1
|
Fasano G, Petrini S, Bonavolontà V, Paradisi G, Pedalino C, Tartaglia M, Lauri A. Assessment of the FRET-based Teen sensor to monitor ERK activation changes preceding morphological defects in a RASopathy zebrafish model and phenotypic rescue by MEK inhibitor. Mol Med 2024; 30:47. [PMID: 38594640 PMCID: PMC11005195 DOI: 10.1186/s10020-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/12/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND RASopathies are genetic syndromes affecting development and having variable cancer predisposition. These disorders are clinically related and are caused by germline mutations affecting key players and regulators of the RAS-MAPK signaling pathway generally leading to an upregulated ERK activity. Gain-of-function (GOF) mutations in PTPN11, encoding SHP2, a cytosolic protein tyrosine phosphatase positively controlling RAS function, underlie approximately 50% of Noonan syndromes (NS), the most common RASopathy. A different class of these activating mutations occurs as somatic events in childhood leukemias. METHOD Here, we evaluated the application of a FRET-based zebrafish ERK reporter, Teen, and used quantitative FRET protocols to monitor non-physiological RASopathy-associated changes in ERK activation. In a multi-level experimental workflow, we tested the suitability of the Teen reporter to detect pan-embryo ERK activity correlates of morphometric alterations driven by the NS-causing Shp2D61G allele. RESULTS Spectral unmixing- and acceptor photobleaching (AB)-FRET analyses captured pathological ERK activity preceding the manifestation of quantifiable body axes defects, a morphological pillar used to test the strength of SHP2 GoF mutations. Last, the work shows that by multi-modal FRET analysis, we can quantitatively trace back the modulation of ERK phosphorylation obtained by low-dose MEK inhibitor treatment to early development, before the onset of morphological defects. CONCLUSION This work proves the usefulness of FRET imaging protocols on both live and fixed Teen ERK reporter fish to readily monitor and quantify pharmacologically- and genetically-induced ERK activity modulations in early embryos, representing a useful tool in pre-clinical applications targeting RAS-MAPK signaling.
Collapse
Affiliation(s)
- Giulia Fasano
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Stefania Petrini
- Microscopy facility, Research laboratories, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Valeria Bonavolontà
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Graziamaria Paradisi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
- Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, Viterbo, 01100, Italy
| | - Catia Pedalino
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy.
| | - Antonella Lauri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy.
| |
Collapse
|
2
|
Asmamaw MD, Shi XJ, Zhang LR, Liu HM. A comprehensive review of SHP2 and its role in cancer. Cell Oncol 2022; 45:729-753. [PMID: 36066752 DOI: 10.1007/s13402-022-00698-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Src homology 2-containing protein tyrosine phosphatase 2 (SHP2) is a non-receptor protein tyrosine phosphatase ubiquitously expressed mainly in the cytoplasm of several tissues. SHP2 modulates diverse cell signaling events that control metabolism, cell growth, differentiation, cell migration, transcription and oncogenic transformation. It interacts with diverse molecules in the cell, and regulates key signaling events including RAS/ERK, PI3K/AKT, JAK/STAT and PD-1 pathways downstream of several receptor tyrosine kinases (RTKs) upon stimulation by growth factors and cytokines. SHP2 acts as both a phosphatase and a scaffold, and plays prominently oncogenic functions but can be tumor suppressor in a context-dependent manner. It typically acts as a positive regulator of RTKs signaling with some inhibitory functions reported as well. SHP2 expression and activity is regulated by such factors as allosteric autoinhibition, microRNAs, ubiquitination and SUMOylation. Dysregulation of SHP2 expression or activity causes many developmental diseases, and hematological and solid tumors. Moreover, upregulated SHP2 expression or activity also decreases sensitivity of cancer cells to anticancer drugs. SHP2 is now considered as a compelling anticancer drug target and several classes of SHP2 inhibitors with different mode of action are developed with some already in clinical trial phases. Moreover, novel SHP2 substrates and functions are rapidly growing both in cell and cancer. In view of this, we comprehensively and thoroughly reviewed literatures about SHP2 regulatory mechanisms, substrates and binding partners, biological functions, roles in human cancers, and different classes of small molecule inhibitors target this oncoprotein in cancer.
Collapse
Affiliation(s)
- Moges Dessale Asmamaw
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China
| | - Xiao-Jing Shi
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan Province, China. .,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, Henan Province, 450001, People's Republic of China.
| |
Collapse
|
3
|
Stagi S, Ferrari V, Ferrari M, Priolo M, Tartaglia M. Inside the Noonan "universe": Literature review on growth, GH/IGF axis and rhGH treatment: Facts and concerns. Front Endocrinol (Lausanne) 2022; 13:951331. [PMID: 36060964 PMCID: PMC9434367 DOI: 10.3389/fendo.2022.951331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/18/2022] [Indexed: 12/21/2022] Open
Abstract
Noonan syndrome (NS) is a disorder characterized by a typical facial gestalt, congenital heart defects, variable cognitive deficits, skeletal defects, and short stature. NS is caused by germline pathogenic variants in genes coding proteins with a role in the RAS/mitogen-activated protein kinase signaling pathway, and it is typically associated with substantial genetic and clinical complexity and variability. Short stature is a cardinal feature in NS, with evidence indicating that growth hormone (GH) deficiency, partial GH insensitivity, and altered response to insulin-like growth factor I (IGF-1) are contributing events for growth failure in these patients. Decreased IGF-I, together with low/normal responses to GH pharmacological provocation tests, indicating a variable presence of GH deficiency/resistance, in particular in subjects with pathogenic PTPN11 variants, are frequently reported. Nonetheless, short- and long-term studies have demonstrated a consistent and significant increase in height velocity (HV) in NS children and adolescents treated with recombinant human GH (rhGH). While the overall experience with rhGH treatment in NS patients with short stature is reassuring, it is difficult to systematically compare published data due to heterogeneous protocols, potential enrolment bias, the small size of cohorts in many studies, different cohort selection criteria and varying durations of therapy. Furthermore, in most studies, the genetic information is lacking. NS is associated with a higher risk of benign and malignant proliferative disorders and hypertrophic cardiomyopathy, and rhGH treatment may further increase risk in these patients, especially as dosages vary widely. Herein we provide an updated review of aspects related to growth, altered function of the GH/IGF axis and cell response to GH/IGF stimulation, rhGH treatment and its possible adverse events. Given the clinical variability and genetic heterogeneity of NS, treatment with rhGH should be personalized and a conservative approach with judicious surveillance is recommended. Depending on the genotype, an individualized follow-up and close monitoring during rhGH treatments, also focusing on screening for neoplasms, should be considered.
Collapse
Affiliation(s)
- Stefano Stagi
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Vittorio Ferrari
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Marta Ferrari
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Manuela Priolo
- Medical Genetics Unit, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, Reggio Calabria, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
4
|
Kontaridis MI, Roberts AE, Schill L, Schoyer L, Stronach B, Andelfinger G, Aoki Y, Axelrad ME, Bakker A, Bennett AM, Broniscer A, Castel P, Chang CA, Cyganek L, Das TK, den Hertog J, Galperin E, Garg S, Gelb BD, Gordon K, Green T, Gripp KW, Itkin M, Kiuru M, Korf BR, Livingstone JR, López‐Juárez A, Magoulas PL, Mansour S, Milner T, Parker E, Pierpont EI, Plouffe K, Rauen KA, Shankar SP, Smith SB, Stevenson DA, Tartaglia M, Van R, Wagner ME, Ware SM, Zenker M. The seventh international RASopathies symposium: Pathways to a cure-expanding knowledge, enhancing research, and therapeutic discovery. Am J Med Genet A 2022; 188:1915-1927. [PMID: 35266292 PMCID: PMC9117434 DOI: 10.1002/ajmg.a.62716] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 12/15/2022]
Abstract
RASopathies are a group of genetic disorders that are caused by genes that affect the canonical Ras/mitogen-activated protein kinase (MAPK) signaling pathway. Despite tremendous progress in understanding the molecular consequences of these genetic anomalies, little movement has been made in translating these findings to the clinic. This year, the seventh International RASopathies Symposium focused on expanding the research knowledge that we have gained over the years to enhance new discoveries in the field, ones that we hope can lead to effective therapeutic treatments. Indeed, for the first time, research efforts are finally being translated to the clinic, with compassionate use of Ras/MAPK pathway inhibitors for the treatment of RASopathies. This biannual meeting, organized by the RASopathies Network, brought together basic scientists, clinicians, clinician scientists, patients, advocates, and their families, as well as representatives from pharmaceutical companies and the National Institutes of Health. A history of RASopathy gene discovery, identification of new disease genes, and the latest research, both at the bench and in the clinic, were discussed.
Collapse
Affiliation(s)
- Maria I. Kontaridis
- Department of Biomedical Research and Translational MedicineMasonic Medical Research InstituteUticaNew YorkUSA
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Amy E. Roberts
- Department of CardiologyBoston Children's HospitalBostonMassachusettsUSA
- Division of Genetics, Department of PediatricsBoston Children's HospitalBostonMassachusettsUSA
| | - Lisa Schill
- RASopathies Network USAAltadenaCaliforniaUSA
| | | | | | - Gregor Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Saint‐Justine Research CentreUniversité de MontréalMontréalCanada
| | - Yoko Aoki
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Marni E. Axelrad
- Section of Psychology, Department of PediatricsBaylor College of MedicineHoustonTexasUSA
| | | | - Anton M. Bennett
- Yale Center for Molecular and Systems MetabolismYale University School of MedicineNew HavenConnecticutUSA
| | - Alberto Broniscer
- Division of Hematology‐OncologyUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Pau Castel
- Department of Biochemistry and Molecular PharmacologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Caitlin A. Chang
- Department of Medical GeneticsBC Women and Children's HospitalVancouverBritish ColumbiaCanada
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Tirtha K. Das
- Department of Cell, Developmental, and Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jeroen den Hertog
- Hubrecht Institute‐KNAW and University Medical Center UtrechtUtrechtThe Netherlands
- Institute Biology LeidenLeiden UniversityLeidenThe Netherlands
| | - Emilia Galperin
- Department of Molecular and Cellular BiochemistryUniversity of KentuckyLexingtonKentuckyUSA
| | - Shruti Garg
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences CentreUniversity of Manchester & Child & Adolescent Mental Health ServicesManchesterUK
| | - Bruce D. Gelb
- Mindich Child Health and Development Institute and the Departments of Pediatrics and Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kristiana Gordon
- Lymphovascular Medicine, Dermatology DepartmentSt. George's UniversityLondonUK
| | - Tamar Green
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Karen W. Gripp
- Department of GeneticsAI duPont Hospital for ChildrenWilmingtonDelawareUSA
| | - Maxim Itkin
- Center for Lymphatic Disorders, Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Maija Kiuru
- Department of Dermatology, Department of Pathology & Laboratory MedicineUniversity of California DavisSacramentoCaliforniaUSA
| | - Bruce R. Korf
- Department of GeneticsUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | | | - Alejandro López‐Juárez
- Department of Health and Biomedical SciencesUniversity of Texas Rio Grande ValleyTexasUSA
| | - Pilar L. Magoulas
- Department of Molecular and Human Genetics, Baylor College of MedicineTexas Children's HospitalHoustonTexasUSA
| | - Sahar Mansour
- Molecular and Clinical Sciences InstituteSt George's UniversityLondonUK
- South West Thames Regional Genetics ServiceSt George's NHS Foundation TrustLondonUK
| | | | | | - Elizabeth I. Pierpont
- Division of Clinical Behavioral Neuroscience, Department of PediatricsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Katherine A. Rauen
- Department of Pediatrics, Division of Genomic Medicine, MIND InstituteUniversity of California DavisSacramentoCaliforniaUSA
| | - Suma P. Shankar
- Department of Pediatrics, Division of Genomic Medicine, MIND InstituteUniversity of California DavisSacramentoCaliforniaUSA
- Department of Ophthalmology and Vision Science, School of MedicineUniversity of California DavisSacramentoCaliforniaUSA
| | | | - David A. Stevenson
- Department of Pediatrics, Division of Medical GeneticsStanford UniversityStanfordCaliforniaUSA
| | - Marco Tartaglia
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - Richard Van
- Helen Diller Family Comprehensive Cancer CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Morgan E. Wagner
- NCI RAS Initiative, Cancer Research Technology ProgramFrederick National Laboratory for Cancer ResearchFrederickMarylandUSA
| | - Stephanie M. Ware
- Department of Pediatrics, Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Martin Zenker
- Institute of Human Genetics, University HospitalOtto‐von‐Guericke UniversityMagdeburgGermany
| |
Collapse
|
5
|
Wintering A, Dvorak CC, Stieglitz E, Loh ML. Juvenile myelomonocytic leukemia in the molecular era: a clinician's guide to diagnosis, risk stratification, and treatment. Blood Adv 2021; 5:4783-4793. [PMID: 34525182 PMCID: PMC8759142 DOI: 10.1182/bloodadvances.2021005117] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/03/2021] [Indexed: 12/03/2022] Open
Abstract
Juvenile myelomonocytic leukemia is an overlapping myeloproliferative and myelodysplastic disorder of early childhood . It is associated with a spectrum of diverse outcomes ranging from spontaneous resolution in rare patients to transformation to acute myeloid leukemia in others that is generally fatal. This unpredictable clinical course, along with initially descriptive diagnostic criteria, led to decades of productive international research. Next-generation sequencing now permits more accurate molecular diagnoses in nearly all patients. However, curative treatment is still reliant on allogeneic hematopoietic cell transplantation for most patients, and additional advances will be required to improve risk stratification algorithms that distinguish those that can be observed expectantly from others who require swift hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Astrid Wintering
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
| | - Christopher C. Dvorak
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| |
Collapse
|
6
|
D'Amico A, Rosano C, Pannone L, Pinna V, Assunto A, Motta M, Ugga L, Daniele P, Mandile R, Mariniello L, Siano MA, Santoro C, Piluso G, Martinelli S, Strisciuglio P, De Luca A, Tartaglia M, Melis D. Clinical variability of neurofibromatosis 1: A modifying role of cooccurring PTPN11 variants and atypical brain MRI findings. Clin Genet 2021; 100:563-572. [PMID: 34346503 DOI: 10.1111/cge.14040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2023]
Abstract
Neurofibromatosis 1 (NF1) is a disorder characterized by variable expressivity caused by loss-of-function variants in NF1, encoding neurofibromin, a protein negatively controlling RAS signaling. We evaluated whether concurrent variation in proteins functionally linked to neurofibromin contribute to the variable expressivity of NF1. Parallel sequencing of a RASopathy gene panel in 138 individuals with molecularly confirmed clinical diagnosis of NF1 identified missense variants in PTPN11, encoding SHP2, a positive regulator of RAS signaling, in four subjects from three unrelated families. Three subjects were heterozygous for a gain-of-function variant and showed a severe expression of NF1 (developmental delay, multiple cerebral neoplasms and peculiar cortical MRI findings), and features resembling Noonan syndrome (a RASopathy caused by activating variants in PTPN11). Conversely, the fourth subject, who showed an attenuated presentation, carried a previously unreported PTPN11 variant that had a hypomorphic behavior in vitro. Our findings document that functionally relevant PTPN11 variants occur in a small but significant proportion of subjects with NF1 modulating disease presentation, suggesting a model in which the clinical expression of pathogenic NF1 variants is modified by concomitant dysregulation of protein(s) functionally linked to neurofibromin. We also suggest targeting of SHP2 function as an approach to treat evolutive complications of NF1.
Collapse
Affiliation(s)
- Alessandra D'Amico
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Carmen Rosano
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Luca Pannone
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Valentina Pinna
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Antonia Assunto
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Marialetizia Motta
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Paola Daniele
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Roberta Mandile
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Lucio Mariniello
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Maria Anna Siano
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| | - Claudia Santoro
- Referral Centre of Neurofibromatosis, Department of Woman and Child, Specialistic and General Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Pietro Strisciuglio
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Alessandro De Luca
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Daniela Melis
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy.,Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| |
Collapse
|
7
|
Rahal N, Sadi A, Cohen-Barak E, Ziv M, Krausz J, Dodiuk-Gad RP. LEPOARD syndrome: A report of a case with a novel PTPN11 mutation. JAAD Case Rep 2021; 11:57-59. [PMID: 33898683 PMCID: PMC8054096 DOI: 10.1016/j.jdcr.2021.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Nader Rahal
- Department of Dermatology, Ha'emek Medical Center, Afula, Israel
| | - Amir Sadi
- Department of Dermatology, Ha'emek Medical Center, Afula, Israel
| | - Eran Cohen-Barak
- Department of Dermatology, Ha'emek Medical Center, Afula, Israel
| | - Michael Ziv
- Department of Dermatology, Ha'emek Medical Center, Afula, Israel
| | - Judit Krausz
- Department of Pathology, Ha'emek Medical Center, Afula, Israel
| | - Roni P Dodiuk-Gad
- Department of Dermatology, Ha'emek Medical Center, Afula, Israel.,Department of Medicine, University of Toronto, Toronto, Canada.,Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current understanding of germline mutations as they contribute to leukemia development and progression. We also discuss how these new insights may help improve clinical management of germline mutations associated with leukemia. RECENT FINDINGS Germline mutations may represent important initial mutations in the development of leukemia where interaction with somatic mutations provide further hits in leukemic progression. In addition, germline mutations may also contribute to leukemogenesis by impacting bone marrow stem-cell microenvironment and immune cell development and function. SUMMARY Leukemia is characterized by the clonal expansion of malignant cells secondary to somatic or germline mutations in a variety of genes. Understanding somatic mutations that drive leukemogenesis has drastically improved our knowledge of leukemia biology and led to novel therapeutic strategies. Advances have also been made in identifying germline mutations that may affect leukemic development and progression. This review will discuss the biological and clinical relationship of germline mutations with clonal hematopoiesis, bone marrow microenvironment, and immunity in the progression of leukemia.
Collapse
Affiliation(s)
- Kevin Chen
- Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- These authors contributed equally to this work
| | - Rafi Kazi
- Department of Pediatrics, Division of Hematology and Oncology, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
- These authors contributed equally to this work
| | - Christopher C. Porter
- Department of Pediatrics, Division of Hematology and Oncology, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Division of Hematology and Oncology, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
9
|
Li L, Xu X, Du Y, Zhang M, Feng Y, Qian X, Li S, Du T, Peng X, Chen F. ATPR induces acute promyelocytic leukemia cells differentiation and growth arrest by blockade of SHP2/Rho/ROCK1 pathway. Toxicol Appl Pharmacol 2020; 399:115053. [DOI: 10.1016/j.taap.2020.115053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 01/04/2023]
|
10
|
Anselmi M, Calligari P, Hub JS, Tartaglia M, Bocchinfuso G, Stella L. Structural Determinants of Phosphopeptide Binding to the N-Terminal Src Homology 2 Domain of the SHP2 Phosphatase. J Chem Inf Model 2020; 60:3157-3171. [PMID: 32395997 PMCID: PMC8007070 DOI: 10.1021/acs.jcim.0c00307] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 11/28/2022]
Abstract
SH2 domain-containing tyrosine phosphatase 2 (SHP2), encoded by PTPN11, plays a fundamental role in the modulation of several signaling pathways. Germline and somatic mutations in PTPN11 are associated with different rare diseases and hematologic malignancies, and recent studies have individuated SHP2 as a central node in oncogenesis and cancer drug resistance. The SHP2 structure includes two Src homology 2 domains (N-SH2 and C-SH2) followed by a catalytic protein tyrosine phosphatase (PTP) domain. Under basal conditions, the N-SH2 domain blocks the active site, inhibiting phosphatase activity. Association of the N-SH2 domain with binding partners containing short amino acid motifs comprising a phosphotyrosine residue (pY) leads to N-SH2/PTP dissociation and SHP2 activation. Considering the relevance of SHP2 in signaling and disease and the central role of the N-SH2 domain in its allosteric regulation mechanism, we performed microsecond-long molecular dynamics (MD) simulations of the N-SH2 domain complexed to 12 different peptides to define the structural and dynamical features determining the binding affinity and specificity of the domain. Phosphopeptide residues at position -2 to +5, with respect to pY, have significant interactions with the SH2 domain. In addition to the strong interaction of the pY residue with its conserved binding pocket, the complex is stabilized hydrophobically by insertion of residues +1, +3, and +5 in an apolar groove of the domain and interaction of residue -2 with both the pY and a protein surface residue. Additional interactions are provided by hydrogen bonds formed by the backbone of residues -1, +1, +2, and +4. Finally, negatively charged residues at positions +2 and +4 are involved in electrostatic interactions with two lysines (Lys89 and Lys91) specific for the SHP2 N-SH2 domain. Interestingly, the MD simulations illustrated a previously undescribed conformational flexibility of the domain, involving the core β sheet and the loop that closes the pY binding pocket.
Collapse
Affiliation(s)
- Massimiliano Anselmi
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133, Rome, Italy
| | - Paolo Calligari
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133, Rome, Italy
| | - Jochen S. Hub
- Theoretical
Physics and Center for Biophysics, Saarland
University, Campus E2 6, 66123 Saarbrücken, Germany
| | - Marco Tartaglia
- Genetics
and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Gianfranco Bocchinfuso
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133, Rome, Italy
| | - Lorenzo Stella
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133, Rome, Italy
| |
Collapse
|
11
|
Alhumaid MS, Dasouki MJ, Ahmed SO, AbalKhail H, Hagos S, Wakil S, Hashmi SK. Comprehensive Genomic Analysis of Noonan Syndrome and Acute Myeloid Leukemia in Adults: A Review and Future Directions. Acta Haematol 2020; 143:583-593. [PMID: 32541138 DOI: 10.1159/000505715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 12/29/2019] [Indexed: 11/19/2022]
Abstract
Acute myeloid leukemia (AML) in the setting of Noonan syndrome (NS) has been reported before without clear guidelines for treatment or prognosis in these subgroups of patients, most likely due to its rarity and incomplete understanding of the pathogenesis of both diseases. In the current era of next-generation sequencing-based genomic analysis, we can better identify patients with NS with more accurate AML-related prognostic markers. Germline mutations in PTPN11 are the most common cause of NS. Somatic mutations in NPM1 occur frequently in AML. Here, we describe a young adult patient with a novel combined germline PTPN11 and somatic NPM1, IDH1,and BCL6 mutations who presented with fatal AML. In addition, a 50.5-Mb interstitial deletion of 7q21.11-q33 in tumor DNA was detected by chromosomal microarray analysis. While mutations in the transcriptional repressor BCL6 are known to contribute to the pathogenesis of diffuse large B cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL), its novel identification in this patient suggests an expanded role in aggressive AML. The identification of key molecular aberrations including the overexpression of SHP2, which drives leukemogenesis and tumorigenesis, has led to the development of novel investigational targeted SHP2 inhibitors.
Collapse
Affiliation(s)
- Muhned S Alhumaid
- Department of Adult Hematology and Stem Cell Transplant, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- King Saud Medical City, Riyadh, Saudi Arabia
| | - Majed J Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Syed O Ahmed
- Department of Adult Hematology and Stem Cell Transplant, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Halah AbalKhail
- Departments of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Samya Hagos
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Salma Wakil
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shahrukh K Hashmi
- Department of Adult Hematology and Stem Cell Transplant, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia,
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,
| |
Collapse
|
12
|
Xu L, Zhou C, Pan R, Tang J, Wang J, Li B, Huang T, Duan S, Xu C. PTPN11 hypomethylation is associated with gastric cancer progression. Oncol Lett 2020; 19:1693-1700. [PMID: 32194661 PMCID: PMC7039138 DOI: 10.3892/ol.2020.11250] [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: 11/16/2018] [Accepted: 10/14/2019] [Indexed: 11/06/2022] Open
Abstract
Protein tyrosine phosphatase non-receptor type 11 (PTPN11) encodes the tyrosine phosphatase SHP-2 that is overexpressed in gastric cancer (GC). In the present study, the association of PTPN11 methylation levels with the incidence of GC and its correlation with SHP-2 overexpression were investigated. The methylation levels of PTPN11 in tumor and adjacent normal tissues of 112 GC patients were assessed by quantitative methylation specific PCR (qMSP). The Cancer Genome Atlas (TCGA) public database was used to analyze the association between PTPN11 methylation and PTPN11 expression. Survival analyses were conducted in order to evaluate the prognostic value of PTPN11 methylation for GC. The results of the qMSP analysis indicated that the methylation levels of PTPN11 in GC tumor tissues were significantly decreased compared with those noted in the normal adjacent tissues (mean with standard deviation: 40.91±26.33 vs. 51.99±37.37, P=0.007). An inverse correlation between PTPN11 methylation levels and PTPN11 mRNA expression levels (P=4×10-6, r=-0.237) was noted. Subgroup analyses indicated that the association of PTPN11 hypomethylation with the incidence of GC was specific to male subjects (P=0.015), heavy drinking patients (P=0.019), patients with poor tumor differentiation (P=0.010) and patients with tumor node and metastasis (TNM) stage III+IV (P=0.008). Kaplan-Meier analyses and log-rank test suggested that PTPN11 hypomethylation was not associated with GC patient overall survival (P=0.605) and recurrence (P=0.485), although it could predict the recurrence of GC patients up to and including 60 years (≤60, P=0.049). The results indicated that PTPN11 levels were hypomethylated in GC patients. TCGA data analysis suggested that PTPN11 hypomethylation could cause an upregulation in the transcription levels of PTPN11. Although, this may explain the pattern of SHP-2 overexpression in GC, additional studies are required to verify this hypothesis. The association of PTPN11 hypomethylation with GC incidence may be specific to male patients, heavy drinking patients, patients with poor tumor differentiation and patients with TNM stage of III+IV. PTPN11 hypomethylation can be considered a biomarker for the recurrence of GC patients with an age of 60 years or lower.
Collapse
Affiliation(s)
- Lele Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| | - Cong Zhou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Ranran Pan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Junjian Tang
- Department of Vascular Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, P.R. China
| | - Jinzhi Wang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Bin Li
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Tianyi Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| |
Collapse
|
13
|
Tafazoli A, Eshraghi P, Pantaleoni F, Vakili R, Moghaddassian M, Ghahraman M, Muto V, Paolacci S, Golyan FF, Abbaszadegan MR. Novel mutations and their genotype-phenotype correlations in patients with Noonan syndrome, using next-generation sequencing. Adv Med Sci 2018; 63:87-93. [PMID: 28957739 DOI: 10.1016/j.advms.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 06/29/2017] [Accepted: 07/07/2017] [Indexed: 11/18/2022]
Abstract
PURPOSE Noonan Syndrome (NS) is an autosomal dominant disorder with many variable and heterogeneous conditions. The genetic basis for 20-30% of cases is still unknown. This study evaluates Iranian Noonan patients both clinically and genetically for the first time. MATERIALS/METHODS Mutational analysis of PTPN11 gene was performed in 15 Iranian patients, using PCR and Sanger sequencing at phase one. Then, as phase two, Next Generation Sequencing (NGS) in the form of targeted resequencing was utilized for analysis of exons from other related genes. Homology modelling for the novel founded mutations was performed as well. The genotype, phenotype correlation was done according to the molecular findings and clinical features. RESULTS Previously reported mutation (p.N308D) in some patients and a novel mutation (p.D155N) in one of the patients were identified in phase one. After applying NGS methods, known and new variants were found in four patients in other genes, including: CBL (p. V904I), KRAS (p. L53W), SOS1 (p. I1302V), and SOS1 (p. R552G). Structural studies of two deduced novel mutations in related genes revealed deficiencies in the mutated proteins. Following genotype, phenotype correlation, a new pattern of the presence of intellectual disability in two patients was registered. CONCLUSIONS NS shows strong variable expressivity along the high genetic heterogeneity especially in distinct populations and ethnic groups. Also possibly unknown other causative genes may be exist. Obviously, more comprehensive and new technologies like NGS methods are the best choice for detection of molecular defects in patients for genotype, phenotype correlation and disease management.
Collapse
Affiliation(s)
- Alireza Tafazoli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peyman Eshraghi
- Department of Pediatrics, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Francesca Pantaleoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy; Research Center, Genetic and Rare Diseases, Ospedale Pediatrico Bambino Gesù, IRCSS, Rome, Italy
| | - Rahim Vakili
- Department of Pediatrics, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morteza Moghaddassian
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, Faculty of Applied Science and Engineering, University of Toronto, ON, Canada
| | - Martha Ghahraman
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Razavi Cancer Research Center, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | - Valentina Muto
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Paolacci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
14
|
Pannone L, Bocchinfuso G, Flex E, Rossi C, Baldassarre G, Lissewski C, Pantaleoni F, Consoli F, Lepri F, Magliozzi M, Anselmi M, Delle Vigne S, Sorge G, Karaer K, Cuturilo G, Sartorio A, Tinschert S, Accadia M, Digilio MC, Zampino G, De Luca A, Cavé H, Zenker M, Gelb BD, Dallapiccola B, Stella L, Ferrero GB, Martinelli S, Tartaglia M. Structural, Functional, and Clinical Characterization of a Novel PTPN11 Mutation Cluster Underlying Noonan Syndrome. Hum Mutat 2017; 38:451-459. [PMID: 28074573 DOI: 10.1002/humu.23175] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/06/2017] [Indexed: 01/12/2023]
Abstract
Germline mutations in PTPN11, the gene encoding the Src-homology 2 (SH2) domain-containing protein tyrosine phosphatase (SHP2), cause Noonan syndrome (NS), a relatively common, clinically variable, multisystem disorder. Here, we report on the identification of five different PTPN11 missense changes affecting residues Leu261 , Leu262 , and Arg265 in 16 unrelated individuals with clinical diagnosis of NS or with features suggestive for this disorder, specifying a novel disease-causing mutation cluster. Expression of the mutant proteins in HEK293T cells documented their activating role on MAPK signaling. Structural data predicted a gain-of-function role of substitutions at residues Leu262 and Arg265 exerted by disruption of the N-SH2/PTP autoinhibitory interaction. Molecular dynamics simulations suggested a more complex behavior for changes affecting Leu261 , with possible impact on SHP2's catalytic activity/selectivity and proper interaction of the PTP domain with the regulatory SH2 domains. Consistent with that, biochemical data indicated that substitutions at codons 262 and 265 increased the catalytic activity of the phosphatase, while those affecting codon 261 were only moderately activating but impacted substrate specificity. Remarkably, these mutations underlie a relatively mild form of NS characterized by low prevalence of cardiac defects, short stature, and cognitive and behavioral issues, as well as less evident typical facial features.
Collapse
Affiliation(s)
- Luca Pannone
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy.,Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy.,Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Elisabetta Flex
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Cesare Rossi
- Genetica Medica, Policlinico S. Orsola-Malpighi, Bologna, Italy
| | | | - Christina Lissewski
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Francesca Pantaleoni
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Federica Consoli
- Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo, Italy
| | - Francesca Lepri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Monia Magliozzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy.,Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo, Italy
| | - Massimiliano Anselmi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Silvia Delle Vigne
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanni Sorge
- Unità Operativa Complessa di Clinica Pediatrica, Dipartimento di Medicina Clinica e Sperimentale, Università di Catania, Catania, Italy
| | - Kadri Karaer
- Dr. Ersin Arslan Research and Training Hospital, Department of Medical Genetics, Gaziantep, Turkey
| | - Goran Cuturilo
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,University Children's Hospital, Belgrade, Serbia
| | - Alessandro Sartorio
- Istituto Auxologico Italiano, Experimental Laboratory for Auxo-Endocrinological Research, Milan and Verbania, Italy.,Istituto Auxologico Italiano, Division of Auxology, Verbania, Italy
| | - Sigrid Tinschert
- Institute of Clinical Genetics, Technical University of Dresden, Dresden, Germany
| | - Maria Accadia
- Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo, Italy
| | - Maria C Digilio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Istituto di Clinica Pediatrica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro De Luca
- Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo, Italy
| | - Hélène Cavé
- Département de Génétique, Hôpital Robert Debré, Paris, France.,INSERM UMR_S1131, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris-Sorbonne-Cité, Paris, France
| | - Martin Zenker
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Giovanni B Ferrero
- Department of Pediatric and Public Health Sciences, University of Torino, Torino, Italy
| | - Simone Martinelli
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
15
|
Identification of protein tyrosine phosphatase SHP-2 as a new target of perfluoroalkyl acids in HepG2 cells. Arch Toxicol 2016; 91:1697-1707. [DOI: 10.1007/s00204-016-1836-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/24/2016] [Indexed: 01/19/2023]
|
16
|
|
17
|
Ju A, Seo H, Kim H, Park BC, Park SG, Kim JH, Choi HK, Min KH, Cho S. Identification of a Novel SHP-2 Protein Tyrosine Phosphatase Inhibitor. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2014. [DOI: 10.1246/bcsj.20130221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Anna Ju
- College of Pharmacy, Chung-Ang University
| | - Huiyun Seo
- College of Pharmacy, Chung-Ang University
| | - Heemun Kim
- College of Pharmacy, Chung-Ang University
| | - Byoung Chul Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology
| | - Sung Goo Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology
| | - Jeong Hoon Kim
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology
| | | | | | - Sayeon Cho
- College of Pharmacy, Chung-Ang University
| |
Collapse
|
18
|
Qiu W, Wang X, Romanov V, Hutchinson A, Lin A, Ruzanov M, Battaile KP, Pai EF, Neel BG, Chirgadze NY. Structural insights into Noonan/LEOPARD syndrome-related mutants of protein-tyrosine phosphatase SHP2 (PTPN11). BMC STRUCTURAL BIOLOGY 2014; 14:10. [PMID: 24628801 PMCID: PMC4007598 DOI: 10.1186/1472-6807-14-10] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 03/06/2014] [Indexed: 12/19/2022]
Abstract
Background The ubiquitous non-receptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) plays a key role in RAS/ERK signaling downstream of most, if not all growth factors, cytokines and integrins, although its major substrates remain controversial. Mutations in PTPN11 lead to several distinct human diseases. Germ-line PTPN11 mutations cause about 50% of Noonan Syndrome (NS), which is among the most common autosomal dominant disorders. LEOPARD Syndrome (LS) is an acronym for its major syndromic manifestations: multiple Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary stenosis, Abnormalities of genitalia, Retardation of growth, and sensorineural Deafness. Frequently, LS patients have hypertrophic cardiomyopathy, and they might also have an increased risk of neuroblastoma (NS) and acute myeloid leukemia (AML). Consistent with the distinct pathogenesis of NS and LS, different types of PTPN11 mutations cause these disorders. Results Although multiple studies have reported the biochemical and biological consequences of NS- and LS-associated PTPN11 mutations, their structural consequences have not been analyzed fully. Here we report the crystal structures of WT SHP2 and five NS/LS-associated SHP2 mutants. These findings enable direct structural comparisons of the local conformational changes caused by each mutation. Conclusions Our structural analysis agrees with, and provides additional mechanistic insight into, the previously reported catalytic properties of these mutants. The results of our research provide new information regarding the structure-function relationship of this medically important target, and should serve as a solid foundation for structure-based drug discovery programs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Benjamin G Neel
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, M5G 2C4, Canada.
| | | |
Collapse
|
19
|
Praetorius C, Sturm RA, Steingrimsson E. Sun-induced freckling: ephelides and solar lentigines. Pigment Cell Melanoma Res 2014; 27:339-50. [DOI: 10.1111/pcmr.12232] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Christian Praetorius
- Department of Biochemistry and Molecular Biology; Biomedical Center; Faculty of Medicine; University of Iceland; Reykjavik Iceland
| | - Richard A. Sturm
- Melanogenix Group; Institute for Molecular Bioscience; The University of Queensland; Brisbane Qld Australia
- Dermatology Research Centre; School of Medicine; The University of Queensland; Princess Alexandra Hospital; Brisbane Qld Australia
| | - Eirikur Steingrimsson
- Department of Biochemistry and Molecular Biology; Biomedical Center; Faculty of Medicine; University of Iceland; Reykjavik Iceland
| |
Collapse
|
20
|
Dong S, Li FQ, Zhang Q, Lv KZ, Yang HL, Gao Y, Yu JR. Expression and clinical significance of SHP2 in gastric cancer. J Int Med Res 2013; 40:2083-9. [PMID: 23321164 DOI: 10.1177/030006051204000605] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To investigate the expression and clinical significance of the protein tyrosine phosphatase, nonreceptor type 11 (PTPN11 or SHP2) gene, which encodes Src homology 2 domain-containing phosphatase (SHP-2) in gastric cancer. METHODS SHP2 expression was detected by immunohistochemical staining and real-time quantitative reverse transcription-polymerase chain reaction in tissue samples of normal gastric mucosa and different grades of gastric cancer. Correlation between SHP2 expression and standard clinico pathological parameters was analysed. RESULTS Immunohistochemical staining revealed significantly higher rates of SHP2 expression in gastric cancer tissues (72.5%) versus normal gastric mucosa (21.9%). SHP-2 mRNA levels were also significantly higher in gastric cancer tissues versus normal gastric mucosa. SHP2 expression correlated significantly with tumour differentiation, clinical classification and lymph node metastases, but was independent of sex and age. CONCLUSIONS SHP-2 is upregulated in gastric cancer and may be related to the development of gastric cancer. SHP-2 may be a potential prognostic marker of, or a therapeutic target for, gastric cancer.
Collapse
Affiliation(s)
- S Dong
- Department of Gastroenterological Surgery, The First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou, Zhejiang Province, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Kawakami T, Xiao W, Yasudo H, Kawakami Y. Regulation of proliferation, survival, differentiation, and activation by the Signaling Platform for SHP-1 phosphatase. Adv Biol Regul 2013; 52:7-15. [PMID: 21982978 DOI: 10.1016/j.advenzreg.2011.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/05/2011] [Indexed: 12/20/2022]
Affiliation(s)
- Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
22
|
Oncogenic Shp2 disturbs microtubule regulation to cause HDAC6-dependent ERK hyperactivation. Oncogene 2013; 33:2938-46. [DOI: 10.1038/onc.2013.241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/29/2013] [Accepted: 05/03/2013] [Indexed: 02/07/2023]
|
23
|
Nunes-Xavier CE, Martín-Pérez J, Elson A, Pulido R. Protein tyrosine phosphatases as novel targets in breast cancer therapy. Biochim Biophys Acta Rev Cancer 2013; 1836:211-26. [PMID: 23756181 DOI: 10.1016/j.bbcan.2013.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 06/01/2013] [Indexed: 02/07/2023]
Abstract
Breast cancer is linked to hyperactivation of protein tyrosine kinases (PTKs), and recent studies have unveiled that selective tyrosine dephosphorylation by protein tyrosine phosphatases (PTPs) of specific substrates, including PTKs, may activate or inactivate oncogenic pathways in human breast cancer cell growth-related processes. Here, we review the current knowledge on the involvement of PTPs in breast cancer, as major regulators of breast cancer therapy-targeted PTKs, such as HER1/EGFR, HER2/Neu, and Src. The functional interplay between PTKs and PTK-activating or -inactivating PTPs, and its implications in novel breast cancer therapies based on targeting of specific PTPs, are discussed.
Collapse
Affiliation(s)
- Caroline E Nunes-Xavier
- BioCruces Health Research Institute, Hospital de Cruces, Plaza Cruces s/n, 48903 Barakaldo, Spain
| | | | | | | |
Collapse
|
24
|
Cianci P, Tono V, Sala A, Locatelli L, Carta C, Rizzari C, Biondi A, Selicorni A. A boy with Burkitt lymphoma associated with Noonan syndrome due to a mutation in RAF1. Am J Med Genet A 2013; 161A:1401-4. [PMID: 23613113 DOI: 10.1002/ajmg.a.35875] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 01/02/2013] [Indexed: 11/09/2022]
Abstract
This article reports on an association between Burkitt lymphoma and Noonan syndrome (NS) due to a RAF1 gene mutation. The patient was a 7-year-old boy with NS, who was included in the first series reporting the association between Noonan and RAF1, and who later presented with a 2-week history of asymptomatic unilateral tonsillar swelling and ipsilateral cervical lymphadenopathy. Histological and biological examinations of the tonsillar biopsy led to the diagnosis of Burkitt lymphoma. While there is a well-established association between NS and solid cell tumors, this is the first case described in the literature of Burkitt lymphoma in a patient with NS, and adds to the growing list of data supporting neoplasia's association with NS.
Collapse
Affiliation(s)
- Paola Cianci
- Pediatric Genetic Unit, Pediatric Department at Monza Brianza per il Bambino e la sua Mamma (MBBM) Foundation, S.Gerardo Hospital, Monza, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Higher bone marrow LGALS3 expression is an independent unfavorable prognostic factor for overall survival in patients with acute myeloid leukemia. Blood 2013; 121:3172-80. [PMID: 23449638 DOI: 10.1182/blood-2012-07-443762] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alterations of galectin-3 expression are often seen in cancers and may contribute to tumorigenesis, cancer progression, and metastasis. The studies concerning clinical implications of galectin-3 expression in patients with acute myeloid leukemia (AML) are scarce. We investigated the expression of LGALS3, the gene encoding galectin-3, in the bone marrow (BM) mononuclear cells from an original cohort comprising 280 adults with primary non-acute promyelocytic leukemia. Higher LGALS3 expression was closely associated with older age, French-American-British M4/M5 subtypes, CD14 expression on leukemic cells, and PTPN11 mutation, but negatively correlated with CEBPA mutation and FLT3-ITD. Compared with patients with lower LGALS3 expression, those with higher expression had lower complete remission rates, higher primary refractory rates, and shorter overall survival. This result was validated in an independent validation cohort. A scoring system incorporating higher LGALS3 expression and 8 other risk factors, including age, white blood cell count, cytogenetics, and gene mutations, into survival analysis proved to be very useful to stratify patients with AML into different prognostic groups (P < .001). In conclusion, BM LGALS3 expression may serve as a new biomarker to predict clinical outcome in patients with AML, and galectin-3 may serve as a potential therapeutic target in those patients with higher expression of this protein.
Collapse
|
26
|
Tsang YH, Han X, Man WY, Lee N, Poon RYC. Novel functions of the phosphatase SHP2 in the DNA replication and damage checkpoints. PLoS One 2012. [PMID: 23189174 PMCID: PMC3506573 DOI: 10.1371/journal.pone.0049943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Replication stress- and DNA damage-induced cell cycle checkpoints are critical for maintaining genome stability. To identify protein phosphatases involved in the activation and maintenance of the checkpoints, we have carried out RNA interference-based screens with a human phosphatome shRNA library. Several phosphatases, including SHP2 (also called PTPN11) were found to be required for cell survival upon hydroxyurea-induced replicative stress in HeLa cells. More detailed studies revealed that SHP2 was also important for the maintenance of the checkpoint after DNA damage induced by cisplatin or ionizing radiation in HeLa cells. Furthermore, SHP2 was activated after replicative stress and DNA damage. Although depletion of SHP2 resulted in a delay in cyclin E accumulation and an extension of G1 phase, these cell cycle impairments were not responsible for the increase in apoptosis after DNA damage. Depletion of SHP2 impaired CHK1 activation, checkpoint-mediated cell cycle arrest, and DNA repair. These effects could be rescued with a shRNA-resistant SHP2. These results underscore the importance of protein phosphatases in checkpoint control and revealed a novel link between SHP2 and cell cycle checkpoints.
Collapse
Affiliation(s)
- Yiu Huen Tsang
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Xianxian Han
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Nelson Lee
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y. C. Poon
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- * E-mail:
| |
Collapse
|
27
|
Alvira D, Naughton R, Bhatt L, Tedesco S, Landry WD, Cotter TG. Inhibition of protein-tyrosine phosphatase 1B (PTP1B) mediates ubiquitination and degradation of Bcr-Abl protein. J Biol Chem 2011; 286:32313-23. [PMID: 21795709 DOI: 10.1074/jbc.m111.249060] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative disorder characterized at the molecular level by the expression of Bcr-Abl, a chimeric protein with deregulated tyrosine kinase activity. The protein-tyrosine phosphatase 1B (PTP1B) is up-regulated in Bcr-Abl-expressing cells, suggesting a regulatory link between the two proteins. To investigate the interplay between these two proteins, we inhibited the activity of PTP1B in Bcr-Abl-expressing TonB.210 cells by either pharmacological or siRNA means and examined the effects of such inhibition on Bcr-Abl expression and function. Herein we describe a novel mechanism by which the phosphatase activity of PTP1B is required for Bcr-Abl protein stability. Inhibition of PTP1B elicits tyrosine phosphorylation of Bcr-Abl that triggers the degradation of Bcr-Abl through ubiquitination via the lysosomal pathway. The degradation of Bcr-Abl consequently inhibits tyrosine phosphorylation of Bcr-Abl substrates and the downstream production of intracellular reactive oxygen species. Furthermore, PTP1B inhibition reduces cell viability and the IC(50) of the Bcr-Abl inhibitor imatinib mesylate. Degradation of Bcr-Abl via PTP1B inhibition is also observed in human CML cell lines K562 and LAMA-84. These results suggest that inhibition of PTP1B may be a useful strategy to explore in the development of novel therapeutic agents for the treatment of CML, particularly because host drugs currently used in CML such as imatinib focus on inhibiting the kinase activity of Bcr-Abl.
Collapse
Affiliation(s)
- Daniel Alvira
- Tumour Biology Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | |
Collapse
|
28
|
Jongmans MCJ, van der Burgt I, Hoogerbrugge PM, Noordam K, Yntema HG, Nillesen WM, Kuiper RP, Ligtenberg MJL, van Kessel AG, van Krieken JHJM, Kiemeney LALM, Hoogerbrugge N. Cancer risk in patients with Noonan syndrome carrying a PTPN11 mutation. Eur J Hum Genet 2011; 19:870-4. [PMID: 21407260 PMCID: PMC3172922 DOI: 10.1038/ejhg.2011.37] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Noonan syndrome (NS) is characterized by short stature, facial dysmorphisms and congenital heart defects. PTPN11 mutations are the most common cause of NS. Patients with NS have a predisposition for leukemia and certain solid tumors. Data on the incidence of malignancies in NS are lacking. Our objective was to estimate the cancer risk and spectrum in patients with NS carrying a PTPN11 mutation. In addition, we have investigated whether specific PTPN11 mutations result in an increased malignancy risk. We have performed a cohort study among 297 Dutch NS patients with a PTPN11 mutation (mean age 18 years). The cancer histories were collected from the referral forms for DNA diagnostics, and by consulting the Dutch national registry of pathology and the Netherlands Cancer Registry. The reported frequencies of cancer among NS patients were compared with the expected frequencies using population-based incidence rates. In total, 12 patients with NS developed a malignancy, providing a cumulative risk for developing cancer of 23% (95% confidence interval (CI), 8–38%) up to age 55 years, which represents a 3.5-fold (95% CI, 2.0–5.9) increased risk compared with that in the general population. Hematological malignancies occurred most frequently. Two malignancies, not previously observed in NS, were found: a malignant mastocytosis and malignant epithelioid angiosarcoma. No correlation was found between specific PTPN11 mutations and cancer occurrence. In conclusion, this study provides first evidence of an increased risk of cancer in patients with NS and a PTPN11 mutation, compared with that in the general population. Our data do not warrant specific cancer surveillance.
Collapse
Affiliation(s)
- Marjolijn C J Jongmans
- Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Tartaglia M, Gelb BD. Disorders of dysregulated signal traffic through the RAS-MAPK pathway: phenotypic spectrum and molecular mechanisms. Ann N Y Acad Sci 2010; 1214:99-121. [PMID: 20958325 PMCID: PMC3010252 DOI: 10.1111/j.1749-6632.2010.05790.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
RAS GTPases control a major signaling network implicated in several cellular functions, including cell fate determination, proliferation, survival, differentiation, migration, and senescence. Within this network, signal flow through the RAF-MEK-ERK pathway-the first identified mitogen-associated protein kinase (MAPK) cascade-mediates early and late developmental processes controlling morphology determination, organogenesis, synaptic plasticity, and growth. Signaling through the RAS-MAPK cascade is tightly controlled; and its enhanced activation represents a well-known event in oncogenesis. Unexpectedly, in the past few years, inherited dysregulation of this pathway has been recognized as the cause underlying a group of clinically related disorders sharing facial dysmorphism, cardiac defects, reduced postnatal growth, ectodermal anomalies, variable cognitive deficits, and susceptibility to certain malignancies as major features. These disorders are caused by heterozygosity for mutations in genes encoding RAS proteins, regulators of RAS function, modulators of RAS interaction with effectors, or downstream signal transducers. Here, we provide an overview of the phenotypic spectrum associated with germline mutations perturbing RAS-MAPK signaling, the unpredicted molecular mechanisms converging toward the dysregulation of this signaling cascade, and major genotype-phenotype correlations.
Collapse
Affiliation(s)
- Marco Tartaglia
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy.
| | | |
Collapse
|
30
|
Hishida A, Matsuo K, Goto Y, Hamajima N. Genetic predisposition to Helicobacter pylori-induced gastric precancerous conditions. World J Gastrointest Oncol 2010; 2:369-79. [PMID: 21160888 PMCID: PMC2999673 DOI: 10.4251/wjgo.v2.i10.369] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 09/20/2010] [Accepted: 09/27/2010] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer is the most common malignancy of the gastrointestinal tract in East Asian populations and the second most frequent cause of cancer-related mortality in the world. While previous studies have investigated the genetic factors involved in gastric carcinogenesis, there still exist relatively few studies that have investigated the genetic traits associated with the risk of gastric precancerous conditions. In this paper we will review the biology and genetic polymorphisms involved in the genesis of gastric precancerous conditions reported to date and discuss the future prospects of this field of study. The associations of gastric precancerous conditions with polymorphisms in the cytotoxin-associated gene A-related genes (e.g. PTPN11 G/A at intron 3, rs2301756), those in the genes involved in host immunity against Helicobacter pylori (H. pylori) infection (e.g. TLR4 +3725G/C, rs11536889) or polymorphisms of the genes essential for the development/ differentiation of the gastric epithelial cells (e.g. RUNX3 T/A polymorphism at intron 3, rs760805) have been reported to date. Genetic epidemiological studies of the associations between H. pylori-induced gastric precancerous conditions and other gene polymorphisms in these pathways as well as polymorphisms of the genes involved in other pathways like oxidative DNA damage repair pathways would provide useful evidence for the individualized prevention of these H. pylori-induced gastric precancerous conditions.
Collapse
Affiliation(s)
- Asahi Hishida
- Asahi Hishida, Yasuyuki Goto, Nobuyuki Hamajima, Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | | | | |
Collapse
|
31
|
Kfir-Erenfeld S, Sionov RV, Spokoini R, Cohen O, Yefenof E. Protein kinase networks regulating glucocorticoid-induced apoptosis of hematopoietic cancer cells: fundamental aspects and practical considerations. Leuk Lymphoma 2010; 51:1968-2005. [PMID: 20849387 DOI: 10.3109/10428194.2010.506570] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are integral components in the treatment protocols of acute lymphoblastic leukemia, multiple myeloma, and non-Hodgkin lymphoma owing to their ability to induce apoptosis of these malignant cells. Resistance to GC therapy is associated with poor prognosis. Although they have been used in clinics for decades, the signal transduction pathways involved in GC-induced apoptosis have only partly been resolved. Accumulating evidence shows that this cell death process is mediated by a communication between nuclear GR affecting gene transcription of pro-apoptotic genes such as Bim, mitochondrial GR affecting the physiology of the mitochondria, and the protein kinase glycogen synthase kinase-3 (GSK3), which interacts with Bim following exposure to GCs. Prevention of Bim up-regulation, mitochondrial GR translocation, and/or GSK3 activation are common causes leading to GC therapy failure. Various protein kinases positively regulating the pro-survival Src-PI3K-Akt-mTOR and Raf-Ras-MEK-ERK signal cascades have been shown to be activated in malignant leukemic cells and antagonize GC-induced apoptosis by inhibiting GSK3 activation and Bim expression. Targeting these protein kinases has proven effective in sensitizing GR-positive malignant lymphoid cells to GC-induced apoptosis. Thus, intervening with the pro-survival kinase network in GC-resistant cells should be a good means of improving GC therapy of hematopoietic malignancies.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- The Lautenberg Center of Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
32
|
El-Deeb IM, Yoo KH, Lee SH. ROS receptor tyrosine kinase: a new potential target for anticancer drugs. Med Res Rev 2010; 31:794-818. [PMID: 20687158 DOI: 10.1002/med.20206] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
ROS kinase is one of the last two remaining orphan receptor tyrosine kinases with an as yet unidentified ligand. The normal functions of human ROS kinase in different body tissues have not been fully identified so far. However, the ectopic expression, as well as the production of variable mutant forms of ROS kinase has been reported in a number of cancers, such as glioblastoma multiforme, and non-small cell lung cancer, suggesting a role for ROS kinase in deriving such tumors. It is thought also that c-ROS gene may have a role in some cardiovascular diseases, and the fact that homozygous male mice targeted against c-ROS gene are healthy but infertile, has inspired researchers to think about ROS inhibition as a method for development of new male contraceptives. The recent discovery of new selective and potent inhibitors for ROS kinase, along with the development of new specific diagnostic methods for the detection of ROS fusion proteins, raises the importance of using these selective inhibitors for targeting ROS mutations as a new method for treatment of cancers harboring such genes. This review focuses on the ectopic expression of ROS and its fusion proteins in different cancer types and highlights the importance of targeting these proteins for treatment of substantial cancers. It describes also the recent advances in the field of ROS kinase inhibition, and the potential clinical applications of ROS kinase inhibitors.
Collapse
Affiliation(s)
- Ibrahim Mustafa El-Deeb
- Life/Health Division, Korea Institute of Science and Technology, Cheongryang, Seoul, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Yuseong-gu, Daejeon, Republic of Korea
| | | | | |
Collapse
|
33
|
Src tyrosine kinase preactivation is associated with platelet hypersensitivity in essential thrombocythemia and polycythemia vera. Blood 2010; 115:667-76. [DOI: 10.1182/blood-2008-12-196535] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Polycythemia vera (PV) and essential thrombocythemia (ET) are chronic myeloproliferative disorders characterized by an increased incidence of thrombo-hemorrhagic complications. The acquired somatic Janus kinase 2 (JAK2) V617F mutation is present in the majority of PV and ET patients. Because aberrant protein Tyr-phosphorylation has been associated with hematopoietic malignancies, the activity of the tyrosine kinases Src and JAK2 was analyzed in resting and thrombin-stimulated platelets from 13 PV and 42 ET patients. JAK2 was found inactive in healthy and pathological resting cells regardless of the V617F mutation. In addition, Src was inactive in all resting platelets, but in the pathological specimens it was present in a preactivated conformation as a consequence of anomalous dephosphorylation of its inhibitory phospho-Tyr527 residue, likely mediated by Src homology-2 domain-containing protein Tyr-phosphatase-2 (SHP-2), whose constitutive activity correlated with its recruitment to Src. Low thrombin concentration triggered a more rapid Src-signaling activation, higher [Ca2+]c increase, and aggregation in pathological platelets compared with controls. Thrombin-induced Src activation preceded JAK2 activation, which occurred simultaneously in normal and pathological platelets. Our results indicate that a constitutive Src kinase preactivation is implicated in platelet hypersensitivity and likely involved, at least partially, in the functional abnormalities of PV and ET platelets.
Collapse
|
34
|
Tartaglia M, Zampino G, Gelb BD. Noonan syndrome: clinical aspects and molecular pathogenesis. Mol Syndromol 2010; 1:2-26. [PMID: 20648242 DOI: 10.1159/000276766] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 10/30/2009] [Indexed: 01/20/2023] Open
Abstract
Noonan syndrome (NS) is a relatively common, clinically variable and genetically heterogeneous developmental disorder characterized by postnatally reduced growth, distinctive facial dysmorphism, cardiac defects and variable cognitive deficits. Other associated features include ectodermal and skeletal defects, cryptorchidism, lymphatic dysplasias, bleeding tendency, and, rarely, predisposition to hematologic malignancies during childhood. NS is caused by mutations in the PTPN11, SOS1, KRAS, RAF1, BRAF and MEK1 (MAP2K1) genes, accounting for approximately 70% of affected individuals. SHP2 (encoded by PTPN11), SOS1, BRAF, RAF1 and MEK1 positively contribute to RAS-MAPK signaling, and possess complex autoinhibitory mechanisms that are impaired by mutations. Similarly, reduced GTPase activity or increased guanine nucleotide release underlie the aberrant signal flow through the MAPK cascade promoted by most KRAS mutations. More recently, a single missense mutation in SHOC2, which encodes a cytoplasmic scaffold positively controlling RAF1 activation, has been discovered to cause a closely related phenotype previously termed Noonan-like syndrome with loose anagen hair. This mutation promotes aberrantly acquired N-myristoylation of the protein, resulting in its constitutive targeting to the plasma membrane and dysregulated function. PTPN11, BRAF and RAF1 mutations also account for approximately 95% of LEOPARD syndrome, a condition which resembles NS phenotypically but is characterized by multiple lentigines dispersed throughout the body, café-au-lait spots, and a higher prevalence of electrocardiographic conduction abnormalities, obstructive cardiomyopathy and sensorineural hearing deficits. These recent discoveries demonstrate that the substantial phenotypic variation characterizing NS and related conditions can be ascribed, in part, to the gene mutated and even the specific molecular lesion involved.
Collapse
Affiliation(s)
- M Tartaglia
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
35
|
Thiel C, Wilken M, Zenker M, Sticht H, Fahsold R, Gusek-Schneider GC, Rauch A. Independent NF1 and PTPN11 mutations in a family with neurofibromatosis-Noonan syndrome. Am J Med Genet A 2009; 149A:1263-7. [PMID: 19449407 DOI: 10.1002/ajmg.a.32837] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurofibromatosis-Noonan syndrome (NFNS), an entity which combines both features of Noonan syndrome (NS) and neurofibromatosis type 1 (NF1), was etiologically unresolved until recent reports demonstrated NF1 mutations in the majority of patients with NFNS. The phenotypic overlap was explained by the involvement of the Ras pathway in both disorders, and, accordingly, clustering of the NF1 mutations in the GTPase-activating protein (GAP) domain of neurofibromin was observed in individuals with NFNS. We report on an 18-month-old girl with typical findings suggestive of NS in combination with multiple café-au-lait spots and bilateral optic gliomas suggestive of NF1. The patient was found to carry a de novo PTPN11 mutation p.T2I as well as the maternally inherited NF1 mutation c.4661+1G>C. Her otherwise healthy mother and brother, who also had the NF1 mutation, showed few café-au-lait spots as the only sign of neurofibromatosis. Since our patient's unique NF1 mutation results in skipping of exon 27a and thus involves the same region, Gap-related domain, that had been shown to be associated with NFNS, her phenotype could have been misleadingly attributed to the NF1 mutation only. Contrarily, absence of both cutaneous neurofibromas and NS features in her relatives with the same NF1 mutation, suggests that the index patient's typical NFNS phenotype is caused by an additive effect of mutations in both NF1 and PTPN11. In contrast to previous findings, we speculate that absence of cutaneous neurofibromas is not solely associated with the recurrent 3-bp in-frame deletion in exon 17.
Collapse
Affiliation(s)
- Christian Thiel
- Insitute of Human Genetics, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
36
|
Hishida A, Matsuo K, Goto Y, Naito M, Wakai K, Tajima K, Hamajima N. Associations of a PTPN11 G/A polymorphism at intron 3 with Helicobactor pylori seropositivity, gastric atrophy and gastric cancer in Japanese. BMC Gastroenterol 2009; 9:51. [PMID: 19589142 PMCID: PMC2715419 DOI: 10.1186/1471-230x-9-51] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 07/09/2009] [Indexed: 12/12/2022] Open
Abstract
Background Previous studies have revealed the significance of Helicobacter pylori (H. pylori) infection as a risk factor of gastric cancer. Cytotoxin-associated gene A (cagA) positivity has been demonstrated to determine the clinical outcome of H. pylori infection in the presence of SHP-2 (src homology 2 domain-containing protein tyrosine phosphatase-2). This study aimed to examine the formerly reported association of G/A PTPN11 (protein-tyrosine phosphatase, nonreceptor-type 11) polymorphism (rs2301756) with gastric atrophy, as well as the association with gastric cancer in a Japanese population using a large sample size. Methods Study subjects were 583 histologically diagnosed patients with gastric cancer (429 males and 154 females) and age- and sex-frequency-matched 1,636 non-cancer outpatients (1,203 males and 433 females), who visited Aichi Cancer Center Hospital between 2001–2005. Serum anti-H. pylori IgG antibody and pepsinogens were measured to evaluate H. pylori infection and gastric atrophy, respectively. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by a logistic model. Results Among H. pylori seropositive non-cancer outpatients, the age- and sex-adjusted OR of gastric atrophy was 0.82 (95% CI 0.62–1.10, P = 0.194) for G/A, 0.84 (95% CI 0.39–1.81, P = 0.650) for A/A, and 0.83 (95% CI 0.62–1.09, P = 0.182) for G/A+A/A, relative to G/G genotype, and that of severe gastric atrophy was 0.70 (95% CI 0.47–1.04, P = 0.079), 0.56 (95% CI 0.17–1.91, P = 0.356), and 0.68 (95% CI 0.46–1.01, P = 0.057), respectively. Among H. pylori infected subjects (H. pylori seropositive subjects and seronegative subjects with gastric atrophy), the adjusted OR of severe gastric atrophy was further reduced; 0.62 (95% CI 0.42–0.90, P = 0.012) for G/A+A/A. The distribution of the genotype in patients with gastric cancer was not significantly different from that for H. pylori infected subjects without gastric atrophy. Conclusion Our study results revealed that those with the A/A genotype of PTPN11 rs2301756 polymorphism are at lower risk of severe gastric atrophy, but are not associated with a decreased risk of gastric cancer, which partially supported our previous finding that the polymorphism in the PTPN11 gene encoding SHP-2 was associated with the gastric atrophy risk in H. pylori infected Japanese. The biological roles of this PTPN11 polymorphism require further investigation.
Collapse
Affiliation(s)
- Asahi Hishida
- Department of Preventive Medicine/Biostatistics and Medical Decision Making, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
37
|
Bergom C, Gao C, Newman PJ. Mechanisms of PECAM-1-mediated cytoprotection and implications for cancer cell survival. Leuk Lymphoma 2009; 46:1409-21. [PMID: 16194886 DOI: 10.1080/10428190500126091] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Defects in apoptotic pathways can promote cancer development and cause cancers to become resistant to chemotherapy. The cell adhesion and signaling molecule PECAM-1 has been shown to potently suppress apoptosis in a variety of cellular systems. PECAM-1 expression has been reported on a variety of human malignancies-especially hematopoietic and vascular cell cancers-but the significance of this expression has not been fully explored. The ability of PECAM-1 to inhibit apoptosis makes it an attractive candidate as a molecule that may promote cancer development and/or confer resistance to chemotherapeutic treatment. The exact mechanisms by which PECAM-1 mediates its cytoprotection have not been fully defined, but its anti-apoptotic effects have been shown to require both homophilic binding and intracellular signaling via its immunoreceptor tyrosine-based inhibitory motif (ITIM) domains. In this review, we will discuss the data regarding PECAM-1's anti-apoptotic effects and ways in which this cytoprotection may be clinically relevant to the development and/or treatment of hematologic malignancies that express this vascular cell-specific surface molecule.
Collapse
Affiliation(s)
- Carmen Bergom
- Blood Research Institute, The Blood Center of Southeastern Wisconsin, Milwaukee, Milwaukee, WI, USA.
| | | | | |
Collapse
|
38
|
Zou GM, Maitra A. Small-molecule inhibitor of the AP endonuclease 1/REF-1 E3330 inhibits pancreatic cancer cell growth and migration. Mol Cancer Ther 2008; 7:2012-21. [PMID: 18645011 DOI: 10.1158/1535-7163.mct-08-0113] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
AP endonuclease 1 (APE1; also known as REF-1) contains a DNA repair domain and a redox regulation domain. APE1 is overexpressed in several human cancers, and disruption of APE1 function has detrimental effects on cancer cell viability. However, the selective contribution of the redox and the DNA repair domains to maintenance of cellular homeostasis in cancer has not been elucidated. In the present study, we used E3330, a small-molecule inhibitor of APE1 redox domain function, to interrogate the functional relevance of sustained redox function in pancreatic cancer. We show that E3330 significantly reduces the growth of human pancreatic cancer cells in vitro. This phenomenon was further confirmed by a small interfering RNA experiment to knockdown APE1 expression in pancreatic cancer cells. Further, the growth-inhibitory effects of E3330 are accentuated by hypoxia, and this is accompanied by striking inhibition in the DNA-binding ability of hypoxia-inducible factor-1alpha, a hypoxia-induced transcription factor. E3330 exposure promotes endogenous reactive oxygen species formation in pancreatic cancer cells, and the resulting oxidative stress is associated with higher levels of oxidized, and hence inactive, SHP-2, an essential protein tyrosine phosphatase that promotes cancer cell proliferation in its active state. Finally, E3330 treatment inhibits pancreatic cancer cell migration as assessed by in vitro chemokine assays. E3330 shows anticancer properties at multiple functional levels in pancreatic cancer, such as inhibition of cancer cell growth and migration. Inhibition of the APE1 redox function through pharmacologic means has the potential to become a promising therapeutic strategy in this disease.
Collapse
Affiliation(s)
- Gang-Ming Zou
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB-2, M341, Baltimore, MD 21231, USA.
| | | |
Collapse
|
39
|
Acquaviva J, Wong R, Charest A. The multifaceted roles of the receptor tyrosine kinase ROS in development and cancer. Biochim Biophys Acta Rev Cancer 2008; 1795:37-52. [PMID: 18778756 DOI: 10.1016/j.bbcan.2008.07.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Accepted: 07/21/2008] [Indexed: 12/26/2022]
Abstract
The proto-oncogene receptor tyrosine kinase ROS was originally discovered through the identification of oncogenic variants isolated from tumors. These discoveries spearheaded a body of work aimed at elucidating the function of this evolutionarily conserved receptor in development and cancer. Through genetic and biochemical approaches, progress in the characterization of ROS points to distinctive roles in the program of epithelial cell differentiation during the development of a variety of organs. Although substantial, these advances remain hampered by the absence of an identified ligand, making ROS one of the last two remaining orphan receptor tyrosine kinases. Recent studies on the oncogenic activation of ROS as a result of different chromosomal rearrangements found in brain and lung cancers have shed light on the molecular mechanisms underlying ROS transforming activities. ROS and its oncogenic variants therefore constitute clinically relevant targets for cancer therapeutic intervention. This review highlights the various roles that this receptor plays in multiple system networks in normalcy and disease and points to future directions towards the elucidation of ROS function in the context of ligand identification, signaling pathways and clinical applications.
Collapse
Affiliation(s)
- Jaime Acquaviva
- Molecular Oncology Research Institute, Tufts University School of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | | | | |
Collapse
|
40
|
Abstract
Diverse cellular processes are regulated by tyrosyl phosphorylation, which is controlled by protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs). De-regulated tyrosyl phosphorylation, evoked by gain-of-function mutations and/or over-expression of PTKs, contributes to the pathogenesis of many cancers and other human diseases. PTPs, because they oppose the action of PTKs, had been considered to be prime suspects for potential tumor suppressor genes. Surprisingly, few, if any, tumor suppressor PTPs have been identified. However, the Src homology-2 domain-containing phosphatase Shp2 (encoded by PTPN11) is a bona fide proto-oncogene. Germline mutations in PTPN11 cause Noonan and LEOPARD syndromes, whereas somatic PTPN11 mutations occur in several types of hematologic malignancies, most notably juvenile myelomonocytic leukemia and, more rarely, in solid tumors. Shp2 also is an essential component in several other oncogene signaling pathways. Elucidation of the events underlying Shp2-evoked transformation may provide new insights into oncogenic mechanisms and novel targets for anti-cancer therapy.
Collapse
|
41
|
Martinelli S, Torreri P, Tinti M, Stella L, Bocchinfuso G, Flex E, Grottesi A, Ceccarini M, Palleschi A, Cesareni G, Castagnoli L, Petrucci TC, Gelb BD, Tartaglia M. Diverse driving forces underlie the invariant occurrence of the T42A, E139D, I282V and T468M SHP2 amino acid substitutions causing Noonan and LEOPARD syndromes. Hum Mol Genet 2008; 17:2018-29. [PMID: 18372317 PMCID: PMC2900904 DOI: 10.1093/hmg/ddn099] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 03/25/2008] [Indexed: 01/22/2023] Open
Abstract
Missense PTPN11 mutations cause Noonan and LEOPARD syndromes (NS and LS), two developmental disorders with pleiomorphic phenotypes. PTPN11 encodes SHP2, an SH2 domain-containing protein tyrosine phosphatase functioning as a signal transducer. Generally, different substitutions of a particular amino acid residue are observed in these diseases, indicating that the crucial factor is the residue being replaced. For a few codons, only one substitution is observed, suggesting the possibility of specific roles for the residue introduced. We analyzed the biochemical behavior and ligand-binding properties of all possible substitutions arising from single-base changes affecting codons 42, 139, 279, 282 and 468 to investigate the mechanisms underlying the invariant occurrence of the T42A, E139D and I282V substitutions in NS and the Y279C and T468M changes in LS. Our data demonstrate that the isoleucine-to-valine change at codon 282 is the only substitution at that position perturbing the stability of SHP2's closed conformation without impairing catalysis, while the threonine-to-alanine change at codon 42, but not other substitutions of that residue, promotes increased phosphopeptide-binding affinity. The recognition specificity of the C-SH2 domain bearing the E139D substitution differed substantially from its wild-type counterpart acquiring binding properties similar to those observed for the N-SH2 domain, revealing a novel mechanism of SHP2's functional dysregulation. Finally, while functional selection does not seem to occur for the substitutions at codons 279 and 468, we point to deamination of the methylated cytosine at nucleotide 1403 as the driving factor leading to the high prevalence of the T468M change in LS.
Collapse
Affiliation(s)
- Simone Martinelli
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Paola Torreri
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Michele Tinti
- Dipartimento di Biologia, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c., 00133 Rome, Italy
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c, 00133 Rome, Italy
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c, 00133 Rome, Italy
| | - Elisabetta Flex
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandro Grottesi
- Consortium for the Application of Super-Computing for Universities and Research (CASPUR), Via dei Tizii 6, 00185 Rome, Italy
| | - Marina Ceccarini
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonio Palleschi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c, 00133 Rome, Italy
| | - Gianni Cesareni
- Dipartimento di Biologia, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c., 00133 Rome, Italy
| | - Luisa Castagnoli
- Dipartimento di Biologia, Università di Roma ‘Tor Vergata’, Via della Ricerca Scientifica s.n.c., 00133 Rome, Italy
| | - Tamara C. Petrucci
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | - Marco Tartaglia
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
42
|
Sarkozy A, Digilio MC, Dallapiccola B. Leopard syndrome. Orphanet J Rare Dis 2008; 3:13. [PMID: 18505544 PMCID: PMC2467408 DOI: 10.1186/1750-1172-3-13] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2008] [Accepted: 05/27/2008] [Indexed: 11/24/2022] Open
Abstract
LEOPARD syndrome (LS, OMIM 151100) is a rare multiple congenital anomalies condition, mainly characterized by skin, facial and cardiac anomalies. LEOPARD is an acronym for the major features of this disorder, including multiple Lentigines, ECG conduction abnormalities, Ocular hypertelorism, Pulmonic stenosis, Abnormal genitalia, Retardation of growth, and sensorineural Deafness. About 200 patients have been reported worldwide but the real incidence of LS has not been assessed. Facial dysmorphism includes ocular hypertelorism, palpebral ptosis and low-set ears. Stature is usually below the 25th centile. Cardiac defects, in particular hypertrophic cardiomyopathy mostly involving the left ventricle, and ECG anomalies are common. The lentigines may be congenital, although more frequently manifest by the age of 4–5 years and increase throughout puberty. Additional common features are café-au-lait spots (CLS), chest anomalies, cryptorchidism, delayed puberty, hypotonia, mild developmental delay, sensorineural deafness and learning difficulties. In about 85% of the cases, a heterozygous missense mutation is detected in exons 7, 12 or 13 of the PTPN11 gene. Recently, missense mutations in the RAF1 gene have been found in two out of six PTPN11-negative LS patients. Mutation analysis can be carried out on blood, chorionic villi and amniotic fluid samples. LS is largely overlapping Noonan syndrome and, during childhood, Neurofibromatosis type 1-Noonan syndrome. Diagnostic clues of LS are multiple lentigines and CLS, hypertrophic cardiomyopathy and deafness. Mutation-based differential diagnosis in patients with borderline clinical manifestations is warranted. LS is an autosomal dominant condition, with full penetrance and variable expressivity. If one parent is affected, a 50% recurrence risk is appropriate. LS should be suspected in foetuses with severe cardiac hypertrophy and prenatal DNA test may be performed. Clinical management should address growth and motor development and congenital anomalies, in particular cardiac defects that should be monitored annually. Hypertrophic cardiomyopathy needs careful risk assessment and prophylaxis against sudden death in patients at risk. Hearing should be evaluated annually until adulthood. With the only exception of ventricular hypertrophy, adults with LS do not require special medical care and long-term prognosis is favourable.
Collapse
Affiliation(s)
- Anna Sarkozy
- IRCCS-CSS, San Giovanni Rotondo and CSS-Mendel Institute, Viale Regina Elena 261, 00198, Rome, Italy.
| | | | | |
Collapse
|
43
|
Roubelakis MG, Martin-Rendon E, Tsaknakis G, Stavropoulos A, Watt SM. The murine ortholog of the SHP-2 binding molecule, PZR accelerates cell migration on fibronectin and is expressed in early embryo formation. J Cell Biochem 2008; 102:955-69. [PMID: 17440960 DOI: 10.1002/jcb.21334] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The human P zero-related protein (hPZR) has a unique function in regulating cell migration. This activity is dependent on both its cytoplasmic immunoreceptor tyrosine inhibitory motif (ITIM) and its interaction with the tyrosine protein phosphatase, src homology phosphatase-2 (SHP-2). Here, using in silico and cDNA cloning approaches, we identify the murine ITIM-containing hPZR ortholog, mPZR, together with its ITIM-less isoform, mPZRb. We demonstrate that, like hPZR, these type 1 integral murine transmembrane isoforms are derived by differential splicing from a single gene transcription unit on mouse chromosome 1, and differ only in the sequence of their cytoplasmic domains. Importantly, mPZR mimicks hPZR functionally by accelerating SHP-2-mediated cell migration on fibronectin. Interestingly, we further demonstrate that although neither mPZR nor mPZRb is expressed in murine pluripotent embryonic stem cells, they first appear at approximately day 3 of blastocyst formation in vivo and of embryoid body formation in vitro. These studies thus provide the basis for defining the function of the mPZR isoforms in vivo, particularly with respect to their roles in regulating SHP-2-dependent cell migration during development.
Collapse
Affiliation(s)
- Maria G Roubelakis
- Stem Cell Research Laboratory, National Blood Service, NHS Blood and Transplant Authority, Oxford OX3 5BG, UK.
| | | | | | | | | |
Collapse
|
44
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of neoplastic disorders with great variability in clinical course and response to therapy, as well as in the genetic and molecular basis of the pathology. Major advances in the understanding of leukemogenesis have been made by the characterization and the study of acquired cytogenetic abnormalities, particularly reciprocal translocations observed in AML. Besides these major cytogenetic abnormalities, gene mutations also constitute key events in AML pathogenesis. In this review, we describe the contribution of known gene mutations to the understanding of AML pathogenesis and their clinical significance. To gain more insight in this understanding, we clustered these alterations in three groups: (1) mutations affecting genes that contribute to cell proliferation (FLT3, c-KIT, RAS, protein tyrosine standard phosphatase nonreceptor 11); (2) mutations affecting genes involved in myeloid differentiation (AML1 and CEBPA) and (3) mutations affecting genes implicated in cell cycle regulation or apoptosis (P53, NPM1). This nonexhaustive review aims to show how gene mutations interact with each other, how they contribute to refine prognosis and how they can be useful for risk-adapted therapeutic management of AML patients.
Collapse
|
45
|
Stuible M, Doody KM, Tremblay ML. PTP1B and TC-PTP: regulators of transformation and tumorigenesis. Cancer Metastasis Rev 2008; 27:215-30. [DOI: 10.1007/s10555-008-9115-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
46
|
Abstract
Abnormalities of cytokine and growth factor signaling pathways are characteristic of all forms of leukemia: lymphoid and myeloid, acute and chronic. In normal hematopoietic cells, cytokines provide the stimulus for proliferation, survival, self-renewal, differentiation and functional activation. In leukemic cells, these pathways are usurped to subserve critical parts of the malignant program. In this review, our current knowledge of leukemic cell cytokine signaling will be summarized, and some speculations on the significance and implications of these insights will be advanced. A better understanding of aberrant cytokine signaling in leukemia should provide additional targets for the rational therapy of these diseases.
Collapse
Affiliation(s)
- R A Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
47
|
Abstract
Ras genes are the most common targets for somatic gain-of-function mutations in human cancer. Recently, germline mutations that affect components of the Ras-Raf-mitogen-activated and extracellular-signal regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway were shown to cause several developmental disorders, including Noonan, Costello and cardio-facio-cutaneous syndromes. Many of these mutant alleles encode proteins with aberrant biochemical and functional properties. Here we will discuss the implications of germline mutations in the Ras-Raf-MEK-ERK pathway for understanding normal developmental processes and cancer pathogenesis.
Collapse
Affiliation(s)
- Suzanne Schubbert
- Department of Pediatrics, University of California, 513 Parnassus Avenue, Room HSE-302, San Francisco, California 94143, USA
| | | | | |
Collapse
|
48
|
Abstract
Tyrosyl phosphorylation plays a critical role in multiple signaling pathways regulating innate and acquired immunity. Although tyrosyl phosphorylation is a reversible process, we know much more about the functions of protein-tyrosine kinases (PTKs) than about protein-tyrosine phosphatases (PTPs). Genome sequencing efforts have revealed a large and diverse superfamily of PTPs, which can be subdivided into receptor-like (RPTPs) and nonreceptor (NRPTPs). The role of the RPTP CD45 in immune cell signaling is well known, but those of most other PTPs remain poorly understood. Here, we review the mechanism of action, regulation, and physiological functions of NRPTPs in immune cell signaling. Such an analysis indicates that PTPs are as important as PTKs in regulating the immune system.
Collapse
Affiliation(s)
- Lily I Pao
- Cancer Biology Program, Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | |
Collapse
|
49
|
Kawai S, Goto Y, Ito LS, Oba-Shinjo SM, Uno M, Shinjo SK, Marie SKN, Ishida Y, Nishio K, Naito M, Hamajima N. Significant association between PTPN11 polymorphism and gastric atrophy among Japanese Brazilians. Gastric Cancer 2007; 9:277-83. [PMID: 17235629 DOI: 10.1007/s10120-006-0391-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 06/13/2006] [Indexed: 02/07/2023]
Abstract
BACKGROUND Helicobacter pylori, especially the cytotoxin-associated antigen A (cagA)-positive strains, plays a crucial role in the development of gastric atrophy and gastric cancer. CagA delivered into gastric epithelial cells combines with src homology 2 domain-containing protein tyrosine phosphatase-2 (SHP-2), possibly leading to atrophy/cancer. Our previous study found that a single-nucleotide polymorphism (SNP; IMS-JST057927) of the PTPN11 gene encoding SHP-2, was associated with gastric atrophy among H. pylori-seropositive subjects. This study aimed to examine the reproducibility of the association among Japanese residing in a different circumstance. METHODS The subjects were 918 healthy adult Japanese Brazilians from four different areas in Brazil. Blood was sampled from March to May 2001. The target SNP in intron 3 of PTPN11 was genotyped by polymerase chain reaction with confronting two-pair primers (PCR-CTPP). Gastric atrophy was evaluated with serum pepsinogens (PGs); PG I, less than 70 ng/dl and PG I/II ratio, less than 3. RESULTS The genotype frequency of PTPN11 was in Hardy-Weinberg equilibrium: 65.5% for G/G, 30.4% for G/A, and 4.1% for A/A. The PTPN11 polymorphism had no significant association with H. pylori seropositivity. Among the H. pylori-seropositive subjects, the odds ratios (ORs) of gastric atrophy were 0.93 (95% confidence interval [CI], 0.59-1.47) for the G/A genotype and 0.31 (95% CI, 0.10-0.95) for the A/A genotype, compared with the G/G genotype. CONCLUSIONS The present study reproduced the significant association between the A/A genotype and reduced risk of gastric atrophy among Japanese outside Japan. According to the Japan Single Nucleotide Polymorphisms (JSNP) database (db)SNP data, the G allele is very frequent among Japanese and rare in Caucasians. This fact may partly explain the distribution of gastric atrophy/cancer in the world.
Collapse
Affiliation(s)
- Sayo Kawai
- Department of Preventive Medicine/Biostatistics and Medical Decision Making, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Touw IP, Bontenbal M. Granulocyte Colony-Stimulating Factor: Key (F)actor or Innocent Bystander in the Development of Secondary Myeloid Malignancy? ACTA ACUST UNITED AC 2007; 99:183-6. [PMID: 17284707 DOI: 10.1093/jnci/djk057] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|