1
|
Sudhakar N, Yan L, Qiryaqos F, Engstrom LD, Laguer J, Calinisan A, Hebbert A, Waters L, Moya K, Bowcut V, Vegar L, Ketcham JM, Ivetac A, Smith CR, Lawson JD, Rahbaek L, Clarine J, Nguyen N, Saechao B, Parker C, Elliott AJ, Vanderpool D, He L, Hover LD, Fernandez-Banet J, Coma S, Pachter JA, Hallin J, Marx MA, Briere DM, Christensen JG, Olson P, Haling J, Khare S. The SOS1 Inhibitor MRTX0902 Blocks KRAS Activation and Demonstrates Antitumor Activity in Cancers Dependent on KRAS Nucleotide Loading. Mol Cancer Ther 2024; 23:1418-1430. [PMID: 38904222 PMCID: PMC11443210 DOI: 10.1158/1535-7163.mct-23-0870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/14/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
KRAS is the most frequently mutated oncogene in human cancer and facilitates uncontrolled growth through hyperactivation of the receptor tyrosine kinase (RTK)/mitogen-activated protein kinase (MAPK) pathway. The Son of Sevenless homolog 1 (SOS1) protein functions as a guanine nucleotide exchange factor (GEF) for the RAS subfamily of small GTPases and represents a druggable target in the pathway. Using a structure-based drug discovery approach, MRTX0902 was identified as a selective and potent SOS1 inhibitor that disrupts the KRAS:SOS1 protein-protein interaction to prevent SOS1-mediated nucleotide exchange on KRAS and translates into an anti-proliferative effect in cancer cell lines with genetic alterations of the KRAS-MAPK pathway. MRTX0902 augmented the antitumor activity of the KRAS G12C inhibitor adagrasib when dosed in combination in eight out of 12 KRAS G12C-mutant human non-small cell lung cancer and colorectal cancer xenograft models. Pharmacogenomic profiling in preclinical models identified cell cycle genes and the SOS2 homolog as genetic co-dependencies and implicated tumor suppressor genes (NF1 and PTEN) in resistance following combination treatment. Lastly, combined vertical inhibition of RTK/MAPK pathway signaling by MRTX0902 with inhibitors of EGFR or RAF/MEK led to greater downregulation of pathway signaling and improved antitumor responses in KRAS-MAPK pathway-mutant models. These studies demonstrate the potential clinical application of dual inhibition of SOS1 and KRAS G12C and additional SOS1 combination strategies that will aide in the understanding of SOS1 and RTK/MAPK biology in targeted cancer therapy.
Collapse
Affiliation(s)
| | - Larry Yan
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | - Jade Laguer
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | - Laura Waters
- Mirati Therapeutics, Inc., San Diego, California
| | - Krystal Moya
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Laura Vegar
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | | | | | - Lisa Rahbaek
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | | | - Cody Parker
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | - Leo He
- Monoceros Biosciences LLC, San Diego, California
| | | | | | | | | | - Jill Hallin
- Mirati Therapeutics, Inc., San Diego, California
| | | | | | | | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Jacob Haling
- Mirati Therapeutics, Inc., San Diego, California
| | - Shilpi Khare
- Mirati Therapeutics, Inc., San Diego, California
| |
Collapse
|
2
|
Faienza MF, Meliota G, Mentino D, Ficarella R, Gentile M, Vairo U, D’amato G. Cardiac Phenotype and Gene Mutations in RASopathies. Genes (Basel) 2024; 15:1015. [PMID: 39202376 PMCID: PMC11353738 DOI: 10.3390/genes15081015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Cardiac involvement is a major feature of RASopathies, a group of phenotypically overlapping syndromes caused by germline mutations in genes encoding components of the RAS/MAPK (mitogen-activated protein kinase) signaling pathway. In particular, Noonan syndrome (NS) is associated with a wide spectrum of cardiac pathologies ranging from congenital heart disease (CHD), present in approximately 80% of patients, to hypertrophic cardiomyopathy (HCM), observed in approximately 20% of patients. Genotype-cardiac phenotype correlations are frequently described, and they are useful indicators in predicting the prognosis concerning cardiac disease over the lifetime. The aim of this review is to clarify the molecular mechanisms underlying the development of cardiac diseases associated particularly with NS, and to discuss the main morphological and clinical characteristics of the two most frequent cardiac disorders, namely pulmonary valve stenosis (PVS) and HCM. We will also report the genotype-phenotype correlation and its implications for prognosis and treatment. Knowing the molecular mechanisms responsible for the genotype-phenotype correlation is key to developing possible targeted therapies. We will briefly address the first experiences of targeted HCM treatment using RAS/MAPK pathway inhibitors.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Giovanni Meliota
- Department of Pediatric Cardiology, Giovanni XXIII Pediatric Hospital, 70126 Bari, Italy; (G.M.); (U.V.)
| | - Donatella Mentino
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Romina Ficarella
- U.O.C. Laboratorio di Genetica Medica, PO Di Venere-ASL Bari, 70012 Bari, Italy; (R.F.); (M.G.)
| | - Mattia Gentile
- U.O.C. Laboratorio di Genetica Medica, PO Di Venere-ASL Bari, 70012 Bari, Italy; (R.F.); (M.G.)
| | - Ugo Vairo
- Department of Pediatric Cardiology, Giovanni XXIII Pediatric Hospital, 70126 Bari, Italy; (G.M.); (U.V.)
| | - Gabriele D’amato
- Neonatal Intensive Care Unit, Di Venere Hospital, 70012 Bari, Italy;
| |
Collapse
|
3
|
Stellacci E, Carter JN, Pannone L, Stevenson D, Moslehi D, Venanzi S, Bernstein JA, Tartaglia M, Martinelli S. Immunological and hematological findings as major features in a patient with a new germline pathogenic CBL variant. Am J Med Genet A 2024; 194:e63627. [PMID: 38613168 PMCID: PMC11223960 DOI: 10.1002/ajmg.a.63627] [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: 02/01/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024]
Abstract
Casitas B-lineage lymphoma (CBL) encodes an adaptor protein with E3-ligase activity negatively controlling intracellular signaling downstream of receptor tyrosine kinases. Somatic CBL mutations play a driver role in a variety of cancers, particularly myeloid malignancies, whereas germline defects in the same gene underlie a RASopathy having clinical overlap with Noonan syndrome (NS) and predisposing to juvenile myelomonocytic leukemia and vasculitis. Other features of the disorder include cardiac defects, postnatal growth delay, cryptorchidism, facial dysmorphisms, and predisposition to develop autoimmune disorders. Here we report a novel CBL variant (c.1202G>T; p.Cys401Phe) occurring de novo in a subject with café-au-lait macules, feeding difficulties, mild dysmorphic features, psychomotor delay, autism spectrum disorder, thrombocytopenia, hepatosplenomegaly, and recurrent hypertransaminasemia. The identified variant affects an evolutionarily conserved residue located in the RING finger domain, a known mutational hot spot of both germline and somatic mutations. Functional studies documented enhanced EGF-induced ERK phosphorylation in transiently transfected COS1 cells. The present findings further support the association of pathogenic CBL variants with immunological and hematological manifestations in the context of a presentation with only minor findings reminiscent of NS or a clinically related RASopathy.
Collapse
Affiliation(s)
- Emilia Stellacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- These authors equally contributed to this work
| | - Jennefer N. Carter
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- These authors equally contributed to this work
| | - Luca Pannone
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - David Stevenson
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dorsa Moslehi
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
| | - Serenella Venanzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Jonathan A. Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
- These authors equally contributed to this work
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- These authors equally contributed to this work
| |
Collapse
|
4
|
Rauen KA, Tidyman WE. RASopathies - what they reveal about RAS/MAPK signaling in skeletal muscle development. Dis Model Mech 2024; 17:dmm050609. [PMID: 38847227 PMCID: PMC11179721 DOI: 10.1242/dmm.050609] [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] [Indexed: 06/12/2024] Open
Abstract
RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
Collapse
Affiliation(s)
- Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, 95817, USA
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| | - William E Tidyman
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
5
|
Huang Y, Sun H, Chen Q, Shen J, Han J, Shan S, Wang S. Computer-based facial recognition as an assisting diagnostic tool to identify children with Noonan syndrome. BMC Pediatr 2024; 24:361. [PMID: 38783283 PMCID: PMC11118109 DOI: 10.1186/s12887-024-04827-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Noonan syndrome (NS) is a rare genetic disease, and patients who suffer from it exhibit a facial morphology that is characterized by a high forehead, hypertelorism, ptosis, inner epicanthal folds, down-slanting palpebral fissures, a highly arched palate, a round nasal tip, and posteriorly rotated ears. Facial analysis technology has recently been applied to identify many genetic syndromes (GSs). However, few studies have investigated the identification of NS based on the facial features of the subjects. OBJECTIVES This study develops advanced models to enhance the accuracy of diagnosis of NS. METHODS A total of 1,892 people were enrolled in this study, including 233 patients with NS, 863 patients with other GSs, and 796 healthy children. We took one to 10 frontal photos of each subject to build a dataset, and then applied the multi-task convolutional neural network (MTCNN) for data pre-processing to generate standardized outputs with five crucial facial landmarks. The ImageNet dataset was used to pre-train the network so that it could capture generalizable features and minimize data wastage. We subsequently constructed seven models for facial identification based on the VGG16, VGG19, VGG16-BN, VGG19-BN, ResNet50, MobileNet-V2, and squeeze-and-excitation network (SENet) architectures. The identification performance of seven models was evaluated and compared with that of six physicians. RESULTS All models exhibited a high accuracy, precision, and specificity in recognizing NS patients. The VGG19-BN model delivered the best overall performance, with an accuracy of 93.76%, precision of 91.40%, specificity of 98.73%, and F1 score of 78.34%. The VGG16-BN model achieved the highest AUC value of 0.9787, while all models based on VGG architectures were superior to the others on the whole. The highest scores of six physicians in terms of accuracy, precision, specificity, and the F1 score were 74.00%, 75.00%, 88.33%, and 61.76%, respectively. The performance of each model of facial recognition was superior to that of the best physician on all metrics. CONCLUSION Models of computer-assisted facial recognition can improve the rate of diagnosis of NS. The models based on VGG19-BN and VGG16-BN can play an important role in diagnosing NS in clinical practice.
Collapse
Affiliation(s)
- Yulu Huang
- Department of Pediatric Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, China
| | - Haomiao Sun
- Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, No. 6 South Science Academy Road, Haidian District, Beijing, China
- University of Chinese Academy of Sciences, No. 80 Zhongguancun Road East, Haidian District, Beijing, China
| | - Qinchang Chen
- Department of Pediatric Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No. 106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, China
| | - Junjun Shen
- Department of Pediatric Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No. 106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, China
| | - Jin Han
- Prenatal diagnosis center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, China
| | - Shiguang Shan
- Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, No. 6 South Science Academy Road, Haidian District, Beijing, China.
- University of Chinese Academy of Sciences, No. 80 Zhongguancun Road East, Haidian District, Beijing, China.
| | - Shushui Wang
- Department of Pediatric Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, China.
- Department of Pediatric Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No. 106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, China.
| |
Collapse
|
6
|
Rodríguez-Martín M, Báez-Flores J, Ribes V, Isidoro-García M, Lacal J, Prieto-Matos P. Non-Mammalian Models for Understanding Neurological Defects in RASopathies. Biomedicines 2024; 12:841. [PMID: 38672195 PMCID: PMC11048513 DOI: 10.3390/biomedicines12040841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
RASopathies, a group of neurodevelopmental congenital disorders stemming from mutations in the RAS/MAPK pathway, present a unique opportunity to delve into the intricacies of complex neurological disorders. Afflicting approximately one in a thousand newborns, RASopathies manifest as abnormalities across multiple organ systems, with a pronounced impact on the central and peripheral nervous system. In the pursuit of understanding RASopathies' neurobiology and establishing phenotype-genotype relationships, in vivo non-mammalian models have emerged as indispensable tools. Species such as Danio rerio, Drosophila melanogaster, Caenorhabditis elegans, Xenopus species and Gallus gallus embryos have proven to be invaluable in shedding light on the intricate pathways implicated in RASopathies. Despite some inherent weaknesses, these genetic models offer distinct advantages over traditional rodent models, providing a holistic perspective on complex genetics, multi-organ involvement, and the interplay among various pathway components, offering insights into the pathophysiological aspects of mutations-driven symptoms. This review underscores the value of investigating the genetic basis of RASopathies for unraveling the underlying mechanisms contributing to broader neurological complexities. It also emphasizes the pivotal role of non-mammalian models in serving as a crucial preliminary step for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Vanessa Ribes
- Institut Jacques Monod, Université Paris Cité, CNRS, F-75013 Paris, France;
| | - María Isidoro-García
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Biochemistry Department, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Pablo Prieto-Matos
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Pediatrics, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Science, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
7
|
Smith CR, Chen D, Christensen JG, Coulombe R, Féthière J, Gunn RJ, Hollander J, Jones B, Ketcham JM, Khare S, Kuehler J, Lawson JD, Marx MA, Olson P, Pearson KE, Ren C, Tsagris D, Ulaganathan T, Van’t Veer I, Wang X, Ivetac A. Discovery of Five SOS2 Fragment Hits with Binding Modes Determined by SOS2 X-Ray Cocrystallography. J Med Chem 2024; 67:774-781. [PMID: 38156904 PMCID: PMC10788894 DOI: 10.1021/acs.jmedchem.3c02140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
SOS1 and SOS2 are guanine nucleotide exchange factors that mediate RTK-stimulated RAS activation. Selective SOS1:KRAS PPI inhibitors are currently under clinical investigation, whereas there are no reports to date of SOS2:KRAS PPI inhibitors. SOS2 activity is implicated in MAPK rebound when divergent SOS1 mutant cell lines are treated with the SOS1 inhibitor BI-3406; therefore, SOS2:KRAS inhibitors are of therapeutic interest. In this report, we detail a fragment-based screening strategy to identify X-ray cocrystal structures of five diverse fragment hits bound to SOS2.
Collapse
Affiliation(s)
| | - Dan Chen
- ZoBio
BV, J.H. Oortweg 19, Leiden 2333 CH, Netherlands
| | | | - René Coulombe
- Inixium, 3000-275 Armand Frappier, Laval, Quebec H7V 4A7, Canada
| | - James Féthière
- Inixium, 3000-275 Armand Frappier, Laval, Quebec H7V 4A7, Canada
| | - Robin J. Gunn
- Mirati
Therapeutics, San Diego, California 92130, United States
| | | | - Benjamin Jones
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - John M. Ketcham
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - Shilpi Khare
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - Jon Kuehler
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - J. David Lawson
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - Matthew A. Marx
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - Peter Olson
- Mirati
Therapeutics, San Diego, California 92130, United States
| | | | - Cynthia Ren
- Mirati
Therapeutics, San Diego, California 92130, United States
| | | | | | | | - Xiaolun Wang
- Mirati
Therapeutics, San Diego, California 92130, United States
| | - Anthony Ivetac
- Mirati
Therapeutics, San Diego, California 92130, United States
| |
Collapse
|
8
|
Lin Q, Qiu M, Wei X, Xiang Z, Zhou Z, Ji I, Liang X, Zhou X, Wen Q, Liu Y, Yu H. Genetic variants of SOS2, MAP2K1 and RASGRF2 in the RAS pathway genes predict survival of HBV-related hepatocellular carcinoma patients. Arch Toxicol 2023; 97:1599-1611. [PMID: 37029817 DOI: 10.1007/s00204-023-03469-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/16/2023] [Indexed: 04/09/2023]
Abstract
The RAS pathway participates in the cascade of proliferation and cell division process, and the activated RAS pathway can lead to tumorigenesis including hepatocellular carcinoma (HCC). However, few studies have explored the effects of genetic variants in the RAS pathway-related genes on the survival of patients with HBV-related HCC. In the present study, we assessed the associations between 11,658 single-nucleotide polymorphisms (SNPs) in 62 RAS pathway genes and the overall survival (OS) of 866 HBV-related HCC individuals, which were randomly split (1:1) into discovery and validation datasets. As a result, three potentially functional SNPs were identified, based on multivariable cox proportional hazards regression analyses, in SOS Ras/Rho guanine nucleotide exchange factor 2 (SOS2, rs4632055 A > G), Ras protein-specific guanine nucleotide releasing factor 2 (RASGRF2, rs26418A > G) and mitogen-activated protein kinase 1 (MAP2K1,rs57120695 C > T), which were significantly and independently associated with OS of HBV-related HCC patients [adjusted hazards ratios (HRs) of 1.42, 1.32 and 1.50, respectively; 95% confidence intervals (CI), 1.14 to 1.76, 1.15 to 1.53 and 1.15 to 1.97, respectively; P = 0.001, < 0.001 and 0.003, respectively]. Additionally, the joint effects as the unfavorable genotypes of these three SNPs showed a significant association with the poor survival of HCC (trend test P < 0.001). The expression quantitative trait loci (eQTL) analysis further revealed that the rs4632055 G allele and the rs26418 A allele were associated with lower mRNA expression levels of SOS2 and RASGRF2, respectively. Collectively, these potentially functional SNPs of RASGRF2, SOS2 and M2PAK1 may become potential prognostic biomarkers for HBV-related HCC after hepatectomy.
Collapse
Affiliation(s)
- Qiuling Lin
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Moqin Qiu
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xueyan Wei
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Zhouyun Xiang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Zihan Zhou
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Iiangyan Ji
- Department of Scientific Research Dept, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiumei Liang
- Department of Disease Process Management, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xianguo Zhou
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Qiuping Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yingchun Liu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
- Key Cultivated Laboratory of Cancer Molecular Medicine, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Hongping Yu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
- Key Cultivated Laboratory of Cancer Molecular Medicine, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China.
| |
Collapse
|
9
|
Weaver KN, Gripp KW. Central nervous system involvement in individuals with RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:494-500. [PMID: 36454176 DOI: 10.1002/ajmg.c.32023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/05/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
Central nervous system (CNS) anomalies are common in individuals with RASopathies. While certain findings, including relative or absolute macrocephaly, are typical for most RASopathies, other findings are more common in certain conditions, with rare low-grade gliomas in Noonan syndrome (NS); Chiari 1 malformation and tethered cord in Costello syndrome (CS); and variable structural anomalies including heterotopia and hydrocephalus in cardio-facio-cutaneous syndrome (CFC). We performed a literature review and present aggregate data on the common and uncommon CNS manifestations in individuals with RASopathies. A gene-based approach to defining risk for specific abnormalities may be considered. However, limited information on the CNS findings of rare RASopathies, such as autosomal recessive LZTR1-related NS or PPP1CB-related NS with loose anagen hair (NSLH), is currently available. Thus, consideration of the RASopathies as a group of distinct syndromic conditions with shared underlying causes and overlapping clinical presentations remains relevant, and individuals with a RASopathy are at risk for many findings seen in these conditions.
Collapse
Affiliation(s)
- K Nicole Weaver
- Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Karen W Gripp
- Division of Medical Genetics, Nemours Children's Hospital, Wilmington, Delaware, USA.,S. Kimmel Medical College, T. Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Delogu AB, Limongelli G, Versacci P, Adorisio R, Kaski JP, Blandino R, Maiolo S, Monda E, Putotto C, De Rosa G, Chatfield KC, Gelb BD, Calcagni G. The heart in RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:440-451. [PMID: 36408797 DOI: 10.1002/ajmg.c.32014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/11/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022]
Abstract
The cardiovascular phenotype associated with RASopathies has expanded far beyond the original descriptions of pulmonary valve stenosis by Dr Jaqueline Noonan in 1968 and hypertrophic cardiomyopathy by Hirsch et al. in 1975. Because of the common underlying RAS/MAPK pathway dysregulation, RASopathy syndromes usually present with a typical spectrum of overlapping cardiovascular anomalies, although less common cardiac defects can occur. The identification of the causative genetic variants has enabled the recognition of specific correlations between genotype and cardiac phenotype. Characterization and understanding of genotype-phenotype associations is not only important for counseling a family of an infant with a new diagnosis of a RASopathy condition but is also critical for their clinical prognosis with respect to cardiac disease, neurodevelopment and other organ system involvement over the lifetime of the patient. This review will focus on the cardiac manifestations of the most common RASopathy syndromes, the relationship between cardiac defects and causal genetic variation, the contribution of cardiovascular abnormalities to morbidity and mortality and the most relevant follow-up issues for patients affected by RAS/MAPK pathway diseases, with respect to cardiac clinical outcomes and management, in children and in the adult population.
Collapse
Affiliation(s)
- Angelica Bibiana Delogu
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paolo Versacci
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Rachele Adorisio
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Juan Pablo Kaski
- Centre for Pediatric Inherited and Rare Cardiovascular Disease, University College London Institute of Cardiovascular Science, London, UK
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London, UK
| | | | - Stella Maiolo
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
- Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Emanuele Monda
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Carolina Putotto
- Pediatric Cardiology Unit, Department of Pediatrics, Obstetrics and Gynecology, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Gabriella De Rosa
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Kathryn C Chatfield
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Departments of Pediatrics and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Giulio Calcagni
- European Reference Network for rare, low-prevalence, or complex disease of the heart (ERN GUARD-Heart), University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy. Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Cardiac Surgery, Cardiology, Heart and Lung Transplantation, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| |
Collapse
|
11
|
Leoni C, Neri G. Molecular advances, clinical management, and treatment opportunities in RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:411-413. [PMID: 36541914 DOI: 10.1002/ajmg.c.32026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Giovanni Neri
- Institute of Genomic Medicine, Catholic University School of Medicine, Rome, Italy
| |
Collapse
|
12
|
Tartaglia M, Aoki Y, Gelb BD. The molecular genetics of RASopathies: An update on novel disease genes and new disorders. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:425-439. [PMID: 36394128 PMCID: PMC10100036 DOI: 10.1002/ajmg.c.32012] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/18/2022]
Abstract
Enhanced signaling through RAS and the mitogen-associated protein kinase (MAPK) cascade underlies the RASopathies, a family of clinically related disorders affecting development and growth. In RASopathies, increased RAS-MAPK signaling can result from the upregulated activity of various RAS GTPases, enhanced function of proteins positively controlling RAS function or favoring the efficient transmission of RAS signaling to downstream transducers, functional upregulation of RAS effectors belonging to the MAPK cascade, or inefficient signaling switch-off operated by feedback mechanisms acting at different levels. The massive effort in RASopathy gene discovery performed in the last 20 years has identified more than 20 genes implicated in these disorders. It has also facilitated the characterization of several molecular activating mechanisms that had remained unappreciated due to their minor impact in oncogenesis. Here, we provide an overview on the discoveries collected during the last 5 years that have delivered unexpected insights (e.g., Noonan syndrome as a recessive disease) and allowed to profile new RASopathies, novel disease genes and new molecular circuits contributing to the control of RAS-MAPK signaling.
Collapse
Affiliation(s)
- Marco Tartaglia
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - Yoko Aoki
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Bruce D. Gelb
- Mindich Child Health and Development InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pediatrics and GeneticsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
13
|
Swarts JW, Kleimeier LER, Leenders EKSM, Rinne T, Klein WM, Draaisma JMT. Lymphatic anomalies during lifetime in patients with Noonan syndrome: Retrospective cohort study. Am J Med Genet A 2022; 188:3242-3261. [PMID: 35979676 PMCID: PMC9804719 DOI: 10.1002/ajmg.a.62955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 01/31/2023]
Abstract
Noonan syndrome (NS) has been associated with an increased risk of lymphatic anomalies, with an estimated prevalence of 20%. The prevalence of lymphatic anomalies seems to differ between pathogenic variants. Therefore, this study aims to describe the clinical presentation, prevalence and genotype-phenotype correlations of lymphatic anomalies during life in patients with NS. This retrospective cohort study included patients (n = 115) who were clinically and genetically diagnosed with NS and visited the Noonan expertise Center of the Radboud University Medical Center between January 2015 and March 2021. Data on lymphatic anomalies during lifetime were obtained from medical records. Lymphatic anomalies most often presented as an increased nuchal translucency, chylothorax and/or lymphedema. Prenatal lymphatic anomalies increased the risk of lymphatic anomalies during infancy (OR 4.9, 95% CI 1.7-14.6). The lifetime prevalence of lymphatic anomalies was 37%. Genotype-phenotype correlations showed an especially high prevalence of lymphatic anomalies during infancy and childhood in patients with a pathogenic SOS2 variant (p = 0.03 and p < 0.01, respectively). This study shows that patients with NS have a high predisposition for developing lymphatic anomalies during life. Especially patients with prenatal lymphatic anomalies have an increased risk of lymphatic anomalies during infancy. Genotype-phenotype correlations were found in pathogenic variants in SOS2.
Collapse
Affiliation(s)
- Jessie W. Swarts
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenNetherlands
| | - Lotte E. R. Kleimeier
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenNetherlands
| | | | - Tuula Rinne
- Department of Human Genetics, Donders Institute for Brain, Cognition and BehaviorRadboud University Medical CenterNijmegenNetherlands
| | - Willemijn M. Klein
- Department of Medical ImagingRadboud University Medical CenterNijmegenNetherlands
| | - Jos M. T. Draaisma
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenNetherlands
| |
Collapse
|
14
|
Bajia D, Bottani E, Derwich K. Effects of Noonan Syndrome-Germline Mutations on Mitochondria and Energy Metabolism. Cells 2022; 11:cells11193099. [PMID: 36231062 PMCID: PMC9563972 DOI: 10.3390/cells11193099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.
Collapse
Affiliation(s)
- Donald Bajia
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Piazzale L. A. Scuro 10, 37134 Verona, Italy
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| |
Collapse
|
15
|
Wingbermühle E, Roelofs RL, Oomens W, Kramer J, Draaisma JMT, Leenders E, Kleefstra T, Kessels RPC, Egger JIM. Cognitive Phenotype and Psychopathology in Noonan Syndrome Spectrum Disorders through Various Ras/MAPK Pathway Associated Gene Variants. J Clin Med 2022; 11:jcm11164735. [PMID: 36012976 PMCID: PMC9410383 DOI: 10.3390/jcm11164735] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Cognitive difficulties are argued to be common in patients with Noonan syndrome spectrum disorders (NSSDs), but findings are based on studies in which patients with variants in PTPN11 (prevalence ~50%) were overrepresented. The current study, using a structured clinical approach, describes the cognitive phenotype and psychopathology of 100 patients (aged 6 to 61 years) with nine different gene variants in the Ras/MAPK pathway underlying NSSDs (PTPN11n = 61, PTPN11 Noonan syndrome with multiple lentigines n = 3, SOS1n = 14, KRASn = 7, LZTR1n = 5, RAF1n = 4, SHOC2n = 2, CBLn = 2, SOS2n = 2). After weighted assessment and bootstrapping of the results of individual neuropsychological assessments and measures of psychopathology, cognitive performances in most variant groups were within the ranges of expectation. IQs were significantly lower in patients with variants in PTPN11, KRAS, RAF1, and SHOC2, but no specific cognitive impairments were found. The performances of younger participants (<16 years of age) did not differ from those of adults. Alexithymia and internalizing problems were more frequent in patients with variants in PTPN11 and SOS1, while PTPN11 patients also showed higher levels of externalizing problems. These results stress the need to take intelligence into account when interpreting lower cognitive performances in individual neuropsychological assessments, which is crucial for an adequate understanding and guidance of patients with NSSDs.
Collapse
Affiliation(s)
- Ellen Wingbermühle
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Correspondence:
| | - Renée L. Roelofs
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Wouter Oomens
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
| | - Jennifer Kramer
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
| | - Jos M. T. Draaisma
- Department of Pediatrics, Amalia Children’s Hospital, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Erika Leenders
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Roy P. C. Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Center of Excellence for Korsakoff and Alcohol-Related Cognitive Disorders, Vincent van Gogh Institute for Psychiatry, 5803 DN Venray, The Netherlands
- Department of Medical Psychology and Radboudumc Alzheimer Center, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Jos I. M. Egger
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Center of Excellence for Korsakoff and Alcohol-Related Cognitive Disorders, Vincent van Gogh Institute for Psychiatry, 5803 DN Venray, The Netherlands
| |
Collapse
|
16
|
Abstract
RAS proteins play major roles in many human cancers, but programs to develop direct RAS inhibitors so far have only been successful for the oncogenic KRAS mutant G12C. As an alternative approach, inhibitors for the RAS guanine nucleotide exchange factor SOS1 have been investigated by several academic groups and companies, and major progress has been achieved in recent years in the optimization of small molecule activators and inhibitors of SOS1. Here, we review the discovery and development of small molecule modulators of SOS1 and their molecular binding modes and modes of action. As targeting the RAS pathway is expected to result in the development of resistance mechanisms, SOS1 inhibitors will most likely be best applied in vertical combination approaches where two nodes of the RAS signaling pathway are hit simultaneously. We summarize the current understanding of which combination partners may be most beneficial for patients with RAS driven tumors.
Collapse
Affiliation(s)
| | - Benjamin Bader
- Screening, Lead Discovery, Nuvisan ICB GmbH, Berlin, Germany
| |
Collapse
|
17
|
Abstract
The RASopathies are a group of disorders caused by a germline mutation in one of the genes encoding a component of the RAS/MAPK pathway. These disorders, including neurofibromatosis type 1, Noonan syndrome, cardiofaciocutaneous syndrome, Costello syndrome and Legius syndrome, among others, have overlapping clinical features due to RAS/MAPK dysfunction. Although several of the RASopathies are very rare, collectively, these disorders are relatively common. In this Review, we discuss the pathogenesis of the RASopathy-associated genetic variants and the knowledge gained about RAS/MAPK signaling that resulted from studying RASopathies. We also describe the cell and animal models of the RASopathies and explore emerging RASopathy genes. Preclinical and clinical experiences with targeted agents as therapeutics for RASopathies are also discussed. Finally, we review how the recently developed drugs targeting RAS/MAPK-driven malignancies, such as inhibitors of RAS activation, direct RAS inhibitors and RAS/MAPK pathway inhibitors, might be leveraged for patients with RASopathies.
Collapse
Affiliation(s)
- Katie E Hebron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Edjay Ralph Hernandez
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Marielle E Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Zhou Y, Bai K, Wang Y, Meng Z, Zhou S, Jiang S, Wang H, Wang J, Yang M, Wang Q, Sun K, Chen S. Identification of Rare Variants in Right Ventricular Outflow Tract Obstruction Congenital Heart Disease by Whole-Exome Sequencing. Front Cardiovasc Med 2022; 8:811156. [PMID: 35141295 PMCID: PMC8818757 DOI: 10.3389/fcvm.2021.811156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/30/2021] [Indexed: 11/18/2022] Open
Abstract
Background Pulmonary atresia (PA) is a kind of congenital heart disease characterized by right ventricular outflow tract obstruction. It is divided into PA with intact ventricular septum (PA/IVS) whose favorable form is pulmonary valvular stenosis (PS), and PA with ventricular septal defect (PA/VSD) whose favorable form is tetralogy of Fallot (TOF). Due to limitations in genetics etiology, whole-exome sequencing (WES) was utilized to identify new variants associated with the diseases. Methods The data from PS-PA/IVS (n = 74), TOF-PA/VSD (n = 100), and 100 controls were obtained. The common sites between PS and PA/IVS, PA/VSD and TOF, were compared. The novel rare damage variants, and candidate genes were identified by gene-based burden analysis. Finally, the enrichment analysis of differential genes was conducted between case and control groups. Results Seventeen rare damage variants located in seven genes were predicted to be associated with the PS through burden analysis. Enrichment analysis identified that the Wnt and cadherin signaling pathways were relevant to PS-PA/IVS. Conclusion This study put forth seven candidate genes (APC, PPP1R12A, PCK2, SOS2, TNR, MED13, and TIAM1), resulting in PS-PA/IVS. The Wnt and cadherin signaling pathways were identified to be related to PS-PA/IVS by enrichment analysis. This study provides new evidence for exploring the genetic mechanism of PS-PA/IVS.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Bai
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatric Cardiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuo Meng
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuang Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiwei Jiang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hualin Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mei Yang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Qingjie Wang
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Kun Sun
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Sun Chen
| |
Collapse
|
19
|
Dussueil P, Sergent JF, Veyssière A, Benateau H. Recurrent labial xanthoma infection in a patient with Neurofibromatosis-Noonan syndrome: case report and literature review. JOURNAL OF ORAL MEDICINE AND ORAL SURGERY 2022. [DOI: 10.1051/mbcb/2022029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Noonan Syndrome is a clinically and genetically heterogeneous syndrome, characterized by marked phenotypic variability. All the clinical manifestations of this syndrome are still not fully known. Observation: We present the case of a 58-year-old woman with a diagnosis of Neurofibromatosis-Noonan syndrome with SOS2 mutation, observed by her general practitioner for a recurrent left upper lip abscess despite drainage and antibiotic therapy. The anatomo-pathological result of the sample was in favor of an infected xanthoma. Discussion: The most common oral manifestation of Noonan syndrome includes malocclusion, dental anomalies and radiologic jaw lesions. Xanthomas of the lip have never been reported in this syndrome. Conclusion: Oral xanthomas could be one of the many oral clinical manifestations of Noonan Syndrome. However, more research is needed to understand clinical consequences of mutations in identified genes.
Collapse
|
20
|
Lioncino M, Monda E, Verrillo F, Moscarella E, Calcagni G, Drago F, Marino B, Digilio MC, Putotto C, Calabrò P, Russo MG, Roberts AE, Gelb BD, Tartaglia M, Limongelli G. Hypertrophic Cardiomyopathy in RASopathies: Diagnosis, Clinical Characteristics, Prognostic Implications, and Management. Heart Fail Clin 2022; 18:19-29. [PMID: 34776080 PMCID: PMC9674037 DOI: 10.1016/j.hfc.2021.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RASopathies are multisystemic disorders caused by germline mutations in genes linked to the RAS/mitogen-activated protein kinase pathway. Diagnosis of RASopathy can be triggered by clinical clues ("red flags") which may direct the clinician toward a specific gene test. Compared with sarcomeric hypertrophic cardiomyopathy, hypertrophic cardiomyopathy in RASopathies (R-HCM) is associated with higher prevalence of congestive heart failure and shows increased prevalence and severity of left ventricular outflow tract obstruction. Biventricular involvement and the association with congenital heart disease, mainly pulmonary stenosis, have been commonly described in R-HCM. The aim of this review is to assess the prevalence and unique features of R-HCM and to define the available therapeutic options.
Collapse
Affiliation(s)
- Michele Lioncino
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples
| | - Emanuele Monda
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples
| | - Federica Verrillo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples;,Division of Cardiology, A.O.R.N. “Sant’Anna & San Sebastiano”, Caserta I-81100, Italy
| | - Giulio Calcagni
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart - ERN GUARD-Heart;,Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children’s Hospital IRCSS, Rome, Italy
| | - Fabrizio Drago
- The European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart - ERN GUARD-Heart;,Pediatric Cardiology and Arrhythmia/Syncope Units, Bambino Gesù Children’s Hospital IRCSS, Rome, Italy
| | - Bruno Marino
- Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Maria Cristina Digilio
- Genetics and Rare Disease Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Carolina Putotto
- Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples;,Division of Cardiology, A.O.R.N. “Sant’Anna & San Sebastiano”, Caserta I-81100, Italy
| | - Maria Giovanna Russo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples;,Department of Pediatric Cardiology, AORN dei Colli, Monaldi Hospital, Naples
| | - Amy E. Roberts
- Department of Cardiology, Children’s Hospital Boston, Boston, MA, USA
| | - Bruce D. Gelb
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marco Tartaglia
- Genetics and Rare Disease Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples;,Division of Cardiology, A.O.R.N. “Sant’Anna & San Sebastiano”, Caserta I-81100, Italy;,Corresponding author. Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples.
| |
Collapse
|
21
|
Brouillard P, Witte MH, Erickson RP, Damstra RJ, Becker C, Quéré I, Vikkula M. Primary lymphoedema. Nat Rev Dis Primers 2021; 7:77. [PMID: 34675250 DOI: 10.1038/s41572-021-00309-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 11/09/2022]
Abstract
Lymphoedema is the swelling of one or several parts of the body owing to lymph accumulation in the extracellular space. It is often chronic, worsens if untreated, predisposes to infections and causes an important reduction in quality of life. Primary lymphoedema (PLE) is thought to result from abnormal development and/or functioning of the lymphatic system, can present in isolation or as part of a syndrome, and can be present at birth or develop later in life. Mutations in numerous genes involved in the initial formation of lymphatic vessels (including valves) as well as in the growth and expansion of the lymphatic system and associated pathways have been identified in syndromic and non-syndromic forms of PLE. Thus, the current hypothesis is that most cases of PLE have a genetic origin, although a causative mutation is identified in only about one-third of affected individuals. Diagnosis relies on clinical presentation, imaging of the structure and functionality of the lymphatics, and in genetic analyses. Management aims at reducing or preventing swelling by compression therapy (with manual drainage, exercise and compressive garments) and, in carefully selected cases, by various surgical techniques. Individuals with PLE often have a reduced quality of life owing to the psychosocial and lifelong management burden associated with their chronic condition. Improved understanding of the underlying genetic origins of PLE will translate into more accurate diagnosis and prognosis and personalized treatment.
Collapse
Affiliation(s)
- Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Marlys H Witte
- Department of Surgery, Neurosurgery, and Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Robert P Erickson
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Robert J Damstra
- VASCERN PPL European Reference Centre; Department of Dermatology, Phlebology and Lymphology, Nij Smellinghe Hospital, Drachten, Netherlands
| | | | - Isabelle Quéré
- Department of Vascular Medicine, Centre de référence des Maladies Lymphatiques et Vasculaires Rares, Inserm IDESP, CHU Montpellier, Université de Montpellier, Montpellier, France
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium. .,VASCERN VASCA European Reference Centre; Center for Vascular Anomalies, Division of Plastic Surgery, University Clinics Saint-Luc, University of Louvain, Brussels, Belgium. .,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), de Duve Institute, University of Louvain, Brussels, Belgium.
| |
Collapse
|
22
|
Leoni C, Blandino R, Delogu AB, De Rosa G, Onesimo R, Verusio V, Marino MV, Lanza GA, Rigante D, Tartaglia M, Zampino G. Genotype-cardiac phenotype correlations in a large single-center cohort of patients affected by RASopathies: Clinical implications and literature review. Am J Med Genet A 2021; 188:431-445. [PMID: 34643321 DOI: 10.1002/ajmg.a.62529] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/11/2021] [Accepted: 09/21/2021] [Indexed: 11/07/2022]
Abstract
Congenital heart disease (CHD) and hypertrophic cardiomyopathy (HCM) are common features in patients affected by RASopathies. The aim of this study was to assess genotype- phenotype correlations, focusing on the cardiac features and outcomes of interventions for cardiac conditions, in a single-center cohort of 116 patients with molecularly confirmed diagnosis of RASopathy, and compare these findings with previously published data. All enrolled patients underwent a comprehensive echocardiographic examination. Relevant information was also retrospectively collected through the analysis of clinical records. As expected, significant associations were found between PTPN11 mutations and pulmonary stenosis (both valvular and supravalvular) and pulmonary valve dysplasia, and between SOS1 mutations and valvular defects. Similarly, HRAS mutations were significantly associated with HCM. Potential associations between less prevalent mutations and cardiac defects were also observed, including RIT1 mutations and HCM, SOS2 mutations and septal defects, and SHOC2 mutations and septal and valve abnormalities. Patients with PTPN11 mutations were the most likely to require both a primary treatment (transcatheter or surgical) and surgical reintervention. Other cardiac anomalies less reported until recently in this population, such as isolated functional and structural mitral valve diseases, as well as a sigmoid-shaped interventricular septum in the absence of HCM, were also reported. In conclusion, our study confirms previous data but also provides new insights on cardiac involvement in RASopathies. Further research concerning genotype/phenotype associations in RASopathies could lead to a more rational approach to surgery and the consideration of drug therapy in patients at higher risk due to age, severity, anatomy, and comorbidities.
Collapse
Affiliation(s)
- Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Rita Blandino
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angelica Bibiana Delogu
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gabriella De Rosa
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Valeria Verusio
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Vittoria Marino
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Donato Rigante
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
23
|
Longo JF, Carroll SL. The RASopathies: Biology, genetics and therapeutic options. Adv Cancer Res 2021; 153:305-341. [PMID: 35101235 DOI: 10.1016/bs.acr.2021.07.007] [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] [Indexed: 10/19/2022]
Abstract
The RASopathies are a group of genetic diseases in which the Ras/MAPK signaling pathway is inappropriately activated as a result of mutations in genes encoding proteins within this pathway. As their causative mutations have been identified, this group of diseases has expanded to include neurofibromatosis type 1 (NF1), Legius syndrome, Noonan syndrome, CBL syndrome, Noonan syndrome-like disorder with loose anagen hair, Noonan syndrome with multiple lentigines, Costello syndrome, cardiofaciocutaneous syndrome, gingival fibromatosis and capillary malformation-arteriovenous malformation syndrome. Many of these genetic disorders share clinical features in common such as abnormal facies, short stature, varying degrees of cognitive impairment, cardiovascular abnormalities, skeletal abnormalities and a predisposition to develop benign and malignant neoplasms. Others are more dissimilar, even though their mutations are in the same gene that is mutated in a different RASopathy. Here, we describe the clinical features of each RASopathy and contrast them with the other RASopathies. We discuss the genetics of these disorders, including the causative mutations for each RASopathy, the impact that these mutations have on the function of an individual protein and how this dysregulates the Ras/MAPK signaling pathway. As several of these individual disorders are genetically heterogeneous, we also consider the different genes that can be mutated to produce disease with the same phenotype. We also discuss how our growing understanding of dysregulated Ras/MAPK signaling had led to the development of new therapeutic agents and what work will be critically important in the future to improve the lives of patients with RASopathies.
Collapse
Affiliation(s)
- Jody Fromm Longo
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
24
|
Baltanás FC, García-Navas R, Santos E. SOS2 Comes to the Fore: Differential Functionalities in Physiology and Pathology. Int J Mol Sci 2021; 22:ijms22126613. [PMID: 34205562 PMCID: PMC8234257 DOI: 10.3390/ijms22126613] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
The SOS family of Ras-GEFs encompasses two highly homologous and widely expressed members, SOS1 and SOS2. Despite their similar structures and expression patterns, early studies of constitutive KO mice showing that SOS1-KO mutants were embryonic lethal while SOS2-KO mice were viable led to initially viewing SOS1 as the main Ras-GEF linking external stimuli to downstream RAS signaling, while obviating the functional significance of SOS2. Subsequently, different genetic and/or pharmacological ablation tools defined more precisely the functional specificity/redundancy of the SOS1/2 GEFs. Interestingly, the defective phenotypes observed in concomitantly ablated SOS1/2-DKO contexts are frequently much stronger than in single SOS1-KO scenarios and undetectable in single SOS2-KO cells, demonstrating functional redundancy between them and suggesting an ancillary role of SOS2 in the absence of SOS1. Preferential SOS1 role was also demonstrated in different RASopathies and tumors. Conversely, specific SOS2 functions, including a critical role in regulation of the RAS-PI3K/AKT signaling axis in keratinocytes and KRAS-driven tumor lines or in control of epidermal stem cell homeostasis, were also reported. Specific SOS2 mutations were also identified in some RASopathies and cancer forms. The relevance/specificity of the newly uncovered functional roles suggests that SOS2 should join SOS1 for consideration as a relevant biomarker/therapy target.
Collapse
|
25
|
Ras Isoforms from Lab Benches to Lives-What Are We Missing and How Far Are We? Int J Mol Sci 2021; 22:ijms22126508. [PMID: 34204435 PMCID: PMC8233758 DOI: 10.3390/ijms22126508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 11/21/2022] Open
Abstract
The central protein in the oncogenic circuitry is the Ras GTPase that has been under intense scrutiny for the last four decades. From its discovery as a viral oncogene and its non-oncogenic contribution to crucial cellular functioning, an elaborate genetic, structural, and functional map of Ras is being created for its therapeutic targeting. Despite decades of research, there still exist lacunae in our understanding of Ras. The complexity of the Ras functioning is further exemplified by the fact that the three canonical Ras genes encode for four protein isoforms (H-Ras, K-Ras4A, K-Ras4B, and N-Ras). Contrary to the initial assessment that the H-, K-, and N-Ras isoforms are functionally similar, emerging data are uncovering crucial differences between them. These Ras isoforms exhibit not only cell-type and context-dependent functions but also activator and effector specificities on activation by the same receptor. Preferential localization of H-, K-, and N-Ras in different microdomains of the plasma membrane and cellular organelles like Golgi, endoplasmic reticulum, mitochondria, and endosome adds a new dimension to isoform-specific signaling and diverse functions. Herein, we review isoform-specific properties of Ras GTPase and highlight the importance of considering these towards generating effective isoform-specific therapies in the future.
Collapse
|
26
|
Das TK, Gatto J, Mirmira R, Hourizadeh E, Kaufman D, Gelb BD, Cagan R. Drosophila RASopathy models identify disease subtype differences and biomarkers of drug efficacy. iScience 2021; 24:102306. [PMID: 33855281 PMCID: PMC8026909 DOI: 10.1016/j.isci.2021.102306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/30/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
RASopathies represent a family of mostly autosomal dominant diseases that are caused by missense variants in the rat sarcoma viral oncogene/mitogen activated protein kinase (RAS/MAPK) pathway including KRAS, NRAS, BRAF, RAF1, and SHP2. These variants are associated with overlapping but distinct phenotypes that affect the heart, craniofacial, skeletal, lymphatic, and nervous systems. Here, we report an analysis of 13 Drosophila transgenic lines, each expressing a different human RASopathy isoform. Similar to their human counterparts, each Drosophila line displayed common aspects but also important differences including distinct signaling pathways such as the Hippo and SAPK/JNK signaling networks. We identified multiple classes of clinically relevant drugs-including statins and histone deacetylase inhibitors-that improved viability across most RASopathy lines; in contrast, several canonical RAS pathway inhibitors proved less broadly effective. Overall, our study compares and contrasts a large number of RASopathy-associated variants including their therapeutic responses.
Collapse
Affiliation(s)
- Tirtha K. Das
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York NY, USA
- The Mindich Child Health and Development Institute, Department of Pediatrics, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Jared Gatto
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York NY, USA
- The Mindich Child Health and Development Institute, Department of Pediatrics, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Rupa Mirmira
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Ethan Hourizadeh
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Dalia Kaufman
- The Mindich Child Health and Development Institute, Department of Pediatrics, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Bruce D. Gelb
- The Mindich Child Health and Development Institute, Department of Pediatrics, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Ross Cagan
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
27
|
Gentile M, Fanelli T, Lepri FR, Gentile A, Orsini P, Volpe P, Novelli A, Ficarella R. First prenatal case of Noonan syndrome with SOS2 mutation: Implications of early diagnosis for genetic counseling. Am J Med Genet A 2021; 185:1897-1902. [PMID: 33750022 DOI: 10.1002/ajmg.a.62180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
RASopathies are a group of syndromes with partially overlapping clinical features caused by germline mutations of the RAS/MAPK signaling pathway genes. The most common disorder is Noonan syndrome (NS; MIM 163950). We report the first prenatal case of NS with SOS2 (NM_006939.4) mutation in a euploid fetus with a severe increase in nuchal translucency (NT > 12 mm). Trio-based custom next-generation sequencing detected a de novo heterozygous missense mutation in the SOS2 gene: c.800 T > A (p.Met267Lys). Owing to the marked variable expressivity of NS and the scarcity of SOS2 mutation-related NS cases reported in the literature, it is difficult to provide appropriate genetic counseling. Several issues such as the best management technique and optimal NT cutoff have been discussed. In addition, in general, the fine balance between the advantages of an early prenatal diagnosis and the challenge of determining if the detected gene variant is pathogenic and, primarily, the stress of the counselees when providing a genetic counseling with limited information on the prenatal phenotype have been discussed. A prenatal path comprising examinations and multidisciplinary counseling is essential to support couples in a shared decision-making process.
Collapse
Affiliation(s)
- Mattia Gentile
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Tiziana Fanelli
- Fetal Medicine Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Francesca Romana Lepri
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Angela Gentile
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Paola Orsini
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Paolo Volpe
- Fetal Medicine Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Romina Ficarella
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| |
Collapse
|
28
|
Hoang HM, Umutesi HG, Heo J. Allosteric autoactivation of SOS and its kinetic mechanism. Small GTPases 2021; 12:44-59. [PMID: 30983499 PMCID: PMC7781538 DOI: 10.1080/21541248.2019.1601954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/15/2019] [Accepted: 03/25/2019] [Indexed: 10/27/2022] Open
Abstract
Son of Sevenless (SOS), one of guanine nucleotide exchange factors (GEFs), activates Ras. We discovered that the allosteric domain of SOS yields SOS to proceed a previously unrecognized autoactivation kinetics. Its essential feature is a time-dependent acceleration of SOS feedback activation with a reaction initiator or with the priming of active Ras. Thus, this mechanistic autoactivation feature explains the notion, previously only conjectured, of accelerative SOS activation followed by the priming of active Ras, an action produced by another GEF Ras guanyl nucleotide-releasing protein (RasGRP). Intriguingly, the kinetic transition from gradual RasGRP activation to accelerative SOS activation has been interpreted as an analog to digital conversion; however, from the perspective of autoactivation kinetics, it is a process of straightforward RasGRP-mediated SOS autoactivation. From the viewpoint of allosteric protein cooperativity, SOS autoactivation is a unique time-dependent cooperative SOS activation because it enables an active SOS to accelerate activation of other SOS as a function of time. This time-dependent SOS cooperativity does not belong to the classic steady-state protein cooperativity, which depends on ligand concentration. Although its hysteretic or sigmoid-like saturation curvature is a classic hallmark of steady-state protein cooperativity, its hyperbolic saturation figure typically represents protein noncooperativity. We also discovered that SOS autoactivation perturbs the previously predicted hysteresis of SOS activation in a steady state to produce a hyperbolic saturation curve. We interpret this as showing that SOS allostery elicits, through SOS autoactivation, cooperativity uniquely time-dependent but not ligand concentration dependent.
Collapse
Affiliation(s)
- Hanh My Hoang
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Hope Gloria Umutesi
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Jongyun Heo
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
29
|
Sheffels E, Sealover NE, Theard PL, Kortum RL. Anchorage-independent growth conditions reveal a differential SOS2 dependence for transformation and survival in RAS-mutant cancer cells. Small GTPases 2021; 12:67-78. [PMID: 31062644 PMCID: PMC7781674 DOI: 10.1080/21541248.2019.1611168] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/15/2019] [Accepted: 04/20/2019] [Indexed: 02/07/2023] Open
Abstract
The RAS family of genes (HRAS, NRAS, and KRAS) is mutated in around 30% of human tumours. Wild-type RAS isoforms play an important role in mutant RAS-driven oncogenesis, indicating that RasGEFs may play a significant role in mutant RAS-driven transformation. We recently reported a hierarchical requirement for SOS2 in mutant RAS-driven transformation in mouse embryonic fibroblasts, with KRAS>NRAS>HRAS (Sheffels et al., 2018). However, whether SOS2 deletion differentially affects mutant RAS isoform-dependent transformation in human tumour cell lines has not been tested. After validating sgRNAs that efficiently deleted HRAS and NRAS, we showed that the differential requirement for SOS2 to support anchorage-independent (3D) growth, which we previously demonstrated in MEFs, held true in cancer cells. KRAS-mutant cells showed a high dependence on SOS2 for 3D growth, as previously shown, whereas HRAS-mutant cells did not require SOS2 for 3D growth. This differential requirement was not due to differences in RTK-stimulated WT RAS activation, as SOS2 deletion reduced RTK-stimulated WT RAS/PI3K/AKT signalling in both HRAS and KRAS mutated cell lines. Instead, this differential requirement of SOS2 to promote transformation was due to the differential sensitivity of RAS-mutated cancer cells to reductions in WT RAS/PI3K/AKT signalling. KRAS mutated cancer cells required SOS2/PI3K signaling to protect them from anoikis, whereas survival of both HRAS and NRAS mutated cancer cells was not altered by SOS2 deletion. Finally, we present an integrated working model of SOS signaling in the context of mutant KRAS based on our findings and those of others.
Collapse
Affiliation(s)
- Erin Sheffels
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nancy E. Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Patricia L. Theard
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert L. Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
30
|
Lores J, Prada CE, Ramírez-Montaño D, Nastasi-Catanese JA, Pachajoa H. Clinical and molecular analysis of 26 individuals with Noonan syndrome in a reference institution in Colombia. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:1042-1051. [PMID: 33300679 DOI: 10.1002/ajmg.c.31869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 11/06/2022]
Abstract
Our aim was to characterize the phenotype and genotype of individuals with Noonan syndrome in Colombia. There are published cohorts of Noonan individuals from several countries in Latin America including Brazil, Chile, and Argentina, but none from Colombia. We described 26 individuals with NS from a single large referral center in the South West of Colombia using an established database in the genetics department and hospital records search using ICD-10 codes. All patients included in this study were evaluated by a medical geneticist and have molecular confirmation of NS diagnosis. The median age at referral was 3.5 years (range, 0-39), and at molecular diagnosis was 5 years (range, 0-40). Patients mostly originated from the southwest region of Colombia (19/26, 73%). Pathogenic variants in PTPN11 are the most common cause of NS in Colombian individuals followed by SHOC2 and SOS1 variants. The prevalence of cardiomyopathy was low in this population compared to other populations. Further research is needed with a larger sample size and including different regions of Colombia to correlate our findings. This study provides new information about time to diagnosis of NS in Colombia, genotypes, and provides important information to help develop guidelines for diagnosis and management of this disease in the region.
Collapse
Affiliation(s)
- Juliana Lores
- Department of Genetics, Fundación Valle del Lili, Cali, Colombia.,Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| | - Carlos E Prada
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Fundación Cardiovascular de Colombia, Bucaramanga, Colombia
| | - Diana Ramírez-Montaño
- Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| | | | - Harry Pachajoa
- Department of Genetics, Fundación Valle del Lili, Cali, Colombia.,Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| |
Collapse
|
31
|
Baltanás FC, Zarich N, Rojas-Cabañeros JM, Santos E. SOS GEFs in health and disease. Biochim Biophys Acta Rev Cancer 2020; 1874:188445. [PMID: 33035641 DOI: 10.1016/j.bbcan.2020.188445] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
SOS1 and SOS2 are the most universal and widely expressed family of guanine exchange factors (GEFs) capable or activating RAS or RAC1 proteins in metazoan cells. SOS proteins contain a sequence of modular domains that are responsible for different intramolecular and intermolecular interactions modulating mechanisms of self-inhibition, allosteric activation and intracellular homeostasis. Despite their homology, analyses of SOS1/2-KO mice demonstrate functional prevalence of SOS1 over SOS2 in cellular processes including proliferation, migration, inflammation or maintenance of intracellular redox homeostasis, although some functional redundancy cannot be excluded, particularly at the organismal level. Specific SOS1 gain-of-function mutations have been identified in inherited RASopathies and various sporadic human cancers. SOS1 depletion reduces tumorigenesis mediated by RAS or RAC1 in mouse models and is associated with increased intracellular oxidative stress and mitochondrial dysfunction. Since WT RAS is essential for development of RAS-mutant tumors, the SOS GEFs may be considered as relevant biomarkers or therapy targets in RAS-dependent cancers. Inhibitors blocking SOS expression, intrinsic GEF activity, or productive SOS protein-protein interactions with cellular regulators and/or RAS/RAC targets have been recently developed and shown preclinical and clinical effectiveness blocking aberrant RAS signaling in RAS-driven and RTK-driven tumors.
Collapse
Affiliation(s)
- Fernando C Baltanás
- Centro de Investigación del Cáncer - IBMCC (CSIC-USAL) and CIBERONC, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Natasha Zarich
- Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC) and CIBERONC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Jose M Rojas-Cabañeros
- Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC) and CIBERONC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Eugenio Santos
- Centro de Investigación del Cáncer - IBMCC (CSIC-USAL) and CIBERONC, Universidad de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
32
|
Variants of SOS2 are a rare cause of Noonan syndrome with particular predisposition for lymphatic complications. Eur J Hum Genet 2020; 29:51-60. [PMID: 32788663 DOI: 10.1038/s41431-020-00708-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 11/09/2022] Open
Abstract
RASopathies are caused by variants in genes encoding components or modulators of the RAS/MAPK signaling pathway. Noonan syndrome is the most common entity among this group of disorders and is characterized by heart defects, short stature, variable developmental delay, and typical facial features. Heterozygous variants in SOS2, encoding a guanine nucleotide exchange factor for RAS, have recently been identified in patients with Noonan syndrome. The number of published cases with SOS2-related Noonan syndrome is still limited and little is known about genotype-phenotype correlations. We collected previously unpublished clinical and genotype data from 17 individuals carrying a disease-causing SOS2 variant. Most individuals had one of the previously reported dominant pathogenic variants; only four had novel changes at the established hotspots for variants that affect protein function. The overall phenotype of the 17 patients fits well into the spectrum of Noonan syndrome and is most similar to the phenotype observed in patients with SOS1-related Noonan syndrome, with ectodermal anomalies as common features and short stature and learning disabilities as relatively infrequent findings compared to the average Noonan syndrome phenotype. The spectrum of heart defects in SOS2-related Noonan syndrome was consistent with the known spectrum of cardiac anomalies in RASopathies, but no specific heart defect was particularly predominating. Notably, lymphatic anomalies were extraordinarily frequent, affecting more than half of the patients. We therefore conclude that SOS2-related Noonan syndrome is associated with a particularly high risk of lymphatic complications that may have a significant impact on morbidity and quality of life.
Collapse
|
33
|
Umutesi HG, Hoang HM, Johnson HE, Nam K, Heo J. Development of Noonan syndrome by deregulation of allosteric SOS autoactivation. J Biol Chem 2020; 295:13651-13663. [PMID: 32753483 DOI: 10.1074/jbc.ra120.013275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/30/2020] [Indexed: 11/06/2022] Open
Abstract
Ras family proteins play an essential role in several cellular functions, including growth, differentiation, and survival. The mechanism of action of Ras mutants in Costello syndrome and cancers has been identified, but the contribution of Ras mutants to Noonan syndrome, a genetic disorder that prevents normal development in various parts of the body, is unknown. Son of Sevenless (SOS) is a Ras guanine nucleotide exchange factor. In response to Ras-activating cell signaling, SOS autoinhibition is released and is followed by accelerative allosteric feedback autoactivation. Here, using mutagenesis-based kinetic and pulldown analyses, we show that Noonan syndrome Ras mutants I24N, T50I, V152G, and D153V deregulate the autoactivation of SOS to populate their active form. This previously unknown process has been linked so far only to the development of Noonan syndrome. In contrast, other Noonan syndrome Ras mutants-V14I, T58I, and G60E-populate their active form by deregulation of the previously documented Ras GTPase activities. We propose a novel mechanism responsible for the deregulation of SOS autoactivation, where I24N, T50I, V152G, and D153V Ras mutants evade SOS autoinhibition. Consequently, they are capable of forming a complex with the SOS allosteric site, thus aberrantly promoting SOS autoactivation, resulting in the population of active Ras mutants in cells. The results of this study elucidate the molecular mechanism of the Ras mutant-mediated development of Noonan syndrome.
Collapse
Affiliation(s)
- Hope Gloria Umutesi
- Department of Chemistry and Biochemistry, University of Texas, Arlington, Texas, USA
| | - Hanh My Hoang
- Department of Chemistry and Biochemistry, University of Texas, Arlington, Texas, USA
| | | | - Kwangho Nam
- Department of Chemistry and Biochemistry, University of Texas, Arlington, Texas, USA
| | - Jongyun Heo
- Department of Chemistry and Biochemistry, University of Texas, Arlington, Texas, USA.
| |
Collapse
|
34
|
Martinelli S, Pannone L, Lissewski C, Brinkmann J, Flex E, Schanze D, Calligari P, Anselmi M, Pantaleoni F, Canale VC, Radio FC, Ioannides A, Rahner N, Schanze I, Josifova D, Bocchinfuso G, Ryten M, Stella L, Tartaglia M, Zenker M. Pathogenic PTPN11 variants involving the poly-glutamine Gln 255 -Gln 256 -Gln 257 stretch highlight the relevance of helix B in SHP2's functional regulation. Hum Mutat 2020; 41:1171-1182. [PMID: 32112654 DOI: 10.1002/humu.24007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/14/2020] [Accepted: 02/27/2020] [Indexed: 01/28/2023]
Abstract
Germline PTPN11 mutations cause Noonan syndrome (NS), the most common disorder among RASopathies. PTPN11 encodes SHP2, a protein tyrosine-phosphatase controlling signaling through the RAS-MAPK and PI3K-AKT pathways. Generally, NS-causing PTPN11 mutations are missense changes destabilizing the inactive conformation of the protein or enhancing its binding to signaling partners. Here, we report on two PTPN11 variants resulting in the deletion or duplication of one of three adjacent glutamine residues (Gln255 -to-Gln257 ). While p.(Gln257dup) caused a typical NS phenotype in carriers of a first family, p.(Gln257del) had incomplete penetrance in a second family. Missense mutations involving Gln256 had previously been reported in NS. This poly-glutamine stretch is located on helix B of the PTP domain, a region involved in stabilizing SHP2 in its autoinhibited state. Molecular dynamics simulations predicted that changes affecting this motif perturb the SHP2's catalytically inactive conformation and/or substrate recognition. Biochemical data showed that duplication and deletion of Gln257 variably enhance SHP2's catalytic activity, while missense changes involving Gln256 affect substrate specificity. Expression of mutants in HEK293T cells documented their activating role on MAPK signaling, uncoupling catalytic activity and modulation of intracellular signaling. These findings further document the relevance of helix B in the regulation of SHP2's function.
Collapse
Affiliation(s)
- Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Pannone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Christina Lissewski
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Julia Brinkmann
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Denny Schanze
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Paolo Calligari
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Massimiliano Anselmi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Francesca Pantaleoni
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Viviana Claudia Canale
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | | | - Adonis Ioannides
- Clinical Genetics, University of Nicosia Medical School, Nicosia, Cyprus.,South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Nils Rahner
- Medical Faculty, Institute of Human Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Ina Schanze
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Dragana Josifova
- South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Mina Ryten
- South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Martin Zenker
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
35
|
Castellanos E, Rosas I, Negro A, Gel B, Alibés A, Baena N, Pineda M, Pi G, Pintos G, Salvador H, Lázaro C, Blanco I, Vilageliu L, Brems H, Grinberg D, Legius E, Serra E. Mutational spectrum by phenotype: panel‐based NGS testing of patients with clinical suspicion of RASopathy and children with multiple café‐au‐lait macules. Clin Genet 2019; 97:264-275. [DOI: 10.1111/cge.13649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Elisabeth Castellanos
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Inma Rosas
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Alex Negro
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Bernat Gel
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Andreu Alibés
- Cancer Genetics and Epigenetics GroupProgram of Predictive and Personalized Medicine of Cancer (PMPPC), Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Neus Baena
- Genetics Laboratory of the UDIAT‐CDParc Tauli Health Corporation Barcelona Spain
| | - Mercè Pineda
- Neuropaediatrics UnitHospital Sant Joan de Déu Barcelona Spain
| | - Graciela Pi
- Neuropaediatrics UnitLa Ribera Hospital Valencia Spain
| | - Guillem Pintos
- Department of PediatricsGermans Trias i Pujol University Hospital and Research Institute (IGTP), Universitat de Barcelona Barcelona Spain
| | - Hector Salvador
- Paediatrics Oncology UnitHospital Sant Joan de Déu Barcelona Spain
| | - Conxi Lázaro
- Hereditary Cancer ProgramCatalan Institute of Oncology (ICO‐IDIBELL‐ONCOBELL), L'Hospitalet de Llobregat Barcelona Spain
- Centro de Investigación Biomédica en RED (CIBERONC)Instituto de Salud Carlos III Madrid Spain
| | - Ignacio Blanco
- Clinical Genetics and Genetic Counseling ProgramGermans Trias i Pujol Hospital Barcelona Spain
| | - Lluïsa Vilageliu
- Department of Genetics, Microbiology and Statistics, Facultat de BiologiaUniversitat de Barcelona (UB), IBUB, IRSJD, CIBERER Barcelona Spain
| | - Hilde Brems
- Laboratory for Neurofibromatosis Research, Department of Human GeneticsKU Leuven University Hospital Leuven Belgium
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Facultat de BiologiaUniversitat de Barcelona (UB), IBUB, IRSJD, CIBERER Barcelona Spain
| | - Eric Legius
- Laboratory for Neurofibromatosis Research, Department of Human GeneticsKU Leuven University Hospital Leuven Belgium
| | - Eduard Serra
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
- Centro de Investigación Biomédica en RED (CIBERONC)Instituto de Salud Carlos III Madrid Spain
| |
Collapse
|
36
|
A report on a girl of Noonan syndrome 9 presenting with bilateral lower limbs lymphedema. Chin Med J (Engl) 2019; 132:480-482. [PMID: 30707178 PMCID: PMC6595711 DOI: 10.1097/cm9.0000000000000096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
37
|
Capri Y, Flex E, Krumbach OH, Carpentieri G, Cecchetti S, Lißewski C, Rezaei Adariani S, Schanze D, Brinkmann J, Piard J, Pantaleoni F, Lepri FR, Goh ESY, Chong K, Stieglitz E, Meyer J, Kuechler A, Bramswig NC, Sacharow S, Strullu M, Vial Y, Vignal C, Kensah G, Cuturilo G, Kazemein Jasemi NS, Dvorsky R, Monaghan KG, Vincent LM, Cavé H, Verloes A, Ahmadian MR, Tartaglia M, Zenker M. Activating Mutations of RRAS2 Are a Rare Cause of Noonan Syndrome. Am J Hum Genet 2019; 104:1223-1232. [PMID: 31130282 DOI: 10.1016/j.ajhg.2019.04.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/18/2019] [Indexed: 01/18/2023] Open
Abstract
Aberrant signaling through pathways controlling cell response to extracellular stimuli constitutes a central theme in disorders affecting development. Signaling through RAS and the MAPK cascade controls a variety of cell decisions in response to cytokines, hormones, and growth factors, and its upregulation causes Noonan syndrome (NS), a developmental disorder whose major features include a distinctive facies, a wide spectrum of cardiac defects, short stature, variable cognitive impairment, and predisposition to malignancies. NS is genetically heterogeneous, and mutations in more than ten genes have been reported to underlie this disorder. Despite the large number of genes implicated, about 10%-20% of affected individuals with a clinical diagnosis of NS do not have mutations in known RASopathy-associated genes, indicating that additional unidentified genes contribute to the disease, when mutated. By using a mixed strategy of functional candidacy and exome sequencing, we identify RRAS2 as a gene implicated in NS in six unrelated subjects/families. We show that the NS-causing RRAS2 variants affect highly conserved residues localized around the nucleotide binding pocket of the GTPase and are predicted to variably affect diverse aspects of RRAS2 biochemical behavior, including nucleotide binding, GTP hydrolysis, and interaction with effectors. Additionally, all pathogenic variants increase activation of the MAPK cascade and variably impact cell morphology and cytoskeletal rearrangement. Finally, we provide a characterization of the clinical phenotype associated with RRAS2 mutations.
Collapse
|
38
|
Tekendo-Ngongang C, Agenbag G, Bope CD, Esterhuizen AI, Wonkam A. Noonan Syndrome in South Africa: Clinical and Molecular Profiles. Front Genet 2019; 10:333. [PMID: 31057598 PMCID: PMC6477999 DOI: 10.3389/fgene.2019.00333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/28/2019] [Indexed: 11/13/2022] Open
Abstract
Noonan Syndrome (NS) is a common autosomal dominant multisystem disorder, caused by mutations in more than 10 genes in the Ras/MAPK signaling pathway. Differential mutation frequencies are observed across populations. Clinical expressions of NS are highly variable and include short stature, distinctive craniofacial dysmorphism, cardiovascular abnormalities, and developmental delay. Little is known about phenotypic specificities and molecular characteristics of NS in Africa. The present study has investigated patients with NS in Cape Town (South Africa). Clinical features were carefully documented in a total of 26 patients. Targeted Next-Generation Sequencing (NGS) was performed on 16 unrelated probands, using a multigene panel comprising 14 genes: PTPN11, SOS1, RIT1, A2ML1, BRAF, CBL, HRAS, KRAS, MAP2K1, MAP2K2, NRAS, RAF1, SHOC2, and SPRED1. The median age at diagnosis was 4.5 years (range: 1 month−51 years). Individuals of mixed-race ancestry were most represented (53.8%), followed by black Africans (30.8%). Our cohort revealed a lower frequency of pulmonary valve stenosis (34.6%) and a less severe developmental milestones phenotype. Molecular analysis found variants predicted to be pathogenic in 5 / 16 cases (31.2%). Among these mutations, two were previously reported: MAP2K1-c.389A>G (p.Tyr130Cys) and PTPN11 - c.1510A>G (p.Met504Val); three are novel: CBL-c.2520T>G (p.Cys840Trp), PTPN11- c.1496C>T (p.Ser499Phe), and MAP2K1- c.200A>C (p.Asp67Ala). Molecular dynamic simulations indicated that novel variants identified impact the stability and flexibility of their corresponding proteins. Genotype-phenotype correlations showed that clinical features of NS were more typical in patients with variants in MAP2K1. This first application of targeted NGS for the molecular diagnosis of NS in South Africans suggests that, while there is no major phenotypic difference compared to other populations, the distribution of genetic variants in NS in South Africans may be different.
Collapse
Affiliation(s)
- Cedrik Tekendo-Ngongang
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gloudi Agenbag
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Christian Domilongo Bope
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Departments of Mathematics and Computer Sciences, Faculty of Sciences, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Alina Izabela Esterhuizen
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Ambroise Wonkam
- Division of Human Genetics, Departments of Medicine and Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
39
|
Saik OV, Nimaev VV, Usmonov DB, Demenkov PS, Ivanisenko TV, Lavrik IN, Ivanisenko VA. Prioritization of genes involved in endothelial cell apoptosis by their implication in lymphedema using an analysis of associative gene networks with ANDSystem. BMC Med Genomics 2019; 12:47. [PMID: 30871556 PMCID: PMC6417156 DOI: 10.1186/s12920-019-0492-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Currently, more than 150 million people worldwide suffer from lymphedema. It is a chronic progressive disease characterized by high-protein edema of various parts of the body due to defects in lymphatic drainage. Molecular-genetic mechanisms of the disease are still poorly understood. Beginning of a clinical manifestation of primary lymphedema in middle age and the development of secondary lymphedema after treatment of breast cancer can be genetically determined. Disruption of endothelial cell apoptosis can be considered as one of the factors contributing to the development of lymphedema. However, a study of the relationship between genes associated with lymphedema and genes involved in endothelial apoptosis, in the associative gene network was not previously conducted. METHODS In the current work, we used well-known methods (ToppGene and Endeavour), as well as methods previously developed by us, to prioritize genes involved in endothelial apoptosis and to find potential participants of molecular-genetic mechanisms of lymphedema among them. Original methods of prioritization took into account the overrepresented Gene Ontology biological processes, the centrality of vertices in the associative gene network, describing the interactions of endothelial apoptosis genes with genes associated with lymphedema, and the association of the analyzed genes with diseases that are comorbid to lymphedema. RESULTS An assessment of the quality of prioritization was performed using criteria, which involved an analysis of the enrichment of the top-most priority genes by genes, which are known to have simultaneous interactions with lymphedema and endothelial cell apoptosis, as well as by genes differentially expressed in murine model of lymphedema. In particular, among genes involved in endothelial apoptosis, KDR, TNF, TEK, BMPR2, SERPINE1, IL10, CD40LG, CCL2, FASLG and ABL1 had the highest priority. The identified priority genes can be considered as candidates for genotyping in the studies involving the search for associations with lymphedema. CONCLUSIONS Analysis of interactions of these genes in the associative gene network of lymphedema can improve understanding of mechanisms of interaction between endothelial apoptosis and lymphangiogenesis, and shed light on the role of disturbance of these processes in the development of edema, chronic inflammation and connective tissue transformation during the progression of the disease.
Collapse
Affiliation(s)
- Olga V. Saik
- Laboratory of Computer-Assisted Proteomics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Prospekt Lavrentyeva 10, Novosibirsk, 630090 Russia
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
| | - Vadim V. Nimaev
- Laboratory of Surgical Lymphology and Lymphodetoxication, Research Institute of Clinical and Experimental Lymрhology – Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, st. Timakova 2, Novosibirsk, 630117 Russia
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
| | - Dilovarkhuja B. Usmonov
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
- Department of Neurosurgery, Ya. L. Tsivyan Novosibirsk Research Institute of Traumatology and Orthopedics, Ministry of Health of the Russian Federation, st. Frunze 17, Novosibirsk, 630091 Russia
| | - Pavel S. Demenkov
- Laboratory of Computer-Assisted Proteomics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Prospekt Lavrentyeva 10, Novosibirsk, 630090 Russia
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
| | - Timofey V. Ivanisenko
- Laboratory of Computer-Assisted Proteomics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Prospekt Lavrentyeva 10, Novosibirsk, 630090 Russia
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
| | - Inna N. Lavrik
- Laboratory of Computer-Assisted Proteomics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Prospekt Lavrentyeva 10, Novosibirsk, 630090 Russia
- Translational Inflammation Research, Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Medical Faculty, Pfalzer Platz 28, 39106 Magdeburg, Germany
| | - Vladimir A. Ivanisenko
- Laboratory of Computer-Assisted Proteomics, Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Prospekt Lavrentyeva 10, Novosibirsk, 630090 Russia
- Novosibirsk State University, st. Pirogova 1, Novosibirsk, 630090 Russia
| |
Collapse
|
40
|
Aljeaid D, Sanchez AI, Wakefield E, Chadwell SE, Moore N, Prada CE, Zhang W. Prevalence of pathogenic and likely pathogenic variants in the RASopathy genes in patients who have had panel testing for cardiomyopathy. Am J Med Genet A 2019; 179:608-614. [DOI: 10.1002/ajmg.a.61072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Deema Aljeaid
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- Department of Genetics Medicine, Faculty of Medicine; King Abdulaziz University; Jeddah Saudi Arabia
| | - Ana Isabel Sanchez
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- Instituto de Genética Humana; Pontificia Universidad Javeriana; Bogotá Colombia
| | - Emily Wakefield
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- RASopathies Program; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
| | - Sarah E. Chadwell
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- RASopathies Program; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
| | - Nicole Moore
- Heart Institute Diagnostic Laboratory; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
| | - Carlos E. Prada
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- RASopathies Program; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- Department of Pediatrics; University of Cincinnati College of Medicine; Cincinnati Ohio
- Division de Genetica, Departamento de Pediatría; Hospital Internacional de Colombia; Bucaramanga Colombia
| | - Wenying Zhang
- Division of Human Genetics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- Heart Institute Diagnostic Laboratory; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
- Department of Pediatrics; University of Cincinnati College of Medicine; Cincinnati Ohio
- Laboratory of Genetics and Genomics; Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
| |
Collapse
|
41
|
Bessis D, Miquel J, Bourrat E, Chiaverini C, Morice-Picard F, Abadie C, Manna F, Baumann C, Best M, Blanchet P, Bursztejn AC, Capri Y, Coubes C, Giuliano F, Guillaumont S, Hadj-Rabia S, Jacquemont ML, Jeandel C, Lacombe D, Mallet S, Mazereeuw-Hautier J, Molinari N, Pallure V, Pernet C, Philip N, Pinson L, Sarda P, Sigaudy S, Vial Y, Willems M, Geneviève D, Verloes A, Cavé H. Dermatological manifestations in Noonan syndrome: a prospective multicentric study of 129 patients positive for mutation. Br J Dermatol 2019; 180:1438-1448. [PMID: 30417923 DOI: 10.1111/bjd.17404] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Data on dermatological manifestations of Noonan syndrome (NS) remain heterogeneous and are based on limited dermatological expertise. OBJECTIVES To describe the dermatological manifestations of NS, compare them with the literature findings, and test for dermatological phenotype-genotype correlations with or without the presence of PTPN11 mutations. METHODS We performed a large 4-year, prospective, multicentric, collaborative dermatological and genetic study. RESULTS Overall, 129 patients with NS were enrolled, including 65 patients with PTPN11-NS, 34 patients with PTPN11-NS with multiple lentigines (NSML), and 30 patients with NS who had a mutation other than PTPN11. Easy bruising was the most frequent dermatological finding in PTPN11-NS, present in 53·8% of patients. Multiple lentigines and café-au-lait macules (n ≥ 3) were present in 94% and 80% of cases of NSML linked to specific mutations of PTPN11, respectively. Atypical forms of NSML could be associated with NS with RAF1 or NRAS mutations. In univariate analysis, patients without a PTPN11 mutation showed (i) a significantly higher frequency of keratinization disorders (P = 0·001), including keratosis pilaris (P = 0·005), ulerythema ophryogenes (P = 0·0001) and palmar and/or plantar hyperkeratosis (P = 0·06, trend association), and (ii) a significantly higher frequency of scarce scalp hair (P = 0·035) and scarce or absent eyelashes (P = 0·06, trend association) than those with PTPN11 mutations. CONCLUSIONS The cutaneous phenotype of NS with a PTPN11 mutation is generally mild and nonspecific, whereas the absence of a PTPN11 mutation is associated with a high frequency of keratinization disorders and hair abnormalities.
Collapse
Affiliation(s)
- D Bessis
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France.,University of Montpellier, Montpellier, France.,INSERM U1058, Montpellier, France
| | - J Miquel
- Department of Paediatric Dermatology, Femme-Mère-Enfant Hospital, University of South Réunion, Saint-Pierre Réunion, France.,Department of Dermatology, University of Rennes, Rennes, France
| | - E Bourrat
- Department of Paediatric Dermatology, Robert-Debré Hospital, AP-HP, Paris, France
| | - C Chiaverini
- Department of Dermatology, L'Archet 2 Hospital, Nice, France.,University of Nice, Nice, France
| | - F Morice-Picard
- Department of Paediatric Dermatology, Pellegrin University Hospital of Bordeaux, Bordeaux, France
| | - C Abadie
- Department of Clinical Genetics, Sud Hospital, Rennes, France.,University Hospital of Rennes, Rennes, France
| | - F Manna
- University of Montpellier, Montpellier, France.,Department of Medical Information, Epidemiological and Clinical Research Unit, La Colombière Hospital, Montpellier, France
| | - C Baumann
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - M Best
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France.,University of Montpellier, Montpellier, France
| | - P Blanchet
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - A-C Bursztejn
- Department of Dermatology, Brabois Hospital, Nancy, France.,University of Nancy, Nancy, France
| | - Y Capri
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - C Coubes
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - F Giuliano
- University of Nice, Nice, France.,Department of Clinical Genetics, L'Archet 2 Hospital, Nice, France
| | - S Guillaumont
- University of Montpellier, Montpellier, France.,Department of Paediatric Cardiology, Arnaud de Villeneuve Hospital, Montpellier, France
| | - S Hadj-Rabia
- Department of Paediatric Dermatology, Reference Centre for Rare Skin Diseases, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - M-L Jacquemont
- Department of Clinical Genetics, Femme-Mère-Enfant Hospital, University of South Réunion, Saint-Pierre Réunion, France
| | - C Jeandel
- University of Montpellier, Montpellier, France.,Department of Paediatric Endocrinology, Arnaud de Villeneuve Hospital, Montpellier, France
| | - D Lacombe
- Department of Clinical Genetics, Pellegrin University Hospital of Bordeaux, AP-HP, Paris, France
| | - S Mallet
- Department of Dermatology, La Timone Hospital, AP-HM, Marseille, France.,University of Marseille, Marseille, France
| | - J Mazereeuw-Hautier
- Department of Dermatology, Larrey Hospital, Reference Centre for Rare Skin Diseases, Toulouse, France.,University of Toulouse, Toulouse, France
| | - N Molinari
- University of Montpellier, Montpellier, France.,Department of Medical Information, Epidemiological and Clinical Research Unit, La Colombière Hospital, Montpellier, France
| | - V Pallure
- Department of Dermatology, CH, Perpignan, Perpignan, France
| | - C Pernet
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France
| | - N Philip
- University of Marseille, Marseille, France.,Department of Clinical Genetics, La Timone Hospital, AP-HM, Marseille, France
| | - L Pinson
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - P Sarda
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - S Sigaudy
- University of Marseille, Marseille, France.,Department of Clinical Genetics, La Timone Hospital, AP-HM, Marseille, France
| | - Y Vial
- University of Paris-Diderot, Paris, France.,Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP, Paris, France
| | - M Willems
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - D Geneviève
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France.,INSERM U1183, Montpellier, France
| | - A Verloes
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - H Cavé
- University of Paris-Diderot, Paris, France.,Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP, Paris, France
| |
Collapse
|
42
|
Koromani F, Trajanoska K, Rivadeneira F, Oei L. Recent Advances in the Genetics of Fractures in Osteoporosis. Front Endocrinol (Lausanne) 2019; 10:337. [PMID: 31231309 PMCID: PMC6559287 DOI: 10.3389/fendo.2019.00337] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/10/2019] [Indexed: 12/15/2022] Open
Abstract
Genetic susceptibility, together with old age, female sex, and low bone mineral density (BMD) are amongst the strongest determinants of fracture risk. Tmost recent large-scale genome-wide association study (GWAS) meta-analysis has yielded fifteen loci. This review focuses on the advances in the research of genetic determinants of fracture risk. We first discuss the genetic architecture of fracture risk, touching upon different methods and overall findings. We then discuss in a second paragraph the most recent advances in the field and focus on the genetics of fracture risk and also of other endophenotypes closely related to fracture risk such as bone mineral density (BMD). Application of state-of-the-art methodology such as Mendelian randzation in fracture GWAS are reviewed. The final part of this review touches upon potential future directions in genetic research of osteoporotic fractures.
Collapse
Affiliation(s)
- Fjorda Koromani
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Ling Oei
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Ling Oei
| |
Collapse
|
43
|
Bustelo XR, Crespo P, Fernández-Pisonero I, Rodríguez-Fdez S. RAS GTPase-dependent pathways in developmental diseases: old guys, new lads, and current challenges. Curr Opin Cell Biol 2018; 55:42-51. [PMID: 30007125 PMCID: PMC7615762 DOI: 10.1016/j.ceb.2018.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/14/2018] [Accepted: 06/14/2018] [Indexed: 10/28/2022]
Abstract
Deregulated RAS signaling is associated with increasing numbers of congenital diseases usually referred to as RASopathies. The spectrum of genes and mutant alleles causing these diseases has been significantly expanded in recent years. This progress has triggered new challenges, including the origin and subsequent selection of the mutations driving these diseases, the specific pathobiological programs triggered by those mutations, the type of correlations that exist between the genotype and the clinical features of patients, and the ancillary genetic factors that influence the severity of the disease in patients. These issues also directly impinge on the feasibility of using RAS pathway drugs to treat RASopathy patients. Here, we will review the main developments and pending challenges in this research topic.
Collapse
Affiliation(s)
- Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain.
| | - Piero Crespo
- CIBERONC, CSIC-University of Cantabria, 39011 Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-University of Cantabria, 39011 Santander, Spain
| | - Isabel Fernández-Pisonero
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW To provide information on the scope of cardiac disease in Noonan syndrome. RECENT FINDINGS Noonan syndrome is a common autosomal dominant RASopathy disorder characterized by clinical findings of facial dysmorphism, congenital heart disease, and short stature. The degree of genetic heterogeneity has recently become evident in that Noonan syndrome is now known to be caused by mutations in a large variety of genes which produce dysregulation of the RAS-MAPK (mitogen-activated protein kinase) signaling pathway. The scope of cardiac disease in Noonan syndrome is quite variable depending on the gene mutation, with some mutations usually associated with a high incidence of congenital heart defects (PTPN11, KRAS, and others) while those with predominantly hypertrophic cardiomyopathy (HCM) have higher risk and morbidity profiles (RAF1, RIT1, and those associated with multiple lentigines). SUMMARY Cardiac disease in Noonan syndrome varies according to the type of gene mutation. The most common forms of cardiac disease include pulmonary stenosis, HCM, and atrial septal defect. HCM in general is associated with increased risk, mortality, and morbidity. New concepts for potential treatments are discussed.
Collapse
|
45
|
Tajan M, Paccoud R, Branka S, Edouard T, Yart A. The RASopathy Family: Consequences of Germline Activation of the RAS/MAPK Pathway. Endocr Rev 2018; 39:676-700. [PMID: 29924299 DOI: 10.1210/er.2017-00232] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Noonan syndrome [NS; Mendelian Inheritance in Men (MIM) #163950] and related syndromes [Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome; MIM #151100), Noonan-like syndrome with loose anagen hair (MIM #607721), Costello syndrome (MIM #218040), cardio-facio-cutaneous syndrome (MIM #115150), type I neurofibromatosis (MIM #162200), and Legius syndrome (MIM #611431)] are a group of related genetic disorders associated with distinctive facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was clinically described more than 50 years ago, and disease genes have been identified throughout the last 3 decades, providing a molecular basis to better understand their physiopathology and identify targets for therapeutic strategies. Most of these genes encode proteins belonging to or regulating the so-called RAS/MAPK signaling pathway, so these syndromes have been gathered under the name RASopathies. In this review, we provide a clinical overview of RASopathies and an update on their genetics. We then focus on the functional and pathophysiological effects of RASopathy-causing mutations and discuss therapeutic perspectives and future directions.
Collapse
Affiliation(s)
- Mylène Tajan
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Romain Paccoud
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Sophie Branka
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Armelle Yart
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| |
Collapse
|
46
|
Bessis D, Morice-Picard F, Bourrat E, Abadie C, Aouinti S, Baumann C, Best M, Bursztejn AC, Capri Y, Chiaverini C, Coubes C, Giuliano F, Hadj-Rabia S, Jacquemont ML, Lacombe D, Lyonnet S, Mallet S, Mazereeuw-Hautier J, Miquel J, Molinari N, Parfait B, Pernet C, Philip N, Pinson L, Pouvreau N, Vial Y, Sarda P, Sigaudy S, Verloes A, Cavé H, Geneviève D. Dermatological manifestations in cardiofaciocutaneous syndrome: a prospective multicentric study of 45 mutation-positive patients. Br J Dermatol 2018; 180:172-180. [PMID: 30141192 DOI: 10.1111/bjd.17077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Data on dermatological manifestations of cardiofaciocutaneous syndrome (CFCS) remain heterogeneous and almost without expert dermatological classification. OBJECTIVES To describe the dermatological manifestations of CFCS; to compare them with the literature findings; to assess those discriminating CFCS from other RASopathies, including Noonan syndrome (NS) and Costello syndrome (CS); and to test for dermatological phenotype-genotype correlations. METHODS We performed a 4-year, large, prospective, multicentric, collaborative dermatological and genetic study. RESULTS Forty-five patients were enrolled. Hair abnormalities were ubiquitous, including scarcity or absence of eyebrows and wavy or curly hair in 73% and 69% of patients, respectively. Keratosis pilaris (KP), ulerythema ophryogenes (UO), palmoplantar hyperkeratosis (PPHK) and multiple melanocytic naevi (MMN; over 50 naevi) were noted in 82%, 44%, 27% and 29% of patients, respectively. Scarcity or absence of eyebrows, association of UO and PPHK, diffuse KP and MMN best differentiated CFCS from NS and CS. Oral acitretin may be highly beneficial for therapeutic management of PPHK, whereas treatment of UO by topical sirolimus 1% failed. No significant dermatological phenotype-genotype correlation was determined. CONCLUSIONS A thorough knowledge of CFCS skin manifestations would help in making a positive diagnosis and differentiating CFCS from CS and NS.
Collapse
Affiliation(s)
- D Bessis
- Department of Dermatology, Saint-Eloi Hospital, University of Montpellier, Montpellier, France.,INSERM U1058, Montpellier, France
| | - F Morice-Picard
- Department of Pediatric Dermatology, Pellegrin University Hospital of Bordeaux, Bordeaux, AP-HP, France
| | - E Bourrat
- Department of Pediatric Dermatology, Robert-Debré Hospital, AP-HP, Paris, France
| | - C Abadie
- Department of Clinical Genetics, Sud Hospital and University Hospital of Rennes, Rennes, France
| | - S Aouinti
- Department of Statistics, La Colombière Hospital and University of Montpellier, Montpellier, France
| | - C Baumann
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - M Best
- Department of Dermatology, Saint-Eloi Hospital, University of Montpellier, Montpellier, France
| | - A-C Bursztejn
- Department of Dermatology, Brabois Hospital, University of Nancy, Nancy, France
| | - Y Capri
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - C Chiaverini
- Department of Dermatology, L'Archet 2 Hospital and University of Nice, Nice, France
| | - C Coubes
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, University of Montpellier, Montpellier, France
| | - F Giuliano
- Department of Clinical Genetics, L'Archet 2 Hospital and University of Nice, Nice, France
| | - S Hadj-Rabia
- Department of Pediatric Dermatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - M-L Jacquemont
- Department of Clinical Genetics, Femme-Mère-Enfant Hospital, University of South Reunion, Saint-Pierre, Réunion, France
| | - D Lacombe
- Department of Clinical Genetics, Pellegrin University Hospital of Bordeaux, Bordeaux, AP-HP, France
| | - S Lyonnet
- Department of Clinical Genetics, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - S Mallet
- Department of Dermatology, La Timone Hospital, AP-HM and University of Marseille, Marseille, France
| | - J Mazereeuw-Hautier
- Department of Dermatology, Larrey Hospital, Reference Center for Rare Skin Diseases, University of Toulouse, Toulouse, France
| | - J Miquel
- Department of Pediatric Dermatology, Femme-Mère-Enfant Hospital, University of South Reunion, Saint-Pierre, Réunion, France.,Department of Dermatology, University of Rennes, Rennes, France
| | - N Molinari
- Department of Statistics, La Colombière Hospital and University of Montpellier, Montpellier, France
| | - B Parfait
- Department of Molecular Genetics and Biology, Cochin Hospital, AP-HP, University Paris V, Paris, France
| | - C Pernet
- Department of Dermatology, Saint-Eloi Hospital, University of Montpellier, Montpellier, France
| | - N Philip
- Department of Clinical Genetics, La Timone Hospital, AP-HM and University of Marseille, Marseille, France
| | - L Pinson
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, University of Montpellier, Montpellier, France
| | - N Pouvreau
- Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - Y Vial
- Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - P Sarda
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, University of Montpellier, Montpellier, France
| | - S Sigaudy
- Department of Clinical Genetics, La Timone Hospital, AP-HM and University of Marseille, Marseille, France
| | - A Verloes
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - H Cavé
- Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP and University of Paris-Diderot, Paris, France
| | - D Geneviève
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, University of Montpellier, Montpellier, France.,INSERM U1183, Montpellier, France
| |
Collapse
|
47
|
Couser NL, Keelean-Fuller D, Davenport ML, Haverfield E, Masood MM, Henin M, Aylsworth AS. Cleft palate and hypopituitarism in a patient with Noonan-like syndrome with loose anagen hair-1. Am J Med Genet A 2018; 176:2024-2027. [PMID: 30240112 DOI: 10.1002/ajmg.a.40432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/04/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022]
Abstract
Noonan syndrome (NS), the most common of the RASopathies, is a developmental disorder caused by heterozygous germline mutations in genes encoding proteins in the RAS-MAPK signaling pathway. Noonan-like syndrome with loose anagen hair (NSLH, including NSLH1, OMIM #607721 and NSLH2, OMIM #617506) is characterized by typical features of NS with additional findings of macrocephaly, loose anagen hair, growth hormone deficiency in some, and a higher incidence of intellectual disability. All NSLH1 reported cases to date have had an SHOC2 c.4A>G, p.Ser2Gly mutation; NSLH2 cases have been reported with a PPP1CB c.146G>C, p.Pro49Arg mutation, or c.166G>C, p.Ala56Pro mutation. True cleft palate does not appear to have been previously reported in individuals with NS or with NSLH. While some patients with NS have had growth hormone deficiency (GHD), other endocrine abnormalities are only rarely documented. We present a female patient with NSLH1 who was born with a posterior cleft palate, micrognathia, and mild hypotonia. Other findings in her childhood and young adulthood years include hearing loss, strabismus, and hypopituitarism with growth hormone, thyroid stimulating hormone (TSH), and gonadotropin deficiencies. The SHOC2 mutation may be responsible for this patient's additional features of cleft palate and hypopituitarism.
Collapse
Affiliation(s)
- Natario L Couser
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Department of Pediatrics, Division of Genetics and Metabolism, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Department of Ophthalmology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Debra Keelean-Fuller
- Department of Pediatrics, Division of Genetics and Metabolism, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Marsha L Davenport
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | | | - Maheer M Masood
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Mark Henin
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Arthur S Aylsworth
- Department of Pediatrics, Division of Genetics and Metabolism, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
48
|
Differential Role of the RasGEFs Sos1 and Sos2 in Mouse Skin Homeostasis and Carcinogenesis. Mol Cell Biol 2018; 38:MCB.00049-18. [PMID: 29844066 DOI: 10.1128/mcb.00049-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/21/2018] [Indexed: 12/24/2022] Open
Abstract
Using Sos1 knockout (Sos1-KO), Sos2-KO, and Sos1/2 double-knockout (Sos1/2-DKO) mice, we assessed the functional role of Sos1 and Sos2 in skin homeostasis under physiological and/or pathological conditions. Sos1 depletion resulted in significant alterations of skin homeostasis, including reduced keratinocyte proliferation, altered hair follicle and blood vessel integrity in dermis, and reduced adipose tissue in hypodermis. These defects worsened significantly when both Sos1 and Sos2 were absent. Simultaneous Sos1/2 disruption led to severe impairment of the ability to repair skin wounds, as well as to almost complete ablation of the neutrophil-mediated inflammatory response in the injury site. Furthermore, Sos1 disruption delayed the onset of tumor initiation, decreased tumor growth, and prevented malignant progression of papillomas in a DMBA (7,12-dimethylbenz[α]anthracene)/TPA (12-O-tetradecanoylphorbol-13-acetate)-induced skin carcinogenesis model. Finally, Sos1 depletion in preexisting chemically induced papillomas resulted also in decreased tumor growth, probably linked to significantly reduced underlying keratinocyte proliferation. Our data unveil novel, distinctive mechanistic roles of Sos 1 and Sos2 in physiological control of skin homeostasis and wound repair, as well as in pathological development of chemically induced skin tumors. These observations underscore the essential role of Sos proteins in cellular proliferation and migration and support the consideration of these RasGEFs as potential biomarkers/therapy targets in Ras-driven epidermal tumors.
Collapse
|
49
|
Tamura A, Uemura S, Matsubara K, Kozuki E, Tanaka T, Nino N, Yokoi T, Saito A, Ishida T, Hasegawa D, Umeki I, Niihori T, Nakazawa Y, Koike K, Aoki Y, Kosaka Y. Co-occurrence of hypertrophic cardiomyopathy and juvenile myelomonocytic leukemia in a neonate with Noonan syndrome, leading to premature death. Clin Case Rep 2018; 6:1202-1207. [PMID: 29988639 PMCID: PMC6028379 DOI: 10.1002/ccr3.1568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/04/2018] [Accepted: 04/12/2018] [Indexed: 11/09/2022] Open
Abstract
We report a case of a neonate with Noonan syndrome presenting with concurrent hypertrophic cardiomyopathy and juvenile myelomonocytic leukemia, which resulted in premature death. Cases with Noonan syndrome diagnosed during the neonatal period might not necessarily show mild clinical course, and premature death is a possible outcome to be considered.
Collapse
Affiliation(s)
- Akihiro Tamura
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
| | - Suguru Uemura
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
- Department of PediatricsKobe University School of MedicineKobeJapan
| | - Kousaku Matsubara
- Department of PediatricsKobe City Nishi‐Kobe Medical CenterKobeJapan
| | - Eru Kozuki
- Department of PediatricsKobe City Nishi‐Kobe Medical CenterKobeJapan
| | | | - Nanako Nino
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
- Department of PediatricsKobe University School of MedicineKobeJapan
| | - Takehito Yokoi
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
- Department of PediatricsOsaka University HospitalSuitaJapan
| | - Atsuro Saito
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
| | - Toshiaki Ishida
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
| | | | - Ikumi Umeki
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Tetsuya Niihori
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Yozo Nakazawa
- Department of PediatricsShinshu University School of MedicineMatsumotoJapan
| | - Kenichi Koike
- Department of PediatricsShinonoi General HospitalMinami Nagano Medical CenterNaganoJapan
| | - Yoko Aoki
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Yoshiyuki Kosaka
- Department of Hematology and OncologyKobe Children's HospitalKobeJapan
| |
Collapse
|
50
|
Comparative assessment of gene-specific variant distribution in prenatal and postnatal cohorts tested for Noonan syndrome and related conditions. Genet Med 2018; 21:417-425. [DOI: 10.1038/s41436-018-0062-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/02/2018] [Indexed: 11/08/2022] Open
|