49
|
Abstract
The AMP-activated protein kinase (AMPK) has been referred to as an "energy sensor" because it binds to and is regulated by both AMP and ATP. The binding of AMP to AMPK allows it to be phosphorylated by upstream kinases, resulting in its activation. In contrast, the binding of ATP prevents its activation. AMPK regulates a multitude of metabolic processes that cumulatively function to maintain cellular energy homeostasis through repression of a number of energy-consuming processes with simultaneous enhancement of energy-producing processes. One downstream AMPK target that has been recently identified is the mammalian target of rapamycin (mTOR), a positive effector of cell growth and division. The focus of the present review is to briefly summarize current knowledge concerning the regulation of mTOR signaling by AMPK.
Collapse
Affiliation(s)
- Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
50
|
York B, Lou D, Noonan DJ. Tuberin nuclear localization can be regulated by phosphorylation of its carboxyl terminus. Mol Cancer Res 2007; 4:885-97. [PMID: 17114346 DOI: 10.1158/1541-7786.mcr-06-0056] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tuberin, the tuberous sclerosis 2 (TSC2) gene product, has been identified as a tumor suppressor protein genetically implicated in the pathology of tuberous sclerosis and the female-specific lung disease lymphangioleiomyomatosis. Tuberin and its predominant cytoplasmic binding partner hamartin have been shown to complex with a variety of intracellular signaling regulators and affect the processes of protein translation, cellular proliferation, cellular migration, and cellular transcription. In previous studies, we have presented evidence for tuberin binding to the calcium-dependent intracellular signaling protein calmodulin (CaM), overlap of tuberin CaM binding domain with a binding domain for estrogen receptor alpha, and the phosphorylation-associated nuclear localization of tuberin. In the study presented here, we expand our findings on the mechanism of tuberin nuclear localization to show that the CaM-estrogen receptor-alpha binding domain of tuberin can also serve as a tuberin nuclear localization sequence. Furthermore, we identify an Akt/p90 ribosomal S6 kinase-1 phosphorylation site within the carboxyl terminus of tuberin that can regulate tuberin nuclear localization and significantly affect the ability of tuberin to modulate estrogen genomic signaling events. These findings suggest a link between tuberin nuclear localization and a variety of intracellular signaling events that have direct implications with respect to the role of tuberin in the pathology of tuberous sclerosis and lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Brian York
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone Avenue, Lexington, KY 40536, USA
| | | | | |
Collapse
|