1
|
Deng H, Zhang Q, Yi J, Yuan L. Unraveling ptosis: a comprehensive review of clinical manifestations, genetics, and treatment. Prog Retin Eye Res 2024:101327. [PMID: 39725023 DOI: 10.1016/j.preteyeres.2024.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Ptosis is defined as an abnormally low-lying upper eyelid margin on the primary gaze, generally resulting from a congenital or acquired abnormality of the nerves or muscles that control the eyelid. Ptosis can occur alone or concurrently as an ocular or systemic syndrome, and the prevalence of ptosis varies among different countries and populations. Isolated ptosis typically causes aesthetic problems in patients and can lead to functional ophthalmic problems in severe cases. In individuals with syndromic ptosis, ptosis can be a warning of serious medical problems. There are different approaches to classification, depending on the onset time or the etiology of ptosis, and the clinical characteristics of congenital and acquired ptosis also differ. Pedigree and genetic analysis have demonstrated that hereditary ptosis is clinically heterogeneous, with incomplete concordance and variable expressivity. A number of genetic loci and genes responsible for hereditary isolated and syndromic ptosis have been reported. Optimal surgical timing and proper method are truly critical for avoiding the risk of potentially severe outcomes from ptosis and minimizing surgical complications, which are challenging as the pathogenesis is still indistinct and the anatomy is complex. This review provides a comprehensive review of ptosis, by summarizing the clinical manifestations, classification, diagnosis, genetics, treatment, and prognosis, as well as the bound anatomy of upper eyelid.
Collapse
Affiliation(s)
- Hao Deng
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Research Center of Medical Experimental Technology, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Disease Genome Research Center, Central South University, Changsha 410013, China; Department of Laboratory Medicine, the Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Qianling Zhang
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Department of Laboratory Medicine, the Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Junhui Yi
- Department of Ophthalmology, the Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Lamei Yuan
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Research Center of Medical Experimental Technology, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha 410013, China; Disease Genome Research Center, Central South University, Changsha 410013, China.
| |
Collapse
|
2
|
Barbero AIS, Valenzuela I, Fernández-Alvarez P, Vazquez É, Cueto-Gonzalez AM, Lasa-Aranzasti A, Trujillano L, Masotto B, Arumí EG, Tizzano EF. New Insights Into the Spectrum of RASopathies: Clinical and Genetic Data in a Cohort of 121 Spanish Patients. Am J Med Genet A 2024:e63905. [PMID: 39484914 DOI: 10.1002/ajmg.a.63905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 11/03/2024]
Abstract
Noonan syndrome and related disorders are a group of well-known genetic conditions caused by dysregulation of the Ras/mitogen-activated protein kinase (RAS/MAPK) pathway. Because of the overlap of clinical and molecular features, they are now called RASopathies. In this study, we retrospectively analyzed the clinical data of 121 patients with a molecularly confirmed diagnosis of RASopathy, describing frequencies for clinical features in all organ systems as well as molecular data. The most common clinical diagnosis was Noonan Syndrome and the most frequently affected gene was PTPN11 followed by SOS1, RAF1, LZTR1, and RIT1. All patients had distinctive craniofacial features indicative of the RASopathy spectrum but we report some atypical features regarding craniofacial shape, such as craniosynostosis and microcephaly. We also describe uncommon clinical characteristics such as aortic dilation, multivalvular heart disease, abnormalities of the posterior fossa, and uterine congenital anomalies in female patients. Furthermore, the presence of multiple giant cell granulomas was observed specifically in patients with SOS1 variants. This comprehensive evaluation allows broadening the phenotypic spectrum of our population and their correlation with the genotype, which are essential to improve the recognition and the follow up of RASopathies as a multisystemic disease.
Collapse
Affiliation(s)
- Ana Isabel Sánchez Barbero
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Paula Fernández-Alvarez
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Élida Vazquez
- Department of Pediatric Radiology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Anna Maria Cueto-Gonzalez
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Amaia Lasa-Aranzasti
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Laura Trujillano
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Bárbara Masotto
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Elena García Arumí
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| | - Eduardo F Tizzano
- Department of Clinical and Molecular Genetics and Rare Disease Unit, Vall Hebron Research Institute, Barcelona, Spain
- Medicine Genetics Group, Vall Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Chatzi D, Kyriakoudi SA, Dermitzakis I, Manthou ME, Meditskou S, Theotokis P. Clinical and Genetic Correlation in Neurocristopathies: Bridging a Precision Medicine Gap. J Clin Med 2024; 13:2223. [PMID: 38673496 PMCID: PMC11050951 DOI: 10.3390/jcm13082223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Neurocristopathies (NCPs) encompass a spectrum of disorders arising from issues during the formation and migration of neural crest cells (NCCs). NCCs undergo epithelial-mesenchymal transition (EMT) and upon key developmental gene deregulation, fetuses and neonates are prone to exhibit diverse manifestations depending on the affected area. These conditions are generally rare and often have a genetic basis, with many following Mendelian inheritance patterns, thus making them perfect candidates for precision medicine. Examples include cranial NCPs, like Goldenhar syndrome and Axenfeld-Rieger syndrome; cardiac-vagal NCPs, such as DiGeorge syndrome; truncal NCPs, like congenital central hypoventilation syndrome and Waardenburg syndrome; and enteric NCPs, such as Hirschsprung disease. Additionally, NCCs' migratory and differentiating nature makes their derivatives prone to tumors, with various cancer types categorized based on their NCC origin. Representative examples include schwannomas and pheochromocytomas. This review summarizes current knowledge of diseases arising from defects in NCCs' specification and highlights the potential of precision medicine to remedy a clinical phenotype by targeting the genotype, particularly important given that those affected are primarily infants and young children.
Collapse
Affiliation(s)
| | | | | | | | | | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.C.); (S.A.K.); (I.D.); (M.E.M.); (S.M.)
| |
Collapse
|
7
|
Shoji Y, Hata A, Maeyama T, Wada T, Hasegawa Y, Nishi E, Ida S, Etani Y, Niihori T, Aoki Y, Okamoto N, Kawai M. Genetic backgrounds and genotype-phenotype relationships in anthropometric parameters of 116 Japanese individuals with Noonan syndrome. Clin Pediatr Endocrinol 2024; 33:50-58. [PMID: 38572385 PMCID: PMC10985011 DOI: 10.1297/cpe.2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/02/2024] [Indexed: 04/05/2024] Open
Abstract
Noonan syndrome (NS) is caused by pathogenic variants in genes encoding components of the RAS/MAPK pathway and presents with a number of symptoms, including characteristic facial features, congenital heart diseases, and short stature. Advances in genetic analyses have contributed to the identification of pathogenic genes in NS as well as genotype-phenotype relationships; however, updated evidence for the detection rate of pathogenic genes with the inclusion of newly identified genes is lacking in Japan. Accordingly, we examined the genetic background of 116 individuals clinically diagnosed with NS and the frequency of short stature. We also investigated genotype-phenotype relationships in the context of body mass index (BMI). Genetic testing revealed the responsible variants in 100 individuals (86%), where PTPN11 variants were the most prevalent (43%) and followed by SOS1 (12%) and RIT1 (9%). The frequency of short stature was the lowest in subjects possessing RIT1 variants. No genotype-phenotype relationships in BMI were observed among the genotypes. In conclusion, this study provides evidence for the detection rate of pathogenic genes and genotype-phenotype relationships in Japanese patients with NS, which will be of clinical importance for accelerating our understanding of the genetic backgrounds of Japanese patients with NS.
Collapse
Affiliation(s)
- Yasuko Shoji
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
- Department of Epidemiology and Health Policy, University of Toyama, Toyama, Japan
| | - Ayaha Hata
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Takatoshi Maeyama
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Tamaki Wada
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Yuiko Hasegawa
- Department of Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Eriko Nishi
- Department of Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Shinobu Ida
- Department of Clinical Laboratory, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Yuri Etani
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University School of Medicine, Miyagi, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, Miyagi, Japan
| | - Nobuhiko Okamoto
- Department of Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Masanobu Kawai
- Department of Gastroenterology and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka, Japan
| |
Collapse
|
8
|
Cherra SJ, Lamb R. Interactions between Ras and Rap signaling pathways during neurodevelopment in health and disease. Front Mol Neurosci 2024; 17:1352731. [PMID: 38463630 PMCID: PMC10920261 DOI: 10.3389/fnmol.2024.1352731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
The Ras family of small GTPases coordinates tissue development by modulating cell proliferation, cell-cell adhesion, and cellular morphology. Perturbations of any of these key steps alter nervous system development and are associated with neurological disorders. While the underlying causes are not known, genetic mutations in Ras and Rap GTPase signaling pathways have been identified in numerous neurodevelopmental disorders, including autism spectrum, neurofibromatosis, intellectual disability, epilepsy, and schizophrenia. Despite diverse clinical presentations, intersections between these two signaling pathways may provide a better understanding of how deviations in neurodevelopment give rise to neurological disorders. In this review, we focus on presynaptic and postsynaptic functions of Ras and Rap GTPases. We highlight various roles of these small GTPases during synapse formation and plasticity. Based on genomic analyses, we discuss how disease-related mutations in Ras and Rap signaling proteins may underlie human disorders. Finally, we discuss how recent observations have identified molecular interactions between these pathways and how these findings may provide insights into the mechanisms that underlie neurodevelopmental disorders.
Collapse
Affiliation(s)
- Salvatore J. Cherra
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | | |
Collapse
|
9
|
Costa ISD, Junot T, Silva FL, Felix W, Cardozo Fh JL, Pereira de Araujo AF, Pais do Amaral C, Gonçalves S, Santos NC, Leite JRSA, Bloch C, Brand GD. Occurrence and evolutionary conservation analysis of α-helical cationic amphiphilic segments in the human proteome. FEBS J 2024; 291:547-565. [PMID: 37945538 DOI: 10.1111/febs.16997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/14/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The existence of encrypted fragments with antimicrobial activity in human proteins has been thoroughly demonstrated in the literature. Recently, algorithms for the large-scale identification of these segments in whole proteomes were developed, and the pervasiveness of this phenomenon was stated. These algorithms typically mine encrypted cationic and amphiphilic segments of proteins, which, when synthesized as individual polypeptide sequences, exert antimicrobial activity by membrane disruption. In the present report, the human reference proteome was submitted to the software kamal for the uncovering of protein segments that correspond to putative intragenic antimicrobial peptides (IAPs). The assessment of the identity of these segments, frequency, functional classes of parent proteins, structural relevance, and evolutionary conservation of amino acid residues within their corresponding proteins was conducted in silico. Additionally, the antimicrobial and anticancer activity of six selected synthetic peptides was evaluated. Our results indicate that cationic and amphiphilic segments can be found in 2% of all human proteins, but are more common in transmembrane and peripheral membrane proteins. These segments are surface-exposed basic patches whose amino acid residues present similar conservation scores to other residues with similar solvent accessibility. Moreover, the antimicrobial and anticancer activity of the synthetic putative IAP sequences was irrespective to whether these are associated to membranes in the cellular setting. Our study discusses these findings in light of the current understanding of encrypted peptide sequences, offering some insights into the relevance of these segments to the organism in the context of their harboring proteins or as separate polypeptide sequences.
Collapse
Affiliation(s)
- Igor S D Costa
- Laboratório de Síntese e Análise de Biomoléculas - LSAB, Instituto de Química, Universidade de Brasília, Brazil
| | - Tiago Junot
- Laboratório de Síntese e Análise de Biomoléculas - LSAB, Instituto de Química, Universidade de Brasília, Brazil
| | - Fernanda L Silva
- Laboratório de Síntese e Análise de Biomoléculas - LSAB, Instituto de Química, Universidade de Brasília, Brazil
| | - Wanessa Felix
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada - NuPMIA, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - José L Cardozo Fh
- Laboratório de Espectrometria de Massa - LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Brazil
| | - Antonio F Pereira de Araujo
- Laboratório de Biofísica Teórica e Computacional, Departamento de Biologia Celular, Universidade de Brasília, Brazil
| | | | - Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - José R S A Leite
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada - NuPMIA, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - Carlos Bloch
- Laboratório de Espectrometria de Massa - LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Brazil
| | - Guilherme D Brand
- Laboratório de Síntese e Análise de Biomoléculas - LSAB, Instituto de Química, Universidade de Brasília, Brazil
| |
Collapse
|
10
|
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
|
11
|
Zheng J, Peng L, Cheng R, Li Z, Xie J, Huang E, Cheng J, Zhao Q. RAF1 mutation leading to hypertrophic cardiomyopathy in a Chinese family with a history of sudden cardiac death: A diagnostic insight into Noonan syndrome. Mol Genet Genomic Med 2024; 12:e2290. [PMID: 37787490 PMCID: PMC10767430 DOI: 10.1002/mgg3.2290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 09/10/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is predominantly caused by mutations in sarcomeric genes. However, a subset of cases is attributed to genetic disorders unrelated to sarcomeric genes, such as Noonan syndrome (NS) and other RASopathies. In this study, we present a family with a history of sudden cardiac death (SCD) and focus on two adults with syndromic left ventricular hypertrophy (LVH). METHODS Clinical evaluations, including echocardiography, were conducted to assess cardiac manifestations. Whole-exome sequencing was performed to identify potential genetic variants underlying syndromic LVH in the study participants. RESULTS Whole-exome sequencing revealed a missense variant in the RAF1 gene, c.782C>T (p.Pro261Leu). This variant confirmed the diagnosis of NS in the affected individuals. CONCLUSION The findings of this study underscore the importance of family history investigation and genetic testing in diagnosing syndromic LVH. By identifying the underlying genetic cause, clinicians can better understand the etiology of RAS-HCM and its association with SCD in young adults.
Collapse
Affiliation(s)
- Jingjing Zheng
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Longyun Peng
- Department of CardiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Ruofei Cheng
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Zhiyan Li
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Jianjie Xie
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Erwen Huang
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Jianding Cheng
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Qianhao Zhao
- Faculty of Forensic Medicine, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
- Guangdong Province Translational Forensic Medicine Engineering Technology Research CenterSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
12
|
Chen T, Tang X, Wang Z, Feng F, Xu C, Zhao Q, Wu Y, Sun H, Chen Y. Inhibition of Son of Sevenless Homologue 1 (SOS1): Promising therapeutic treatment for KRAS-mutant cancers. Eur J Med Chem 2023; 261:115828. [PMID: 37778239 DOI: 10.1016/j.ejmech.2023.115828] [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: 07/04/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
Kristen rat sarcoma (KRAS) is one of the most common oncogenes in human cancers. As a guanine nucleotide exchange factor, Son of Sevenless Homologue 1 (SOS1) represents a potential therapeutic concept for the treatment of KRAS-mutant cancers because of its activation on KRAS and downstream signaling pathways. In this review, we provide a comprehensive overview of the structure, biological function, and regulation of SOS1. We also focus on the recent advances in SOS1 inhibitors and emphasize their binding modes, structure-activity relationships and pharmacological activities. We hope that this publication can provide a comprehensive compendium on the rational design of SOS1 inhibitors.
Collapse
Affiliation(s)
- Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zhenqi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China
| | - Chunlei Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qun Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yulan Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
13
|
Yi JS, Perla S, Bennett AM. An Assessment of the Therapeutic Landscape for the Treatment of Heart Disease in the RASopathies. Cardiovasc Drugs Ther 2023; 37:1193-1204. [PMID: 35156148 DOI: 10.1007/s10557-022-07324-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
The RAS/mitogen-activated protein kinase (MAPK) pathway controls a plethora of developmental and post-developmental processes. It is now clear that mutations in the RAS-MAPK pathway cause developmental diseases collectively referred to as the RASopathies. The RASopathies include Noonan syndrome, Noonan syndrome with multiple lentigines, cardiofaciocutaneous syndrome, neurofibromatosis type 1, and Costello syndrome. RASopathy patients exhibit a wide spectrum of congenital heart defects (CHD), such as valvular abnormalities and hypertrophic cardiomyopathy (HCM). Since the cardiovascular defects are the most serious and recurrent cause of mortality in RASopathy patients, it is critical to understand the pathological signaling mechanisms that drive the disease. Therapies for the treatment of HCM and other RASopathy-associated comorbidities have yet to be fully realized. Recent developments have shown promise for the use of repurposed antineoplastic drugs that target the RAS-MAPK pathway for the treatment of RASopathy-associated HCM. However, given the impact of the RAS-MAPK pathway in post-developmental physiology, establishing safety and evaluating risk when treating children will be paramount. As such insight provided by preclinical and clinical information will be critical. This review will highlight the cardiovascular manifestations caused by the RASopathies and will discuss the emerging therapies for treatment.
Collapse
Affiliation(s)
- Jae-Sung Yi
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, SHM B226D, 333 Cedar Street, New Haven, CT, 06520-8066, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
14
|
Papadopoulou A, Bountouvi E. Skeletal defects and bone metabolism in Noonan, Costello and cardio-facio-cutaneous syndromes. Front Endocrinol (Lausanne) 2023; 14:1231828. [PMID: 37964950 PMCID: PMC10641803 DOI: 10.3389/fendo.2023.1231828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Noonan, Costello and Cardio-facio-cutaneous syndromes belong to a group of disorders named RASopathies due to their common pathogenetic origin that lies on the Ras/MAPK signaling pathway. Genetics has eased, at least in part, the distinction of these entities as they are presented with overlapping clinical features which, sometimes, become more pronounced with age. Distinctive face, cardiac and skeletal defects are among the primary abnormalities seen in these patients. Skeletal dysmorphisms range from mild to severe and may include anterior chest wall anomalies, scoliosis, kyphosis, short stature, hand anomalies, muscle weakness, osteopenia or/and osteoporosis. Patients usually have increased serum concentrations of bone resorption markers, while markers of bone formation are within normal range. The causative molecular defects encompass the members of the Ras/MAPK/ERK pathway and the adjacent cascades, important for the maintenance of normal bone homeostasis. It has been suggested that modulation of the expression of specific molecules involved in the processes of bone remodeling may affect the osteogenic fate decision, potentially, bringing out new pharmaceutical targets. Currently, the laboratory imprint of bone metabolism on the clinical picture of the affected individuals is not clear, maybe due to the rarity of these syndromes, the small number of the recruited patients and the methods used for the description of their clinical and biochemical profiles.
Collapse
Affiliation(s)
- Anna Papadopoulou
- Laboratory of Clinical Biochemistry, University General Hospital “Attikon”, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
15
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
16
|
Ou Y, Yuan JC, Zheng Y, Zhang JM, He T, Liang Z, Zhou YK. Case report: Noonan syndrome with protein-losing enteropathy. Front Genet 2023; 14:1237821. [PMID: 37829277 PMCID: PMC10565653 DOI: 10.3389/fgene.2023.1237821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Background: Noonan syndrome (NS) is characterized by typical facial features, short stature, congenital heart defects and other comorbidities. Lymphedema and chylous pleural effusions are also common in NS, but protein-losing enteropathy (PLE) is rarely reported. Case presentation: We present the case of a 19-year-old Chinese woman presenting with PLE. Small intestine biopsy showed obvious expansion of lymphatic vessels. The gene mutation results of the patient indicated a c.184T>G missense mutation (p.Tyr62Asp) in the PTPN11 gene (NM_002834.3). Conclusion: NS accompanied by PLE is not common, but hypoproteinemia attributable to PLE may be more common in patients with NS than previously thought. It remains uncertain whether mutation of the PTPN11 gene is related to PLE, indicating that further research is needed.
Collapse
Affiliation(s)
- Yang Ou
- Department of Endocrinology and Metabolism, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jun-Chao Yuan
- Department of Endocrinology and Metabolism, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yao Zheng
- Department of Endocrinology and Metabolism, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jin-Man Zhang
- Department of Medical Genetics, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tian He
- Department of Gastroenterology, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zhi Liang
- Department of Information Center, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yi-Kun Zhou
- Department of Endocrinology and Metabolism, First People’s Hospital of Yunnan Province, The Kunhua Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
17
|
Alem D, Yang X, Beato F, Sarcar B, Tassielli AF, Dai R, Hogenson TL, Park MA, Jiang K, Cai J, Yuan Y, Fernandez-Zapico ME, Tan AC, Fleming JB, Xie H. Translational relevance of SOS1 targeting for KRAS-mutant colorectal cancer. Mol Carcinog 2023; 62:1025-1037. [PMID: 37042566 PMCID: PMC10330439 DOI: 10.1002/mc.23543] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/13/2023]
Abstract
It has been challenging to target mutant KRAS (mKRAS) in colorectal cancer (CRC) and other malignancies. Recent efforts have focused on developing inhibitors blocking molecules essential for KRAS activity. In this regard, SOS1 inhibition has arisen as an attractive approach for mKRAS CRC given its essential role as a guanine nucleotide exchange factor for this GTPase. Here, we demonstrated the translational value of SOS1 blockade in mKRAS CRC. We used CRC patient-derived organoids (PDOs) as preclinical models to evaluate their sensitivity to SOS1 inhibitor BI3406. A combination of in silico analyses and wet lab techniques was utilized to define potential predictive markers for SOS1 sensitivity and potential mechanisms of resistance in CRC. RNA-seq analysis of CRC PDOs revealed two groups of CRC PDOs with differential sensitivities to SOS1 inhibitor BI3406. The resistant group was enriched in gene sets involving cholesterol homeostasis, epithelial-mesenchymal transition, and TNF-α/NFκB signaling. Expression analysis identified a significant correlation between SOS1 and SOS2 mRNA levels (Spearman's ρ 0.56, p < 0.001). SOS1/2 protein expression was universally present with heterogeneous patterns in CRC cells but only minimal to none in surrounding nonmalignant cells. Only SOS1 protein expression was associated with worse survival in patients with RAS/RAF mutant CRC (p = 0.04). We also found that SOS1/SOS2 protein expression ratio >1 by immunohistochemistry (p = 0.03) instead of KRAS mutation (p = 1) was a better predictive marker to BI3406 sensitivity of CRC PDOs, concordant with the significant positive correlation between SOS1/SOS2 protein expression ratio and SOS1 dependency. Finally, we showed that GTP-bound RAS level underwent rebound even in BI3406-sensitive PDOs with no change of KRAS downstream effector genes, thus suggesting upregulation of guanine nucleotide exchange factor as potential cellular adaptation mechanisms to SOS1 inhibition. Taken together, our results show that high SOS1/SOS2 protein expression ratio predicts sensitivity to SOS1 inhibition and support further clinical development of SOS1-targeting agents in CRC.
Collapse
Affiliation(s)
- Diego Alem
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Xinrui Yang
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Francisca Beato
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Bhaswati Sarcar
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Alexandra F Tassielli
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Ruifan Dai
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Tara L Hogenson
- Department of Oncology, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Margaret A Park
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Kun Jiang
- Department of Pathology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida, USA
| | - Yu Yuan
- Department of Chemistry, University of Central Florida, Orlando, Florida, USA
| | - Martin E Fernandez-Zapico
- Department of Oncology, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Hao Xie
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
18
|
D’Antona L, Amato R, Brescia C, Rocca V, Colao E, Iuliano R, Blazer-Yost BL, Perrotti N. Kinase Inhibitors in Genetic Diseases. Int J Mol Sci 2023; 24:ijms24065276. [PMID: 36982349 PMCID: PMC10048847 DOI: 10.3390/ijms24065276] [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: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Over the years, several studies have shown that kinase-regulated signaling pathways are involved in the development of rare genetic diseases. The study of the mechanisms underlying the onset of these diseases has opened a possible way for the development of targeted therapies using particular kinase inhibitors. Some of these are currently used to treat other diseases, such as cancer. This review aims to describe the possibilities of using kinase inhibitors in genetic pathologies such as tuberous sclerosis, RASopathies, and ciliopathies, describing the various pathways involved and the possible targets already identified or currently under study.
Collapse
Affiliation(s)
- Lucia D’Antona
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rosario Amato
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Carolina Brescia
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Rocca
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Department of Experimental and Clinical Medicine, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Emma Colao
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rodolfo Iuliano
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Bonnie L. Blazer-Yost
- Department of Biology, Indiana University Purdue University, Indianapolis, IN 46202, USA
| | - Nicola Perrotti
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Correspondence:
| |
Collapse
|
19
|
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] [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
|
20
|
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
|
21
|
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] [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 Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Pediatrics and Genetics, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
22
|
Chen CP. Prenatal Diagnosis of Euploid Increased Nuchal Translucency on Fetal Ultrasound (I): Noonan Syndrome: Prenatal Diagnosis and Genetic Testing. J Med Ultrasound 2022; 30:257-260. [PMID: 36844761 PMCID: PMC9944828 DOI: 10.4103/jmu.jmu_78_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/28/2022] Open
Abstract
Prenatal diagnosis of euploid increased nuchal translucency (NT) remains a challenge to obstetricians and genetic counselors although increased euploid NT at prenatal diagnosis can be associated with a favorable outcome. Prenatal diagnosis of euploid increased NT should include a differential diagnosis of pathogenetic copy number variants and RASopathy disorders (RDs) including Noonan syndrome (NS). Therefore, chromosomal microarray analysis, whole-exome sequencing, RD testing, and protein-tyrosine phosphatase, nonreceptor type 11 (PTPN11) gene testing may be necessary under such a circumstance. In this report, a comprehensive review of NS with its prenatal diagnosis and genetic testing is presented.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan,Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan,Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan,Address for correspondence: Prof. Chih-Ping Chen, Department of Obstetrics and Gynecology, MacKay Memorial Hospital, No. 92, Section 2, Chung-Shan North Road, Taipei 10449, Taiwan. E-mail:
| |
Collapse
|
23
|
Chaves Rabelo N, Gomes ME, de Oliveira Moraes I, Cantagalli Pfisterer J, Loss de Morais G, Antunes D, Caffarena ER, Llerena Jr J, Gonzalez S. RASopathy Cohort of Patients Enrolled in a Brazilian Reference Center for Rare Diseases: A Novel Familial LZTR1 Variant and Recurrent Mutations. Appl Clin Genet 2022; 15:153-170. [PMID: 36304179 PMCID: PMC9595068 DOI: 10.2147/tacg.s372761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/03/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose Noonan syndrome and related disorders are genetic conditions affecting 1:1000-2000 individuals. Variants causing hyperactivation of the RAS/MAPK pathway lead to phenotypic overlap between syndromes, in addition to an increased risk of pediatric tumors. DNA sequencing methods have been optimized to provide a molecular diagnosis for clinical and genetic heterogeneity conditions. This work aimed to investigate the genetic basis in RASopathy patients through Next Generation Sequencing in a Reference Center for Rare Diseases (IFF/Fiocruz) and implement the precision medicine at a public health institute in Brazil. Patients and Methods This study comprises 26 cases with clinical suspicion of RASopathies. Sanger sequencing was used to screen variants in exons usually affected in the PTPN11 and HRAS genes for cases with clinical features of Noonan and Costello syndrome, respectively. Posteriorly, negative and new cases with clinical suspicion of RASopathy were analyzed by clinical or whole-exome sequencing. Results Molecular analysis revealed recurrent variants and a novel LZTR1 missense variant: 24 unrelated individuals with pathogenic variants [PTPN11(11), NF1(2), SOS1(2), SHOC2(2), HRAS(1), BRAF(1), LZTR (1), RAF1(1), KRAS(1), RIT1(1), a patient with co-occurrence of PTPN11 and NF1 mutations (1)]; familial cases carrying a known pathogenic variant in PTPN11 (mother-two children), and a previously undescribed paternally inherited variant in LZTR1. The comparative modeling analysis of the novel LZTR1 variant p.Pro225Leu showed local and global changes in the secondary and tertiary structures, showing a decrease of about 1% in the β-sheet content. Furthermore, evolutionary conservation indicated that Pro225 is in a highly conserved region, as observed for known dominant pathogenic variants in this protein. Conclusion Bringing precision medicine through NGS towards congenital syndromes promotes a better understanding of complex clinical and/or undiagnosed cases. The National Policy for Rare Diseases in Brazil emphasizes the importance of incorporating and optimizing diagnostic methodologies in the Unified Brazilian Health System (SUS). Therefore, this work is an important step for the NGS inclusion in diagnostic genetic routine in the public health system.
Collapse
Affiliation(s)
- Natana Chaves Rabelo
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Maria Eduarda Gomes
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Isabelle de Oliveira Moraes
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Juliana Cantagalli Pfisterer
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | | | - Deborah Antunes
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Ernesto Raúl Caffarena
- Grupo de Biofísica Computacional e Modelagem Molecular, Programa de Computação Científica, Fundação Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Juan Llerena Jr
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Faculdade de Medicina de Petrópolis, FASE, Petrópolis, RJ, Brazil,INAGEMP, Rio de Janeiro, RJ, Brazil,Correspondence: Juan Llerena Jr, Email
| | - Sayonara Gonzalez
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
24
|
Molecular and clinical profile of patients referred as Noonan or Noonan-like syndrome in Greece: a cohort of 86 patients. Eur J Pediatr 2022; 181:3691-3700. [PMID: 35904599 DOI: 10.1007/s00431-022-04574-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
UNLABELLED Noonan syndrome (NS) is an autosomal dominant disorder characterized by clinical and genetic heterogeneity. It belongs to a wider group of pathologies, known as Rasopathies, due to the implication of genes encoding components of the Ras/MAPK signalling pathway. Recording the genetic alterations across populations helps assessing specific features to specific genes which is essential for better disease's recognition, prognosis and monitoring. Herein, we report the clinical and molecular data of a Greek cohort comprising of 86 NS or NS-like patients admitted at a single tertiary Centre in Athens, Greece. The analysis was performed using Sanger and next-generation sequencing, comprising 14 different genes. The mutational rates of the confirmed NS-associated genes in the Greek NS population are as follows: PTPN11 32.5%; RIT1 5.8%; SOS1 4.7%; BRAF 1.2%; CBL 1.2%; KRAS 1.2%; MAP2K1 1.2%; RAF1 1.2%; SHOC2 1.2%, corresponding to 50% of positivity in total NS population. The genotype-phenotype analysis showed statistically significant differences in craniofacial dysmorphisms (p = 0.005) and pulmonary valve stenosis (PS) (p < 0.001) frequencies between patients harbouring a pathogenic variant and patients without pathogenic variant in any of the tested genes. Patients with at least a pathogenic variant had 6.71 times greater odds to develop PS compared to pathogenic variant-negative patients (OR = 6.71, 95%; CI = (2.61, 17.27)). PTPN11 positive patients showed higher frequency of epicanthal folds (p = 0.004), ptosis (p = 0.001) and coarseness (p = 0.001) and lower frequency of neurological findings (p = 0.006), compared to patients carrying pathogenic variants in other genes. CONCLUSION Craniofacial dysmorphism and PS prevail among pathogenic variant positive compared to pathogenic variant negative NS and NS-like patients while neurological defects are less common in PTPN11-affected NS patients compared to patients harbouring pathogenic variants in other genes. The significant prevalence of the Ras/MAPK pathogenic variants (17.4%), other than PTPN11, in Greek NS patients, highlights the necessity of a wider spectrum of molecular diagnosis. WHAT IS KNOWN • Noonan syndrome (NS) has been associated with pathogenic variants in molecules-components of the Ras/MAPK pathway. • Clinical and genetic description of NS patients worldwide helps establishing personalized monitoring. WHAT IS NEW • NS and NS-like mutational rate in Greece reaches 50% with pathogenic variants identified mostly in PTPN11 (32.5%), RIT1 (6%) and SOS1 (4.7%) genes. • The risk for pulmonary stenosis increases 6.71-fold in NS patients with a pathogenic variant compared to patients without genetic alterations.
Collapse
|
25
|
Janas-Naze A, Malkiewicz K, Zhang W. Clinical Findings in Children with Noonan Syndrome—A 17-Year Retrospective Study in an Oral Surgery Center. CHILDREN 2022; 9:children9101486. [PMID: 36291422 PMCID: PMC9600410 DOI: 10.3390/children9101486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022]
Abstract
To date, only a limited number of publications have studied the specific oral and maxillofacial findings in patients diagnosed with Noonan syndrome (NS), which is an example of a genetically heterogeneous RASopathy. In this retrospective study, we aimed to ascertain the genotype–phenotype correlations between genetic mutations and certain diagnoses in the field of oral surgery. We collected surgical and genetic data from 42 children (median age, 12 years) who had a confirmed diagnosis of NS and underwent surgery in the Department of Oral Surgery, Medical University of Lodz, over a 17-year period, from 2004 to 2021. In total, 17 patients with mutations of the PTPN11 gene were diagnosed with over-retained deciduous teeth and supernumerary teeth. An amount of 7 patients with mutations of the SOS1 gene were diagnosed with mandibular compound odontomas. Finally, 12 patients with mutations of the LZTR1 gene were diagnosed with bilateral or unilateral central giant cell granulomas in the mandible. Although craniofacial features of many genetic disorders have been previously described in the literature, this study determined the genotype–phenotype correlations in the field of oral surgery.
Collapse
Affiliation(s)
- Anna Janas-Naze
- Department of Oral Surgery, Medical University of Lodz, 92-213 Lodz, Poland
- Correspondence: ; Tel.: +48-426757529
| | - Konrad Malkiewicz
- Department of Orthodontics, Medical University of Lodz, 92-213 Lodz, Poland
| | - Wei Zhang
- Shanxi Oral Disease Prevention and Treatment Center, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| |
Collapse
|
26
|
Mercadante F, Piro E, Busè M, Salzano E, Ferrara A, Serra G, Passarello C, Corsello G, Piccione M. Cutis verticis gyrata and Noonan syndrome: report of two cases with pathogenetic variant in SOS1 gene. Ital J Pediatr 2022; 48:152. [PMID: 35986401 PMCID: PMC9392323 DOI: 10.1186/s13052-022-01340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Background Noonan and Noonan-like syndromes are multisystem genetic disorders, mainly with autosomal dominant trasmission, caused by mutations in several genes. Missense pathogenetic variants of SOS1 gene are the second most common cause of Noonan syndrome (NS) and account approximately for 13% to 17% of cases. Subjects carrying a pathogenetic variant in SOS1 gene tend to exhibit a distinctive phenotype that is characterized by ectodermal abnormalities. Cutis verticis gyrata (CVG) is a rare disease, congenital or acquired, characterized by the redundancy of skin on scalp, forming thick skin folds and grooves of similar aspect to cerebral cortex gyri. Several references in the literature have reported association between nonessential primary form of CVG and NS. Case presentation we report two cases of newborns with CVG and phenotype suggestive for NS who have been diagnosed to harbour the same pathogenetic variant in SOS1 gene. Conclusions previously described patients with NS presenting CVG had received only clinical diagnosis. Therefore we report the first patients with CVG in which the clinical suspicion of NS is confirmed by molecolar analysis.
Collapse
|
27
|
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
|
28
|
Ketcham JM, Haling J, Khare S, Bowcut V, Briere DM, Burns AC, Gunn RJ, Ivetac A, Kuehler J, Kulyk S, Laguer J, Lawson JD, Moya K, Nguyen N, Rahbaek L, Saechao B, Smith CR, Sudhakar N, Thomas NC, Vegar L, Vanderpool D, Wang X, Yan L, Olson P, Christensen JG, Marx MA. Design and Discovery of MRTX0902, a Potent, Selective, Brain-Penetrant, and Orally Bioavailable Inhibitor of the SOS1:KRAS Protein-Protein Interaction. J Med Chem 2022; 65:9678-9690. [PMID: 35833726 PMCID: PMC9340770 DOI: 10.1021/acs.jmedchem.2c00741] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
SOS1 is one of the major guanine nucleotide exchange
factors that
regulates the ability of KRAS to cycle through its “on”
and “off” states. Disrupting the SOS1:KRASG12C protein–protein interaction (PPI) can increase the proportion
of GDP-loaded KRASG12C, providing a strong mechanistic
rationale for combining inhibitors of the SOS1:KRAS complex with inhibitors
like MRTX849 that target GDP-loaded KRASG12C. In this report,
we detail the design and discovery of MRTX0902—a potent, selective,
brain-penetrant, and orally bioavailable SOS1 binder that disrupts
the SOS1:KRASG12C PPI. Oral administration of MRTX0902
in combination with MRTX849 results in a significant increase in antitumor
activity relative to that of either single agent, including tumor
regressions in a subset of animals in the MIA PaCa-2 tumor mouse xenograft
model.
Collapse
Affiliation(s)
- John M Ketcham
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Jacob Haling
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Shilpi Khare
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Vickie Bowcut
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - David M Briere
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Aaron C Burns
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Robin J Gunn
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Anthony Ivetac
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Jon Kuehler
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Svitlana Kulyk
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Jade Laguer
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - J David Lawson
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Krystal Moya
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Natalie Nguyen
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Lisa Rahbaek
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Barbara Saechao
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Christopher R Smith
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Niranjan Sudhakar
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Nicole C Thomas
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Laura Vegar
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Darin Vanderpool
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Xiaolun Wang
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Larry Yan
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Peter Olson
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - James G Christensen
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Matthew A Marx
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| |
Collapse
|
29
|
Gao W, Li X. Prenatal ultrasound diagnosis of primary myxomatous degeneration of cardiac valves in a fetus: Case report. J Obstet Gynaecol Res 2022; 48:2620-2623. [PMID: 35810462 DOI: 10.1111/jog.15331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 04/27/2022] [Accepted: 05/20/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Wen‐Juan Gao
- Department of Ultrasound Weifang People's Hospital, the Affiliated Hospital of Weifang Medical University Weifang Shandong China
| | - Xue‐Ning Li
- Weifang Medical University Weifang Shandong China
| |
Collapse
|
30
|
Bonetti G, Paolacci S, Samaja M, Maltese PE, Michelini S, Michelini S, Michelini S, Ricci M, Cestari M, Dautaj A, Medori MC, Bertelli M. Low Efficacy of Genetic Tests for the Diagnosis of Primary Lymphedema Prompts Novel Insights into the Underlying Molecular Pathways. Int J Mol Sci 2022; 23:ijms23137414. [PMID: 35806420 PMCID: PMC9267137 DOI: 10.3390/ijms23137414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Lymphedema is a chronic inflammatory disorder caused by ineffective fluid uptake by the lymphatic system, with effects mainly on the lower limbs. Lymphedema is either primary, when caused by genetic mutations, or secondary, when it follows injury, infection, or surgery. In this study, we aim to assess to what extent the current genetic tests detect genetic variants of lymphedema, and to identify the major molecular pathways that underlie this rather unknown disease. We recruited 147 individuals with a clinical diagnosis of primary lymphedema and used established genetic tests on their blood or saliva specimens. Only 11 of these were positive, while other probands were either negative (63) or inconclusive (73). The low efficacy of such tests calls for greater insight into the underlying mechanisms to increase accuracy. For this purpose, we built a molecular pathways diagram based on a literature analysis (OMIM, Kegg, PubMed, Scopus) of candidate and diagnostic genes. The PI3K/AKT and the RAS/MAPK pathways emerged as primary candidates responsible for lymphedema diagnosis, while the Rho/ROCK pathway appeared less critical. The results of this study suggest the most important pathways involved in the pathogenesis of lymphedema, and outline the most promising diagnostic and candidate genes to diagnose this disease.
Collapse
Affiliation(s)
- Gabriele Bonetti
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- Correspondence: ; Tel.: +39-0365-62-061
| | - Stefano Paolacci
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | | | | | - Sandro Michelini
- Vascular Diagnostics and Rehabilitation Service, Marino Hospital, ASL Roma 6, 00047 Marino, Italy;
| | - Serena Michelini
- Unit of Physical Medicine, “Sapienza” University of Rome, 00185 Rome, Italy;
| | | | - Maurizio Ricci
- Division of Rehabilitation Medicine, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy;
| | - Marina Cestari
- Study Centre Pianeta Linfedema, 05100 Terni, Italy;
- Lymphology Sector of the Rehabilitation Service, USLUmbria2, 05100 Terni, Italy
| | - Astrit Dautaj
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Maria Chiara Medori
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Matteo Bertelli
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- MAGI Group, 25010 San Felice del Benaco, Italy;
- MAGI Euregio, 39100 Bolzano, Italy
| |
Collapse
|
31
|
MR lymphangiography of lymphatic abnormalities in children and adults with Noonan syndrome. Sci Rep 2022; 12:11164. [PMID: 35778409 PMCID: PMC9249771 DOI: 10.1038/s41598-022-13806-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Noonan syndrome is associated with complex lymphatic abnormalities. We report dynamic-contrast enhanced MR lymphangiography (DCMRL) findings in children and adults with Noonan syndrome to further elucidate this complex disease spectrum. A retrospective evaluation of patients with confirmed Noonan syndrome and clinical signs of lymphatic dysfunction undergoing DCMRL between 01/2019 and 04/2021 was performed. MRL included T2-weighted imaging (T2w) and DCMRL. Clinical history/presentation and genetic variants were recorded. T2w-imaging was evaluated for central lymphatic abnormalities and edema distribution. DCMRL was evaluated regarding the presence of cisterna chyli/thoracic duct, lymphatic leakages, pathological lymphatic reflux and abnormal lymphatic perfusion. The time from start of contrast-injection to initial enhancement of the thoracic duct venous junction was measured to calculate the speed of contrast propagation. Eleven patients with Noonan syndrome with lymphatic abnormalities (5 female, 6 male; 7 infants, 4 adults; mean age 10.8 ± 16.4 years) were identified (PTPN11 n = 5/11 [45.5%], RIT1 n = 5/11 [45.5%], KRAS n = 1/11 [9%]). Patients had a chylothorax (n = 10/11 [91%]) and/or pulmonary lymphangiectasia [dilated pulmonary lymph vessels] (n = 9/11 [82%]). Mediastinal/pulmonary edema was depicted in 9/11 (82%) patients. The thoracic duct (TD) was (partially) absent in 10/11 (91%) cases. DCMRL showed lymphatic reflux into intercostal (n = 11/11 [100%]), mediastinal (n = 9/11 [82%]), peribronchial (n = 8/11 [73%]), peripheral (n = 5/11 [45.5%]) and genital lymphatics (n = 4/11 [36%]). Abnormal pulmonary/pleural lymphatic perfusion was seen in 8/11 patients (73%). At infancy peripheral/genital edema was more prevalent in patients with RIT1 than PTPN11 (n = 3/5 vs. n = 0/5). Compared to patients with PTPN11 who had fast lymphatic enhancement in 4/5 patients, enhancement took markedly longer in 4/5 patients with RIT1-mutations. Thoracic duct dysplasia, intercostal reflux and pulmonary/pleural lymphatic perfusion are characteristic findings in patients with Noonan syndrome presenting with chylothorax and/or pulmonary lymphangiectasia. Central lymphatic flow abnormalities show possible phenotypical differences between PTPN11 and RIT1-mutations.
Collapse
|
32
|
Abstract
Noonan syndrome is a genetic disorder characteried by short stature, typical facial features, developmental delay, and CHD. In this single-centre retrospective study, we analysed typical Noonan syndrome-related electrocardiographic features in 95 patients with clinically and molecularly confirmed Noonan syndrome. Typical Noonan syndrome-related electrocardiographic features are left axis deviation, small left precordial R-waves, large right precordial S-waves, abnormal Q-wave, and abnormal wide QRS complex. In this representative cohort, CHD was found in 59 patients (62.1%) and typical Noonan syndrome-related electrographic features in 60 patients (63.2%). The typical Noonan syndrome-related electrographic features were also increased over baseline in patients without CHD (41.7%). Of all 95 patients, left axis deviation was seen in 46.3%, small left precordial R-waves in 30.5%, large right precordial S-waves in 5.3%, and abnormal Q-wave and wide QRS complex in 2.1%. There was no significant difference in the frequency of the individual-specific electrographic features between the group with CHD and the group without CHD. However, there were significantly more patients with a small left precordial R-wave in the subgroup with pulmonary stenosis compared to patients without pulmonary stenosis. Conclusion: Specific Noonan syndrome-related electrographic features are frequently present in patients with Noonan syndrome, also in the absence of CHD. These results suggest that there may be a continuum of cardiac anomalies from overt CHD to milder abnormalities that are only seen on electrocardiogram.
Collapse
|
33
|
Bruno JL, Shrestha SB, Reiss AL, Saggar M, Green T. Altered canonical and striatal-frontal resting state functional connectivity in children with pathogenic variants in the Ras/mitogen-activated protein kinase pathway. Mol Psychiatry 2022; 27:1542-1551. [PMID: 35087195 PMCID: PMC9106817 DOI: 10.1038/s41380-021-01422-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022]
Abstract
Mounting evidence supports the role of the Ras/mitogen-activated protein kinase (Ras/MAPK) pathway in neurodevelopmental disorders. Here, the authors used a genetics-first approach to examine how Ras/MAPK pathogenic variants affect the functional organization of the brain and cognitive phenotypes including weaknesses in attention and inhibition. Functional MRI was used to examine resting state functional connectivity (RSFC) in association with Ras/MAPK pathogenic variants in children with Noonan syndrome (NS). Participants (age 4-12 years) included 39 children with NS (mean age 8.44, SD = 2.20, 25 females) and 49 typically developing (TD) children (mean age 9.02, SD = 9.02, 33 females). Twenty-eight children in the NS group and 46 in the TD group had usable MRI data and were included in final analyses. The results indicated significant hyperconnectivity for the NS group within canonical visual, ventral attention, left frontoparietal and limbic networks (p < 0.05 FWE). Higher connectivity within canonical left frontoparietal and limbic networks positively correlated with cognitive function within the NS but not the TD group. Further, the NS group demonstrated significant group differences in seed-based striatal-frontal connectivity (Z > 2.6, p < 0.05 FWE). Hyperconnectivity within canonical brain networks may represent an intermediary phenotype between Ras/MAPK pathogenic variants and cognitive phenotypes, including weaknesses in attention and inhibition. Altered striatal-frontal connectivity corresponds with smaller striatal volume and altered white matter connectivity previously documented in children with NS. These results may indicate delayed maturation and compensatory mechanisms and they are important for understanding the pathophysiology underlying cognitive phenotypes in NS and in the broader population of children with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jennifer L Bruno
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Sharon B Shrestha
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Allan L Reiss
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics and Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Manish Saggar
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tamar Green
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
34
|
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
|
35
|
Chou YT, Bivona TG. Inhibition of SHP2 as an approach to block RAS-driven cancers. Adv Cancer Res 2022; 153:205-236. [PMID: 35101231 DOI: 10.1016/bs.acr.2021.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The non-receptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) is a critical component of RAS/MAPK signaling by acting upstream of RAS to promote oncogenic signaling and tumor growth. Over three decades, SHP2 was considered "undruggable" because enzymatic active-site inhibitors generally showed off-target inhibition of other proteins and low membrane permeability. More recently, allosteric SHP2 inhibitors with striking inhibitory potency have been developed. These small molecules effectively block the signal transduction between receptor tyrosine kinases (RTKs) and RAS/MAPK signaling and show efficacy in preclinical cancer models. Moreover, clinical evaluation of these allosteric SHP2 inhibitors is ongoing. RAS proteins which harbor transforming properties by gain-of-function mutations are present in various cancer types. While inhibitors of KRASG12C show early clinical promise, resistance remains a challenge and other forms of oncogenic RAS remain to be selectively inhibited. Here, we summarize the role of SHP2 in RAS-driven cancers and the therapeutic potential of allosteric SHP2 inhibitors as a strategy to block RAS-driven cancers.
Collapse
Affiliation(s)
- Yu-Ting Chou
- Department of Medicine, Division of Hematology and Oncology, and The Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Trever G Bivona
- Department of Medicine, Division of Hematology and Oncology, and The Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
36
|
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
|
37
|
Abstract
Activating mutations in RAS genes are the most common genetic driver of human cancers. Yet, drugging this small GTPase has proven extremely challenging and therapeutic strategies targeting these recurrent alterations have long had limited success. To circumvent this difficulty, research has focused on the molecular dissection of the RAS pathway to gain a more-precise mechanistic understanding of its regulation, with the hope to identify new pharmacological approaches. Here, we review the current knowledge on the (dys)regulation of the RAS pathway, using melanoma as a paradigm. We first present a map of the main proteins involved in the RAS pathway, highlighting recent insights into their molecular roles and diverse mechanisms of regulation. We then overview genetic data pertaining to RAS pathway alterations in melanoma, along with insight into other cancers, that inform the biological function of members of the pathway. Finally, we describe the clinical implications of RAS pathway dysregulation in melanoma, discuss past and current approaches aimed at drugging the RAS pathway, and outline future opportunities for therapeutic development. Summary: This Review describes the molecular regulation of the RAS pathway, presents the clinical consequences of its pathological activation in human cancer, and highlights recent advances towards its therapeutic inhibition, using melanoma as an example.
Collapse
Affiliation(s)
- Amira Al Mahi
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, INSERM U1052 CNRS UMR5286, Tumor Escape, Resistance and Immunity Department, 69008 Lyon, France
| | - Julien Ablain
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, INSERM U1052 CNRS UMR5286, Tumor Escape, Resistance and Immunity Department, 69008 Lyon, France
| |
Collapse
|
38
|
Butler MG, Miller BS, Romano A, Ross J, Abuzzahab MJ, Backeljauw P, Bamba V, Bhangoo A, Mauras N, Geffner M. Genetic conditions of short stature: A review of three classic examples. Front Endocrinol (Lausanne) 2022; 13:1011960. [PMID: 36339399 PMCID: PMC9634554 DOI: 10.3389/fendo.2022.1011960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan, Turner, and Prader-Willi syndromes are classical genetic disorders that are marked by short stature. Each disorder has been recognized for several decades and is backed by extensive published literature describing its features, genetic origins, and optimal treatment strategies. These disorders are accompanied by a multitude of comorbidities, including cardiovascular issues, endocrinopathies, and infertility. Diagnostic delays, syndrome-associated comorbidities, and inefficient communication among the members of a patient's health care team can affect a patient's well-being from birth through adulthood. Insufficient information is available to help patients and their multidisciplinary team of providers transition from pediatric to adult health care systems. The aim of this review is to summarize the clinical features and genetics associated with each syndrome, describe best practices for diagnosis and treatment, and emphasize the importance of multidisciplinary teams and appropriate care plans for the pediatric to adult health care transition.
Collapse
Affiliation(s)
- Merlin G. Butler
- Department of Psychiatry & Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, United States
- *Correspondence: Merlin G. Butler,
| | - Bradley S. Miller
- Pediatric Endocrinology, University of Minnesota Masonic Children’s Hospital, Minneapolis, MN, United States
| | - Alicia Romano
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Judith Ross
- Department of Pediatrics, Nemours Children’s Health, Wilmington, DE, United States
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Philippe Backeljauw
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Vaneeta Bamba
- Division of Endocrinology, Children’s Hospital of Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Amrit Bhangoo
- Pediatric Endocrinology, Children's Health of Orange County (CHOC) Children’s Hospital, Orange, CA, United States
| | - Nelly Mauras
- Division of Endocrinology, Nemours Children’s Health, Jacksonville, FL, United States
| | - Mitchell Geffner
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
39
|
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
|
40
|
Brozou T, Yasin L, Brandes D, Picard D, Walter C, Varghese J, Dugas M, Fischer U, Borkhardt A, Haas OA. Resolving inherited and de novo germline predisposing sequence variants by means of whole exome trio analyses in childhood hematological malignancies. Front Pediatr 2022; 10:1080347. [PMID: 36824296 PMCID: PMC9941195 DOI: 10.3389/fped.2022.1080347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/28/2022] [Indexed: 02/10/2023] Open
Abstract
Molecular screening tools have significantly eased the assessment of potential germline susceptibility factors that may underlie the development of pediatric malignancies. Most of the hitherto published studies utilize the comparative analyses of the respective patients' germline and tumor tissues for this purpose. Since this approach is not able to discriminate between de novo and inherited sequence variants, we performed whole exome trio analyses in a consecutive series of 131 children with various forms of hematologic malignancies and their parents. In total, we identified 458 de novo variants with a range from zero to 28 (median value = 3) per patient, although most of them (58%) had only up to three per exome. Overall, we identified bona fide cancer predisposing alterations in five of the investigated 131 (3.8%) patients. Three of them had de novo pathogenic lesions in the SOS1, PTPN11 and TP53 genes and two of them parentally inherited ones in the STK11 and PMS2 genes that are specific for a Peutz-Jeghers and a constitutional mismatch repair deficiency (CMMRD) syndrome, respectively. Notwithstanding that we did not identify a disease-specific alteration in the two cases with the highest number of de novo variants, one of them developed two almost synchronous malignancies: a myelodysplastic syndrome and successively within two months a cerebral astrocytoma. Moreover, we also found that the rate of de novo sequence variants in the offspring increased especially with the age of the father, but less so with that of the mother. We therefore conclude that trio analyses deliver an immediate overview about the inheritance pattern of the entire spectrum of sequence variants, which not only helps to securely identify the de novo or inherited nature of genuinely disease-related lesions, but also of all other less obvious variants that in one or the other way may eventually advance our understanding of the disease process.
Collapse
Affiliation(s)
- Triantafyllia Brozou
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Layal Yasin
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Danielle Brandes
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Martin Dugas
- Insititute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Oskar A Haas
- St. Anna Children's Hospital, Pediatric Clinic, Medical University, Vienna, Austria
| |
Collapse
|
41
|
Sigamani V, Rajasingh S, Gurusamy N, Panda A, Rajasingh J. In-Silico and In-Vitro Analysis of Human SOS1 Protein Causing Noonan Syndrome - A Novel Approach to Explore the Molecular Pathways. Curr Genomics 2021; 22:526-540. [PMID: 35386434 PMCID: PMC8905634 DOI: 10.2174/1389202922666211130144221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 11/22/2022] Open
Abstract
Aims Perform in-silico analysis of human SOS1 mutations to elucidate their pathogenic role in Noonan syndrome (NS). Background NS is an autosomal dominant genetic disorder caused by single nucleotide mutation in PTPN11, SOS1, RAF1, and KRAS genes. NS is thought to affect approximately 1 in 1000. NS patients suffer different pathogenic effects depending on the mutations they carry. Analysis of the mutations would be a promising predictor in identifying the pathogenic effect of NS. Methods We performed computational analysis of the SOS1 gene to identify the pathogenic nonsynonymous single nucleotide polymorphisms (nsSNPs) th a t cause NS. SOS1 variants were retrieved from the SNP database (dbSNP) and analyzed by in-silico tools I-Mutant, iPTREESTAB, and MutPred to elucidate their structural and functional characteristics. Results We found that 11 nsSNPs of SOS1 that were linked to NS. 3D modeling of the wild-type and the 11 nsSNPs of SOS1 showed that SOS1 interacts with cardiac proteins GATA4, TNNT2, and ACTN2. We also found that GRB2 and HRAS act as intermediate molecules between SOS1 and cardiac proteins. Our in-silico analysis findings were further validated using induced cardiomyocytes (iCMCs) derived from NS patients carrying SOS1 gene variant c.1654A>G (NSiCMCs) and compared to control human skin fibroblast-derived iCMCs (C-iCMCs). Our in vitro data confirmed that the SOS1, GRB2 and HRAS gene expressions as well as the activated ERK protein, were significantly decreased in NS-iCMCs when compared to C-iCMCs. Conclusion This is the first in-silico and in vitro study demonstrating that 11 nsSNPs of SOS1 play deleterious pathogenic roles in causing NS.
Collapse
Affiliation(s)
- Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Arunima Panda
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
42
|
Vriz O, AlSergani H, Elshaer AN, Shaik A, Mushtaq AH, Lioncino M, Alamro B, Monda E, Caiazza M, Mauro C, Bossone E, Al-Hassnan ZN, Albert-Brotons D, Limongelli G. A complex unit for a complex disease: the HCM-Family Unit. Monaldi Arch Chest Dis 2021; 92. [PMID: 34964577 DOI: 10.4081/monaldi.2021.2147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a group of heterogeneous disorders that are most commonly passed on in a heritable manner. It is a relatively rare disease around the globe, but due to increased rates of consanguinity within the Kingdom of Saudi Arabia, we speculate a high incidence of undiagnosed cases. The aim of this paper is to elucidate a systematic approach in dealing with HCM patients and since HCM has variable presentation, we have summarized differentials for diagnosis and how different subtypes and genes can have an impact on the clinical picture, management and prognosis. Moreover, we propose a referral multi-disciplinary team HCM-Family Unit in Saudi Arabia and an integrated role in a network between King Faisal Hospital and Inherited and Rare Cardiovascular Disease Unit-Monaldi Hospital, Italy (among the 24 excellence centers of the European Reference Network (ERN) GUARD-Heart). Graphical Abstract.
Collapse
Affiliation(s)
- Olga Vriz
- Department of Cardiology, King Faisal Specialist Hospital and Research Center, Riyadh.
| | - Hani AlSergani
- Department of Cardiology, King Faisal Specialist Hospital and Research Center, Riyadh.
| | | | | | | | - Michele Lioncino
- Inherited and Rare Cardiovascular Disease Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", AORN dei Colli, Monaldi Hospital, Naples.
| | - Bandar Alamro
- Department of Cardiology, King Faisal Specialist Hospital and Research Center, Riyadh.
| | - Emanuele Monda
- Inherited and Rare Cardiovascular Disease Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", AORN dei Colli, Monaldi Hospital, Naples.
| | - Martina Caiazza
- Inherited and Rare Cardiovascular Disease Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", AORN dei Colli, Monaldi Hospital, Naples.
| | - Ciro Mauro
- Department of Cardiology, Cardarelli Hospital, Naples.
| | | | - Zuhair N Al-Hassnan
- Cardiovascular Genetics Program and Department of Medical Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh.
| | - Dimpna Albert-Brotons
- Department of Cardiology, King Faisal Specialist Hospital and Research Center, Riyadh.
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Disease Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", AORN dei Colli, Monaldi Hospital, Naples.
| |
Collapse
|
43
|
Fowlkes JL, Thrailkill KM, Bunn RC. RASopathies: The musculoskeletal consequences and their etiology and pathogenesis. Bone 2021; 152:116060. [PMID: 34144233 PMCID: PMC8316423 DOI: 10.1016/j.bone.2021.116060] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023]
Abstract
The RASopathies comprise an ever-growing number of clinical syndromes resulting from germline mutations in components of the RAS/MAPK signaling pathway. While multiple organs and tissues may be affected by these mutations, this review will focus on how these mutations specifically impact the musculoskeletal system. Herein, we review the genetics and musculoskeletal phenotypes of these syndromes in humans. We discuss how mutations in the RASopathy syndromes have been studied in translational mouse models. Finally, we discuss how signaling molecules within the RAS/MAPK pathway are involved in normal and abnormal bone biology in the context of osteoblasts, osteoclasts and chondrocytes.
Collapse
Affiliation(s)
- John L Fowlkes
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America.
| | - Kathryn M Thrailkill
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - R Clay Bunn
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| |
Collapse
|
44
|
Vasic V, Jones MSO, Haslinger D, Knaus LS, Schmeisser MJ, Novarino G, Chiocchetti AG. Translating the Role of mTOR- and RAS-Associated Signalopathies in Autism Spectrum Disorder: Models, Mechanisms and Treatment. Genes (Basel) 2021; 12:genes12111746. [PMID: 34828352 PMCID: PMC8624393 DOI: 10.3390/genes12111746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
Mutations affecting mTOR or RAS signaling underlie defined syndromes (the so-called mTORopathies and RASopathies) with high risk for Autism Spectrum Disorder (ASD). These syndromes show a broad variety of somatic phenotypes including cancers, skin abnormalities, heart disease and facial dysmorphisms. Less well studied are the neuropsychiatric symptoms such as ASD. Here, we assess the relevance of these signalopathies in ASD reviewing genetic, human cell model, rodent studies and clinical trials. We conclude that signalopathies have an increased liability for ASD and that, in particular, ASD individuals with dysmorphic features and intellectual disability (ID) have a higher chance for disruptive mutations in RAS- and mTOR-related genes. Studies on rodent and human cell models confirm aberrant neuronal development as the underlying pathology. Human studies further suggest that multiple hits are necessary to induce the respective phenotypes. Recent clinical trials do only report improvements for comorbid conditions such as epilepsy or cancer but not for behavioral aspects. Animal models show that treatment during early development can rescue behavioral phenotypes. Taken together, we suggest investigating the differential roles of mTOR and RAS signaling in both human and rodent models, and to test drug treatment both during and after neuronal development in the available model systems.
Collapse
Affiliation(s)
- Verica Vasic
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
| | - Mattson S. O. Jones
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
| | - Denise Haslinger
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Lisa S. Knaus
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Michael J. Schmeisser
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Andreas G. Chiocchetti
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-80658
| |
Collapse
|
45
|
García-Miñaúr S, Burkitt-Wright E, Verloes A, Shaikh G, Lebl J, Östman-Smith I, Wolf CM, Ortega Castelló E, Tartaglia M, Zenker M, Edouard T. European Medical Education Initiative on Noonan syndrome: A clinical practice survey assessing the diagnosis and clinical management of individuals with Noonan syndrome across Europe. Eur J Med Genet 2021; 65:104371. [PMID: 34757053 DOI: 10.1016/j.ejmg.2021.104371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Noonan syndrome (NS) is a rare genetic disorder caused by mutations in genes encoding components of the RAS/mitogen-activated protein kinase (MAPK) signalling pathway. Patients with NS exhibit certain characteristic features, including cardiac defects, short stature, distinctive facial appearance, skeletal abnormalities, cognitive deficits, and predisposition to certain cancers. Here, a clinical practice survey was developed to learn more about differences in the diagnosis and management of this disease across Europe. The aim was to identify gaps in the knowledge and management of this rare disorder. MATERIALS AND METHODS The European Medical Education Initiative on NS, which comprised a group of 10 experts, developed a 60-question clinical practice survey to gather information from European physicians on the diagnosis and clinical management of patients with diseases in the NS phenotypic spectrum. Physicians from three specialities (clinical genetics, paediatric endocrinology, paediatric cardiology) were invited to complete the survey by several national and European societies. Differences in answers provided by respondents between specialities and countries were analysed using contingency tables and the Chi-Squared test for independence. The Friedman's test was used for related samples. RESULTS Data were analysed from 364 respondents from 20 European countries. Most respondents came from France (21%), Spain (18%), Germany (16%), Italy (15%), United Kingdom (8%) and the Czech Republic (6%). Respondents were distributed evenly across three specialities: clinical genetics (30%), paediatric endocrinology (40%) and paediatric cardiology (30%). Care practices were generally aligned across the countries participating in the survey. Delayed diagnosis did not emerge as a critical issue, but certain unmet needs were identified, including transition of young patients to adult medical services and awareness of family support groups. CONCLUSION Data collected from this survey provide a comprehensive summary of the diagnosis and clinical management practices for patients with NS across different European countries.
Collapse
Affiliation(s)
- Sixto García-Miñaúr
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz Research Institute (IdiPAZ), Hospital Universitario La Paz, Madrid, Spain.
| | - Emma Burkitt-Wright
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Alain Verloes
- Department of Genetics, Hospital Robert Debré, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
| | - Guftar Shaikh
- Department of Paediatric Endocrinology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Jan Lebl
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Ingegerd Östman-Smith
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Cordula M Wolf
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Eduardo Ortega Castelló
- Department of Statistics and Data Science, Faculty of Statistical Studies, Complutense University of Madrid, Madrid, Spain
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, RESTORE INSERM UMR1301, Toulouse, France
| |
Collapse
|
46
|
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
|
47
|
Follansbee CW, Malloy-Walton L. Ventricular fibrillation due to a likely pathogenic SOS1 variant: An unrecognized etiology of infantile sudden death? HeartRhythm Case Rep 2021; 7:510-513. [PMID: 34434697 PMCID: PMC8377259 DOI: 10.1016/j.hrcr.2021.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Christopher W Follansbee
- Ward Family Heart Center, Children's Mercy Hospital Kansas City, Kansas City, Missouri.,University of Missouri School of Medicine Kansas City, Kansas City, Missouri
| | - Lindsey Malloy-Walton
- Ward Family Heart Center, Children's Mercy Hospital Kansas City, Kansas City, Missouri.,University of Missouri School of Medicine Kansas City, Kansas City, Missouri
| |
Collapse
|
48
|
Schreuder WH, van der Wal JE, de Lange J, van den Berg H. Multiple versus solitary giant cell lesions of the jaw: Similar or distinct entities? Bone 2021; 149:115935. [PMID: 33771761 DOI: 10.1016/j.bone.2021.115935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
The majority of giant cell lesions of the jaw present as a solitary focus of disease in bones of the maxillofacial skeleton. Less frequently they occur as multifocal lesions. This raises the clinical dilemma if these should be considered distinct entities and therefore each need a specific therapeutic approach. Solitary giant cell lesions of the jaw present with a great diversity of symptoms. Recent molecular analysis revealed that these are associated with somatic gain-of-function mutations in KRAS, FGFR1 or TRPV4 in a large component of the mononuclear stromal cells which all act on the RAS/MAPK pathway. For multifocal lesions, a small group of neoplastic multifocal giant cell lesions of the jaw remain after ruling out hyperparathyroidism. Strikingly, most of these patients are diagnosed with jaw lesions before the age of 20 years, thus before the completion of dental and jaw development. These multifocal lesions are often accompanied by a diagnosis or strong clinical suspicion of a syndrome. Many of the frequently reported syndromes belong to the so-called RASopathies, with germline or mosaic mutations leading to downstream upregulation of the RAS/MAPK pathway. The other frequently reported syndrome is cherubism, with gain-of-function mutations in the SH3BP2 gene leading through assumed and unknown signaling to an autoinflammatory bone disorder with hyperactive osteoclasts and defective osteoblastogenesis. Based on this extensive literature review, a RAS/MAPK pathway activation is hypothesized in all giant cell lesions of the jaw. The different interaction between and contribution of deregulated signaling in individual cell lineages and crosstalk with other pathways among the different germline- and non-germline-based alterations causing giant cell lesions of the jaw can be explanatory for the characteristic clinical features. As such, this might also aid in the understanding of the age-dependent symptomatology of syndrome associated giant cell lesions of the jaw; hopefully guiding ideal timing when installing treatment strategies in the future.
Collapse
Affiliation(s)
- Willem H Schreuder
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands; Department of Head and Neck Surgery and Oncology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Jacqueline E van der Wal
- Department of Pathology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan de Lange
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Henk van den Berg
- Department of Pediatrics / Oncology, Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands
| |
Collapse
|
49
|
Uludağ Alkaya D, Lissewski C, Yeşil G, Zenker M, Tüysüz B. Expanding the clinical phenotype of RASopathies in 38 Turkish patients, including the rare LZTR1, RAF1, RIT1 variants, and large deletion in NF1. Am J Med Genet A 2021; 185:3623-3633. [PMID: 34184824 DOI: 10.1002/ajmg.a.62410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/30/2021] [Accepted: 06/12/2021] [Indexed: 12/31/2022]
Abstract
RASopathies are a group of disorders caused by pathogenic variants in the genes encoding Ras/mitogen-activated protein kinase pathway and share overlapping clinical and molecular features. This study is aimed to describe the clinical and molecular features of 38 patients with RASopathies. Sanger or targeted next-generation sequencing of related genes and multiplex ligation-dependent-probe amplification analysis for NF1 were performed. The pathogenic variant detection rate was 94.4%. While PTPN11 was responsible for 50% of 18 patients with Noonan syndrome (NS), SOS1, LZTR1, RIT1, and RAF1 were responsible for the remaining 27.8%, 11.1%, 5.5%, and 5.5%, respectively. Three variants in LZTR1 were novel, of which two were identified in the compound heterozygous state in a patient with intellectual disability and hypertrophic cardiomyopathy, whereas the third variant was found in the heterozygous state in a patient with pulmonary stenosis and normal intelligence. We described pyloric stenosis, knee dislocation, and cleft palate in patients with SOS1, RIT1, and RAF1 variants, respectively, that was not previously reported. We detected a PTPN11 variant in three patients from same family with NS with multiple lentigines. BRAF and MAP2K2 variants were found in eight patients with Cardiofaciocutaneous syndrome. Two variants in HRAS were detected in two Costello syndrome patients, one with a mild and the other with a severe phenotype. While large NF1 deletions were identified in four Neurofibromatosis-NS patients with intellectual disability, intelligence was normal in one patient with missense variant. In conclusion, this study provided three novel variants in LZTR1 and expanded the clinical phenotype of rare RASopathies.
Collapse
Affiliation(s)
- Dilek Uludağ Alkaya
- Department of Pediatric Genetics, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Christina Lissewski
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Gözde Yeşil
- Department of Medical Genetics, Medical School, Bezmialem University, Istanbul, Turkey
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Beyhan Tüysüz
- Department of Pediatric Genetics, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
50
|
RASopathies: from germline mutations to somatic and multigenic diseases. Biomed J 2021; 44:422-432. [PMID: 34175492 PMCID: PMC8514848 DOI: 10.1016/j.bj.2021.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
The RAS-RAF-MEK-ERK signaling pathway is vital for different cellular mechanisms including cell proliferation, differentiation and apoptosis. This importance is highlighted by the high prevalence of mutations in RAS or related proteins of the pathway in cancers. More recently, development abnormalities have been linked to various germline mutations in this pathway and called RASopathies. Interestingly, rare disorders such as RAS-associated leukoproliferative diseases and histiocytosis have also been recently linked to multiple mutations in the same pathway, sometimes with the same mutation. This review will focus on germline RASopathies and rare somatic RASopathies and focus on how gain-of-function mutations in the same pathway can lead to various diseases.
Collapse
|