1
|
Cui Z, Wang Y, Luo F, Diao J, Yuan B. Mapping the current status and outlook of research on noonan syndrome over the last 26 years: a bibliometric and visual analysis. Front Genet 2024; 15:1488425. [PMID: 39726952 PMCID: PMC11669677 DOI: 10.3389/fgene.2024.1488425] [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: 08/30/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Noonan syndrome (NS) is a rare group of autosomal genetic disorders. In recent years, with the exploration and development of molecular diagnostic techniques, more and more researchers have begun to pay attention to NS. However, there is still a lack of reports on the bibliometric analysis of NS worldwide. This study aims to assess the current research status and development trend of NS, to explore the research hotspots and emerging topics, and to point out the direction for future scientific research. Methods Web of Science Core Collection was selected as the search database for bibliometric analysis of NS-related publications from 1998 to 2023. Statistical and visual analysis of the number of publications, countries, institutions, authors, journals, keywords, and references were analyzed using Citespace, VOSviewer, Scimago Graphica, and BibliometrixR. Results A total of 2041 articles were included in this study. The United States had the highest number of publications, and Istituto Superiore di Sanità, Italy, was the institution with the highest number of publications. TARTAGLIA M was the scientist with the highest number of publications and citations. Among the journals, AMERICAN JOURNAL OF MEDICAL GENETICS PART A has the highest output, and Nature Genetics is the most frequently cited. The reference with the highest outburst intensity is Roberts AE, LANCET, 2013. the cluster diagram divides all the keywords into seven categories, with the most vigorous outburst being "of function mutations." Conclusion Research hotspots in the field of NS focus on the correspondence between NS genotype and phenotype and the precise diagnosis of NS. Future research efforts will explore more deeply from the perspective of long-term intervention strategies for NS. There is an urgent need to rely on significant research countries, institutions, journals, and authors to lead the construction of a more robust global collaborative network that will enhance research efficacy.
Collapse
Affiliation(s)
- Zhengjiu Cui
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanyuan Wang
- Department of Pediatrics, Suqian Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian, China
| | - Fei Luo
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Juanjuan Diao
- Department of Pediatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Yuan
- Department of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Chen X, Zhang Q, Lu M, Feng Q, Qin L, Liao S. Prenatal finding of isolated ventricular septal defect: genetic association, outcomes and counseling. Front Genet 2024; 15:1447216. [PMID: 39415979 PMCID: PMC11479991 DOI: 10.3389/fgene.2024.1447216] [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: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
The innovation in ultrasound has greatly promoted the prenatal diagnosis of ventricular septal defect. As a minor lesion of congenital heart disease, the prenatal genetic counseling of isolated ventricular septal defect faces some challenges, including the true genetic correlationship, selection of appropriated testing methods to identify deleterious mutations, and avoidance of overdiagnosis and overintervention. Researchers have explored the prenatal diagnosis efficiency of commonly used cytogenetic and molecular genetic technologies. Small insertions/deletions and monogenic variants with phenotypic heterogeneity play important role and contribute to the comprehend of pathogenesis. Isolated ventricular septal defect fetuses without genetic finding and extracardiac structural abnormality generally have good pregnancy outcome. Long-term follow-up data is needed to describe the comprehensive map, such as the potential missed diagnosis especially late-onset syndromes, the impact on the quality of life and life expectancy. When conducting prenatal genetic counseling, strict adherence to ethical principles is needed to ensure that the rights of all parties involved are fully protected. Clinicians should carefully evaluate the risks and benefits and provide parents with sufficient information and advice to enable them to make informed decisions.
Collapse
Affiliation(s)
- Xin Chen
- Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Zhengzhou, China
| | - Qian Zhang
- Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Man Lu
- Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Qiuxia Feng
- National Health Commission Key Laboratory of Birth Defects Prevention, Zhengzhou, China
- Reproduction Medical Center, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Litao Qin
- Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shixiu Liao
- Medical Genetics Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Vijayakumar P, Mishra A, Deka RP, Pinto SM, Subbannayya Y, Sood R, Prasad TSK, Raut AA. Proteomics Analysis of Duck Lung Tissues in Response to Highly Pathogenic Avian Influenza Virus. Microorganisms 2024; 12:1288. [PMID: 39065055 PMCID: PMC11278641 DOI: 10.3390/microorganisms12071288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 07/28/2024] Open
Abstract
Domestic ducks (Anas platyrhynchos domesticus) are resistant to most of the highly pathogenic avian influenza virus (HPAIV) infections. In this study, we characterized the lung proteome and phosphoproteome of ducks infected with the HPAI H5N1 virus (A/duck/India/02CA10/2011/Agartala) at 12 h, 48 h, and 5 days post-infection. A total of 2082 proteins were differentially expressed and 320 phosphorylation sites mapping to 199 phosphopeptides, corresponding to 129 proteins were identified. The functional annotation of the proteome data analysis revealed the activation of the RIG-I-like receptor and Jak-STAT signaling pathways, which led to the induction of interferon-stimulated gene (ISG) expression. The pathway analysis of the phosphoproteome datasets also confirmed the activation of RIG-I, Jak-STAT signaling, NF-kappa B signaling, and MAPK signaling pathways in the lung tissues. The induction of ISG proteins (STAT1, STAT3, STAT5B, STAT6, IFIT5, and PKR) established a protective anti-viral immune response in duck lung tissue. Further, the protein-protein interaction network analysis identified proteins like AKT1, STAT3, JAK2, RAC1, STAT1, PTPN11, RPS27A, NFKB1, and MAPK1 as the main hub proteins that might play important roles in disease progression in ducks. Together, the functional annotation of the proteome and phosphoproteome datasets revealed the molecular basis of the disease progression and disease resistance mechanism in ducks infected with the HPAI H5N1 virus.
Collapse
Affiliation(s)
- Periyasamy Vijayakumar
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Salem 600051, Tamil Nadu, India
| | - Anamika Mishra
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| | - Ram Pratim Deka
- International Livestock Research Institute, National Agricultural Science Complex, Pusa 110012, New Delhi, India;
| | - Sneha M. Pinto
- Centre for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; (S.M.P.); (Y.S.)
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Yashwanth Subbannayya
- Centre for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; (S.M.P.); (Y.S.)
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Richa Sood
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| | | | - Ashwin Ashok Raut
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| |
Collapse
|
4
|
Liu Y, Zhang W, Jang H, Nussinov R. SHP2 clinical phenotype, cancer, or RASopathies, can be predicted by mutant conformational propensities. Cell Mol Life Sci 2023; 81:5. [PMID: 38085330 PMCID: PMC11072105 DOI: 10.1007/s00018-023-05052-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/20/2023] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
SHP2 phosphatase promotes full activation of the RTK-dependent Ras/MAPK pathway. Its mutations can drive cancer and RASopathies, a group of neurodevelopmental disorders (NDDs). Here we ask how same residue mutations in SHP2 can lead to both cancer and NDD phenotypes, and whether we can predict what the outcome will be. We collected and analyzed mutation data from the literature and cancer databases and performed molecular dynamics simulations of SHP2 mutants. We show that both cancer and Noonan syndrome (NS, a RASopathy) mutations favor catalysis-prone conformations. As to cancer versus RASopathies, we demonstrate that cancer mutations are more likely to accelerate SHP2 activation than the NS mutations at the same genomic loci, in line with NMR data for K-Ras4B more aggressive mutations. The compiled experimental data and dynamic features of SHP2 mutants lead us to propose that different from strong oncogenic mutations, SHP2 activation by NS mutations is less likely to induce a transition of the ensemble from the SHP2 inactive state to the active state. Strong signaling promotes cell proliferation, a hallmark of cancer. Weak, or moderate signals are associated with differentiation. In embryonic neural cells, dysregulated differentiation is connected to NDDs. Our innovative work offers structural guidelines for identifying and correlating mutations with clinical outcomes, and an explanation for why bearers of RASopathy mutations may have a higher probability of cancer. Finally, we propose a drug strategy against SHP2 variants-promoting cancer and RASopathies.
Collapse
Affiliation(s)
- Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
5
|
Zhao Q, Li Y, Shao Q, Zhang C, Kou S, Yang W, Zhang M, Ban B. Clinical and genetic evaluation of children with short stature of unknown origin. BMC Med Genomics 2023; 16:194. [PMID: 37605180 PMCID: PMC10441754 DOI: 10.1186/s12920-023-01626-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Short stature is a common human trait. More severe and/or associated short stature is usually part of the presentation of a syndrome and may be a monogenic disease. The present study aimed to identify the genetic etiology of children with short stature of unknown origin. METHODS A total of 232 children with short stature of unknown origin from March 2013 to May 2020 were enrolled in this study. Whole exome sequencing (WES) was performed for the enrolled patients to determine the underlying genetic etiology. RESULTS We identified pathogenic or likely pathogenic genetic variants in 18 (7.8%) patients. All of these variants were located in genes known to be associated with growth disorders. Five of the genes are associated with paracrine signaling or cartilage extracellular matrix in the growth plate, including NPR2 (N = 1), ACAN (N = 1), CASR (N = 1), COMP (N = 1) and FBN1 (N = 1). Two of the genes are involved in the RAS/MAPK pathway, namely, PTPN11 (N = 6) and NF1 (N = 1). Two genes are associated with the abnormal growth hormone-insulin-like growth factor 1 (GH-IGF1) axis, including GH1 (N = 1) and IGF1R (N = 1). Two mutations are located in PROKR2, which is associated with gonadotropin-releasing hormone deficiency. Mutations were found in the remaining two patients in genes with miscellaneous mechanisms: ANKRD11 (N = 1) and ARID1A (N = 1). CONCLUSIONS The present study identified pathogenic or likely pathogenic genetic variants in eighteen of the 232 patients (7.8%) with short stature of unknown origin. Our findings suggest that in the absence of prominent malformation, genetic defects in hormones, paracrine factors, and matrix molecules may be the causal factors for this group of patients. Early genetic testing is necessary for accurate diagnosis and precision treatment.
Collapse
Affiliation(s)
- Qianqian Zhao
- School of Medicine, Qingdao University, Qingdao, Shandong, 266071, P.R. China
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
| | - Yanying Li
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
| | - Qian Shao
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
| | - Chuanpeng Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
| | - Shuang Kou
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, 999077, P.R. China
| | - Mei Zhang
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China.
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China.
| | - Bo Ban
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China.
- Chinese Research Center for Behavior Medicine in Growth and Development, 89 Guhuai Road, Jining, Shandong, 272029, P.R. China.
| |
Collapse
|
6
|
Nakhleh Francis Y, Hershkovitz T, Ekhilevitch N, Habib C, Ravid S, Tal G, Schertz M, Mory A, Zinger A, Baris Feldman H, Zaid R, Paperna T, Weiss K. Publicly funded exome sequencing for outpatients with neurodevelopmental disorders demonstrates a high rate of unexpected findings impacting medical management. GENETICS IN MEDICINE OPEN 2023; 1:100828. [PMID: 39669259 PMCID: PMC11613680 DOI: 10.1016/j.gimo.2023.100828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 12/14/2024]
Abstract
Purpose Exome sequencing (ES) is a powerful tool that facilitates the diagnosis of patients with rare Mendelian syndromes. In 2018 the Israeli Ministry of Health initiated a national pilot program that funds ES for outpatients with global developmental delay (GDD). Here, we describe the 3-year impact of this program on patient care in a single tertiary hospital. Methods From 2018 to 2020, trio ES was performed on 170 participants fulfilling Israeli Ministry of Health criteria: (1) moderate to severe GDD and (2) mild GDD with epilepsy or a major congenital anomaly. We retrospectively analyzed this cohort. Results A diagnosis was achieved in 74 individuals (43%). There were 82 clinically significant variants, the majority being novel. Consanguinity was reported in 22% and was not associated with a higher diagnostic rate. The presence of autism spectrum was associated with a lower diagnostic rate of 8/33 (24%). Autosomal dominant inheritance was identified in 14% of participants, and the parental phenotype ranged between fully affected and asymptomatic. Among the diagnosed participants, 16% had an unexpected diagnosis that did not fit the typical clinical presentation. In 9%, the diagnosis changed short-term active clinical management, in 19%, the surveillance recommendations, and in 23%, the family-focused outcomes. Conclusion The introduction of a national program that funds ES for GDD has transformed patient care, leading to a significant effect on medical management and treatment. The high rate of an unexpected inheritance mode and variable phenotypes emphasizes the diagnostic complexity of neurodevelopmental disorders and the strength of a non-targeted approach.
Collapse
Affiliation(s)
- Yara Nakhleh Francis
- Department of obstetrics and gynecology, Galilee medical center, Nahariya, Israel
- The Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Tova Hershkovitz
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nina Ekhilevitch
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Clair Habib
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Sarit Ravid
- Pediatric neurology unit, Rambam Health Care Campus, Haifa, Israel
| | - Galit Tal
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Metabolic Clinic, Ruth Rappaport Children’s Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Mitchell Schertz
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Child Development and Pediatric Neurology Service, Meuhedet, Tel Aviv, Israel
| | - Adi Mory
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- Current address: The Genetics Institute, Tel Aviv Sourasky Medical Center Tel Aviv, Israel
| | - Amihood Zinger
- Community Genetics, Public Health Services, Ministry of Health, Jerusalem, Israel
| | - Hagit Baris Feldman
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- Current address: The Genetics Institute, Tel Aviv Sourasky Medical Center Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rinat Zaid
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Tamar Paperna
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Karin Weiss
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
7
|
Borowczyk M, Dobosz P, Szczepanek-Parulska E, Budny B, Dębicki S, Filipowicz D, Wrotkowska E, Oszywa M, Verburg FA, Janicka-Jedyńska M, Ziemnicka K, Ruchała M. Follicular Thyroid Adenoma and Follicular Thyroid Carcinoma-A Common or Distinct Background? Loss of Heterozygosity in Comprehensive Microarray Study. Cancers (Basel) 2023; 15:638. [PMID: 36765597 PMCID: PMC9913827 DOI: 10.3390/cancers15030638] [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: 12/04/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Pre- and postsurgical differentiation between follicular thyroid adenoma (FTA) and follicular thyroid cancer (FTC) represents a significant diagnostic challenge. Furthermore, it remains unclear whether they share a common or distinct background and what the mechanisms underlying follicular thyroid lesions malignancy are. The study aimed to compare FTA and FTC by the comprehensive microarray and to identify recurrent regions of loss of heterozygosity (LOH). We analyzed formalin-fixed paraffin-embedded (FFPE) samples acquired from 32 Caucasian patients diagnosed with FTA (16) and FTC (16). We used the OncoScan™ microarray assay (Affymetrix, USA), using highly multiplexed molecular inversion probes for single nucleotide polymorphism (SNP). The total number of LOH was higher in FTC compared with FTA (18 vs. 15). The most common LOH present in 21 cases, in both FTA (10 cases) and FTC (11 cases), was 16p12.1, which encompasses many cancer-related genes, such as TP53, and was followed by 3p21.31. The only LOH present exclusively in FTA patients (56% vs. 0%) was 11p11.2-p11.12. The alteration which tended to be detected more often in FTC (6 vs. 1 in FTA) was 12q24.11-q24.13 overlapping FOXN4, MYL2, PTPN11 genes. FTA and FTC may share a common genetic background, even though differentiating rearrangements may also be detected.
Collapse
Affiliation(s)
- Martyna Borowczyk
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
- Department of Medical Simulation, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Paula Dobosz
- Department of Genetics and Genomics, Central Clinical Hospital of the Ministry of Interior Affairs and Administration, 02-507 Warsaw, Poland
| | - Ewelina Szczepanek-Parulska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Szymon Dębicki
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Dorota Filipowicz
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Michalina Oszywa
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Frederik A. Verburg
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
8
|
Idiopathic Short Stature: What to Expect from Genomic Investigations. ENDOCRINES 2023. [DOI: 10.3390/endocrines4010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Short stature is a common concern for physicians caring for children. In traditional investigations, about 70% of children are healthy, without producing clinical and laboratory findings that justify their growth disorder, being classified as having constitutional short stature or idiopathic short stature (ISS). In such scenarios, the genetic approach has emerged as a great potential method to understand ISS. Over the last 30 years, several genes have been identified as being responsible for isolated short stature, with almost all of them being inherited in an autosomal-dominant pattern. Most of these defects are in genes related to the growth plate, followed by genes related to the growth hormone (GH)–insulin-like growth factor 1 (IGF1) axis and RAS-MAPK pathway. These patients usually do not have a specific phenotype, which hinders the use of a candidate gene approach. Through multigene sequencing analyses, it has been possible to provide an answer for short stature in 10–30% of these cases, with great impacts on treatment and follow-up, allowing the application of the concept of precision medicine in patients with ISS. This review highlights the historic aspects and provides an update on the monogenic causes of idiopathic short stature and suggests what to expect from genomic investigations in this field.
Collapse
|
9
|
Eboreime J, Choi SK, Yoon SR, Sadybekov A, Katritch V, Calabrese P, Arnheim N. Germline selection of PTPN11 (HGNC:9644) variants make a major contribution to both Noonan syndrome's high birth rate and the transmission of sporadic cancer variants resulting in fetal abnormality. Hum Mutat 2022; 43:2205-2221. [PMID: 36349709 PMCID: PMC10099774 DOI: 10.1002/humu.24493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/20/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022]
Abstract
Some spontaneous germline gain-of-function mutations promote spermatogonial stem cell clonal expansion and disproportionate variant sperm production leading to unexpectedly high transmission rates for some human genetic conditions. To measure the frequency and spatial distribution of de novo mutations we divided three testes into 192 pieces each and used error-corrected deep-sequencing on each piece. We focused on PTPN11 (HGNC:9644) Exon 3 that contains 30 different PTPN11 Noonan syndrome (NS) mutation sites. We found 14 of these variants formed clusters among the testes; one testis had 11 different variant clusters. The mutation frequencies of these different clusters were not correlated with their case-recurrence rates nor were case recurrence rates of PTPN11 variants correlated with their tyrosine phosphatase levels thereby confusing PTPN11's role in germline clonal expansion. Six of the PTPN11 exon 3 de novo variants associated with somatic mutation-induced sporadic cancers (but not NS) also formed testis clusters. Further, three of these six variants were observed among fetuses that underwent prenatal ultrasound screening for NS-like features. Mathematical modeling showed that germline selection can explain both the mutation clusters and the high incidence of NS (1/1000-1/2500).
Collapse
Affiliation(s)
- Jordan Eboreime
- Department of Biological Sciences, Molecular and Computational Biology Program, University of Southern California, Los Angeles, California, USA
| | - Soo-Kyung Choi
- Department of Biological Sciences, Molecular and Computational Biology Program, University of Southern California, Los Angeles, California, USA
| | - Song-Ro Yoon
- Department of Biological Sciences, Molecular and Computational Biology Program, University of Southern California, Los Angeles, California, USA
| | - Anastasiia Sadybekov
- Department of Chemistry, Bridge Institute, University of Southern California, Los Angeles, California, USA
| | - Vsevolod Katritch
- Department of Chemistry, Bridge Institute, University of Southern California, Los Angeles, California, USA
| | - Peter Calabrese
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA
| | - Norman Arnheim
- Department of Biological Sciences, Molecular and Computational Biology Program, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
10
|
Gao Y, Wu D, Chen B, Chen Y, Zhang Q, Zhao P. Rare Variants in Novel Candidate Genes Associated With Nonsyndromic Patent Ductus Arteriosus Identified With Whole-Exome Sequencing. Front Genet 2022; 13:921925. [PMID: 35734438 PMCID: PMC9207465 DOI: 10.3389/fgene.2022.921925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Patent ductus arteriosus (PDA) is one of the most common congenital heart defects causing pulmonary hypertension, infective endocarditis, and even death. The important role of genetics in determining spontaneous ductal closure has been well-established. However, as many of the identified variants are rare, thorough identification of the associated genetic factors is necessary to further explore the genetic etiology of PDA. Methods: We performed whole-exome sequencing (WES) on 39 isolated nonsyndromic PDA patients and 100 healthy controls. Rare variants and novel genes were identified through bioinformatic filtering strategies. The expression patterns of candidate genes were explored in human embryo heart samples. Results: Eighteen rare damaging variants of six novel PDA-associated genes (SOX8, NES, CDH2, ANK3, EIF4G1, and HIPK1) were newly identified, which were highly expressed in human embryo hearts. Conclusions: WES is an efficient diagnostic tool for exploring the genetic pathogenesis of PDA. These findings contribute new insights into the molecular basis of PDA and may inform further studies on genetic risk factors for congenital heart defects.
Collapse
Affiliation(s)
- Ying Gao
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Dan Wu
- Department of Pediatric, Shidong Hospital, Shanghai, China
| | - Bo Chen
- Department of Cardiothoracic Surgery, School of Medicine, Heart Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengjun Zhao
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Pengjun Zhao,
| |
Collapse
|
11
|
Calligari P, Santucci V, Stella L, Bocchinfuso G. Discriminating between competing models for the allosteric regulation of oncogenic phosphatase SHP2 by characterizing its active state. Comput Struct Biotechnol J 2021; 19:6125-6139. [PMID: 34900129 PMCID: PMC8632847 DOI: 10.1016/j.csbj.2021.10.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 11/07/2022] Open
Abstract
The Src-homology 2 domain containing phosphatase 2 (SHP2) plays a critical role in crucial signaling pathways and is involved in oncogenesis and in developmental disorders. Its structure includes two SH2 domains (N-SH2 and C-SH2), and a protein tyrosine phosphatase (PTP) domain. Under basal conditions, SHP2 is auto-inhibited, with the N-SH2 domain blocking the PTP active site. Activation involves a rearrangement of the domains that makes the catalytic site accessible, coupled to the association between the SH2 domains and cognate proteins containing phosphotyrosines. Several aspects of this transition are debated and competing mechanistic models have been proposed. A crystallographic structure of SHP2 in an active state has been reported (PDB code 6crf), but several lines of evidence suggests that it is not fully representative of the conformations populated in solution. To clarify the structural rearrangements involved in SHP2 activation, enhanced sampling simulations of the autoinhibited and active states have been performed, for wild type SHP2 and its pathogenic E76K variant. Our results demonstrate that the crystallographic conformation of the active state is unstable in solution, and multiple interdomain arrangements are populated, thus allowing association to bisphosphorylated sequences. Contrary to a recent proposal, activation is coupled to the conformational changes of the N-SH2 binding site, which is significantly more accessible in the active sate, rather than to the structure of the central β-sheet of the domain. In this coupling, a previously undescribed role for the N-SH2 BG loop emerged.
Collapse
Key Words
- BTLA, B and T lymphocyte attenuator
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- FRET, Förster resonance energy transfer
- Inter-domain dynamics
- JMML, juvenile myelomonocytic leukemia
- MD, molecular dynamics
- NS, Noonan syndrome
- NSML, Noonan syndrome with multiple lentigines
- PD-1, programmed cell death protein 1
- PDB, protein data bank
- PMF, potential of mean force
- PTP, protein tyrosine phosphatase
- Protein flexibility
- REMD, replica exchange molecular dynamics
- RMSD, root mean square deviation
- RMSF, root mean square fluctuation
- RTK, receptor tyrosine kinase
- Replica exchange molecular dynamics simulations
- SASA, solvent accessible surface area
- SAXS, small angle X-ray scattering
- SH2, Src homology 2
- SHP2 regulatory mechanism
- SHP2, Src homology 2 domain-containing phosphatase 2
- SIRPalpha, signal regulatory protein alpha
- pY, phosphorylated tyrosine
Collapse
Affiliation(s)
- Paolo Calligari
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Valerio Santucci
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| |
Collapse
|
12
|
D'Amico A, Rosano C, Pannone L, Pinna V, Assunto A, Motta M, Ugga L, Daniele P, Mandile R, Mariniello L, Siano MA, Santoro C, Piluso G, Martinelli S, Strisciuglio P, De Luca A, Tartaglia M, Melis D. Clinical variability of neurofibromatosis 1: A modifying role of cooccurring PTPN11 variants and atypical brain MRI findings. Clin Genet 2021; 100:563-572. [PMID: 34346503 DOI: 10.1111/cge.14040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2023]
Abstract
Neurofibromatosis 1 (NF1) is a disorder characterized by variable expressivity caused by loss-of-function variants in NF1, encoding neurofibromin, a protein negatively controlling RAS signaling. We evaluated whether concurrent variation in proteins functionally linked to neurofibromin contribute to the variable expressivity of NF1. Parallel sequencing of a RASopathy gene panel in 138 individuals with molecularly confirmed clinical diagnosis of NF1 identified missense variants in PTPN11, encoding SHP2, a positive regulator of RAS signaling, in four subjects from three unrelated families. Three subjects were heterozygous for a gain-of-function variant and showed a severe expression of NF1 (developmental delay, multiple cerebral neoplasms and peculiar cortical MRI findings), and features resembling Noonan syndrome (a RASopathy caused by activating variants in PTPN11). Conversely, the fourth subject, who showed an attenuated presentation, carried a previously unreported PTPN11 variant that had a hypomorphic behavior in vitro. Our findings document that functionally relevant PTPN11 variants occur in a small but significant proportion of subjects with NF1 modulating disease presentation, suggesting a model in which the clinical expression of pathogenic NF1 variants is modified by concomitant dysregulation of protein(s) functionally linked to neurofibromin. We also suggest targeting of SHP2 function as an approach to treat evolutive complications of NF1.
Collapse
Affiliation(s)
- Alessandra D'Amico
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Carmen Rosano
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Luca Pannone
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Valentina Pinna
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Antonia Assunto
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Marialetizia Motta
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Paola Daniele
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Roberta Mandile
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Lucio Mariniello
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Maria Anna Siano
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| | - Claudia Santoro
- Referral Centre of Neurofibromatosis, Department of Woman and Child, Specialistic and General Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Pietro Strisciuglio
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Alessandro De Luca
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Daniela Melis
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy.,Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| |
Collapse
|
13
|
Menon S, Salhotra A, Shailendra S, Tevlin R, Ransom RC, Januszyk M, Chan CKF, Behr B, Wan DC, Longaker MT, Quarto N. Skeletal stem and progenitor cells maintain cranial suture patency and prevent craniosynostosis. Nat Commun 2021; 12:4640. [PMID: 34330896 PMCID: PMC8324898 DOI: 10.1038/s41467-021-24801-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
Cranial sutures are major growth centers for the calvarial vault, and their premature fusion leads to a pathologic condition called craniosynostosis. This study investigates whether skeletal stem/progenitor cells are resident in the cranial sutures. Prospective isolation by FACS identifies this population with a significant difference in spatio-temporal representation between fusing versus patent sutures. Transcriptomic analysis highlights a distinct signature in cells derived from the physiological closing PF suture, and scRNA sequencing identifies transcriptional heterogeneity among sutures. Wnt-signaling activation increases skeletal stem/progenitor cells in sutures, whereas its inhibition decreases. Crossing Axin2LacZ/+ mouse, endowing enhanced Wnt activation, to a Twist1+/- mouse model of coronal craniosynostosis enriches skeletal stem/progenitor cells in sutures restoring patency. Co-transplantation of these cells with Wnt3a prevents resynostosis following suturectomy in Twist1+/- mice. Our study reveals that decrease and/or imbalance of skeletal stem/progenitor cells representation within sutures may underlie craniosynostosis. These findings have translational implications toward therapeutic approaches for craniosynostosis.
Collapse
Affiliation(s)
- Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ankit Salhotra
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruth Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles K F Chan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Björn Behr
- Department of Plastic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Napoli, Italy.
| |
Collapse
|
14
|
Kawashima I, Oishi N, Kasai K, Inoue T, Hosokawa E, Nakadate A, Matsuura M, Kumagai T, Koshiishi M, Yamamoto T, Nakajima K, Tanaka M, Kondo T, Kirito K. Transdifferentiation of mantle cell lymphoma into sarcoma with limited neuromuscular differentiation after conventional chemotherapy. Virchows Arch 2021; 480:1101-1105. [PMID: 34226971 DOI: 10.1007/s00428-021-03148-9] [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: 04/21/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 01/26/2023]
Abstract
We report an exceptionally rare case of mantle cell lymphoma (MCL) that transdifferentiated into sarcoma with limited neuromuscular differentiation. An 81-year-old man with t(11;14)-positive MCL was treated with rituximab and bendamustine and achieved complete remission; however, just 2 months later, the patient developed multiple systemic tumors. Pathologic studies revealed round cell sarcoma expressing synaptophysin, CD56, and myogenin without any B-cell markers. The CCND1 translocation and an identical IGL gene rearrangement were shared by both the MCL and sarcoma. Whole-exome sequencing detected 189 single nucleotide variants (SNVs) in the MCL and 205 SNVs in the sarcoma; 160 SNVs including NSD2, ATM, RB1, and TP53 mutations were shared between MCL and sarcoma cells. An additional PTPN11 mutation was specifically found in the sarcoma. These findings confirmed the shared clonal origin of MCL and sarcoma in this patient and indicated that MCL can transdifferentiate into sarcoma in rare cases.
Collapse
Affiliation(s)
- Ichiro Kawashima
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Naoki Oishi
- Department of Pathology, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kazunari Kasai
- Department of Pathology, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Tomohiro Inoue
- Department of Pathology, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Eriko Hosokawa
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Ayato Nakadate
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Minori Matsuura
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Takuma Kumagai
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Megumi Koshiishi
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Takeo Yamamoto
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Kei Nakajima
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Masaru Tanaka
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Tetsuo Kondo
- Department of Pathology, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keita Kirito
- Department of Hematology and Oncology, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
| |
Collapse
|
15
|
Montero-Bullón JF, González-Velasco Ó, Isidoro-García M, Lacal J. Integrated in silico MS-based phosphoproteomics and network enrichment analysis of RASopathy proteins. Orphanet J Rare Dis 2021; 16:303. [PMID: 34229750 PMCID: PMC8258961 DOI: 10.1186/s13023-021-01934-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 06/27/2021] [Indexed: 11/30/2022] Open
Abstract
Background RASopathies are a group of syndromes showing clinical overlap caused by mutations in genes affecting the RAS-MAPK pathway. Consequent disruption on cellular signaling leads and is driven by phosphoproteome remodeling. However, we still lack a comprehensive picture of the different key players and altered downstream effectors. Methods An in silico interactome of RASopathy proteins was generated using pathway enrichment analysis/STRING tool, including identification of main hub proteins. We also integrated phosphoproteomic and immunoblotting studies using previous published information on RASopathy proteins and their neighbors in the context of RASopathy syndromes. Data from Phosphosite database (www.phosphosite.org) was collected in order to obtain the potential phosphosites subjected to regulation in the 27 causative RASopathy proteins. We compiled a dataset of dysregulated phosphosites in RASopathies, searched for commonalities between syndromes in harmonized data, and analyzed the role of phosphorylation in the syndromes by the identification of key players between the causative RASopathy proteins and the associated interactome. Results In this study, we provide a curated data set of 27 causative RASopathy genes, identify up to 511 protein–protein associations using pathway enrichment analysis/STRING tool, and identify 12 nodes as main hub proteins. We found that a large group of proteins contain tyrosine residues and their biological processes include but are not limited to the nervous system. Harmonizing published RASopathy phosphoproteomic and immunoblotting studies we identified a total of 147 phosphosites with increased phosphorylation, whereas 47 have reduced phosphorylation. The PKB signaling pathway is the most represented among the dysregulated phosphoproteins within the RASopathy proteins and their neighbors, followed by phosphoproteins implicated in the regulation of cell proliferation and the MAPK pathway. Conclusions This work illustrates the complex network underlying the RASopathies and the potential of phosphoproteomics for dissecting the molecular mechanisms in these syndromes. A combined study of associated genes, their interactome and phosphorylation events in RASopathies, elucidates key players and mechanisms to direct future research, diagnosis and therapeutic windows. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01934-x.
Collapse
Affiliation(s)
- Javier-Fernando Montero-Bullón
- Metabolic Engineering Group, Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007, Salamanca, Spain
| | - Óscar González-Velasco
- Bioinformatics and Functional Genomics Group, IBMCC Cancer Research Center, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - María Isidoro-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.,Network for Cooperative Research in Health-RETICS ARADyAL, 37007, Salamanca, Spain.,Department of Clinical Biochemistry, University Hospital of Salamanca, 37007, Salamanca, Spain.,Department of Medicine, University of Salamanca, 37007, Salamanca, Spain
| | - Jesus Lacal
- Institute for Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain. .,Molecular Genetics of Human Diseases Group, Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
16
|
Guo Y, Xu Y, Dong X, Zhang J. Cross the Undruggable Barrier, the Development of SHP2 Inhibitors: From Catalytic Site Inhibitors to Allosteric Inhibitors. ChemistrySelect 2021. [DOI: 10.1002/slct.202100186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yu Guo
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Yaping Xu
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Jianjun Zhang
- Department of Pharmacy Institution The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine) Hangzhou 310006 P.R. China
| |
Collapse
|
17
|
Dong L, Han D, Meng X, Xu M, Zheng C, Xia Q. Activating Mutation of SHP2 Establishes a Tumorigenic Phonotype Through Cell-Autonomous and Non-Cell-Autonomous Mechanisms. Front Cell Dev Biol 2021; 9:630712. [PMID: 33777940 PMCID: PMC7991796 DOI: 10.3389/fcell.2021.630712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 01/18/2023] Open
Abstract
Gain-of-function mutation of SHP2 is a central regulator in tumorigenesis and cancer progression through cell-autonomous mechanisms. Activating mutation of SHP2 in microenvironment was identified to promote cancerous transformation of hematopoietic stem cell in non-autonomous mechanisms. It is interesting to see whether therapies directed against SHP2 in tumor or microenvironmental cells augment antitumor efficacy. In this review, we summarized different types of gain-of-function SHP2 mutations from a human disease. In general, gain-of-function mutations destroy the auto-inhibition state from wild-type SHP2, leading to consistency activation of SHP2. We illustrated how somatic or germline mutation of SHP2 plays an oncogenic role in tumorigenesis, stemness maintenance, invasion, etc. Moreover, the small-molecule SHP2 inhibitors are considered as a potential strategy for enhancing the efficacy of antitumor immunotherapy and chemotherapy. We also discussed the interconnection between phase separation and activating mutation of SHP2 in drug resistance of antitumor therapy.
Collapse
Affiliation(s)
- Lei Dong
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Da Han
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Xinyi Meng
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Mengchuan Xu
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Chuwen Zheng
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Qin Xia
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
18
|
Targeting SHP2 as a therapeutic strategy for inflammatory diseases. Eur J Med Chem 2021; 214:113264. [PMID: 33582386 DOI: 10.1016/j.ejmech.2021.113264] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/19/2022]
Abstract
With the change of lifestyle and the acceleration of aging process, inflammatory diseases have increasingly become one of the most vital threats to global human health. SHP2 protein is a non-receptor tyrosine phosphatase encoded by PTPN11 gene, and it is widely expressed in various tissues and cells. Numerous studies have shown that SHP2 plays important roles in the regulation of inflammatory diseases, including cancer-related inflammation, neurodegenerative diseases and metabolic diseases. In this paper, the roles of SHP2 in inflammatory diseases of various physiological systems were reviewed. At the same time, the latest SHP2 inhibitors were summarized, which will hold a promise for the therapeutic potential in future.
Collapse
|
19
|
Lorca R, Pannone L, Cuesta-Llavona E, Bocchinfuso G, Rodríguez-Reguero J, Carpentieri G, Hernando I, Flex E, Tartaglia M, Coto E, Gómez J, Martinelli S. Compound heterozygosity for PTPN11 variants in a subject with Noonan syndrome provides insights into the mechanism of SHP2-related disorders. Clin Genet 2021; 99:457-461. [PMID: 33354767 DOI: 10.1111/cge.13904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/26/2020] [Accepted: 12/19/2020] [Indexed: 12/31/2022]
Abstract
The RASopathies are a family of clinically related disorders caused by mutations affecting genes participating in the RAS-MAPK signaling cascade. Among them, Noonan syndrome (NS) and Noonan syndrome with multiple lentigines (NSML) are allelic conditions principally associated with dominant mutations in PTPN11, which encodes the nonreceptor SH2 domain-containing protein tyrosine phosphatase SHP2. Individual PTPN11 mutations are specific to each syndrome and have opposite consequences on catalysis, but all favor SHP2's interaction with signaling partners. Here, we report on a subject with NS harboring biallelic variants in PTPN11. While the former (p.Leu261Phe) had previously been reported in NS, the latter (p.Thr357Met) is a novel change impairing catalysis. Members of the family carrying p.Thr357Met, however, did not show any obvious feature fitting NSML or within the RASopathy phenotypic spectrum. A major impact of this change on transcript processing and protein stability was excluded. These findings further support the view that NSML cannot be ascribed merely to impaired SHP2's catalytic activity and suggest that PTPN11 mutations causing this condition act through an alternative dominant mechanism.
Collapse
Affiliation(s)
- Rebeca Lorca
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Luca Pannone
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Elías Cuesta-Llavona
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Grupo Eje Cardio-Renal, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Julian Rodríguez-Reguero
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Grupo Eje Cardio-Renal, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Giovanna Carpentieri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Inés Hernando
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Eliecer Coto
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Grupo Eje Cardio-Renal, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Medicine Department, Universidad de Oviedo, Oviedo, Spain.,HUCA. Eje Cardio-Renal, Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Juan Gómez
- Unidad de Referencia de Cardiopatías Familiares-HUCA, Hospital Universitario Central de Asturias (HUCA) - Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Grupo Eje Cardio-Renal, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,HUCA. Eje Cardio-Renal, Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
20
|
Malaquias AC, Jorge AAL. Activation of the MAPK pathway (RASopathies) and partial growth hormone insensitivity. Mol Cell Endocrinol 2021; 519:111040. [PMID: 33011209 DOI: 10.1016/j.mce.2020.111040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
RASopathies are a heterogeneous group of syndromes caused by germline mutations in genes encoding components of the RAS/MAPK pathway. Postnatal short stature is a cardinal feature of the RASopathies. Although the pathophysiology of these conditions is not fully understood to date, growth hormone insensitivity is one possibility, based on the observation of low IGF-1 values, generally preserved GH secretion and suboptimal growth response to recombinant human GH therapy. In this review, we will discuss the clinical and experimental evidence of GH insensitivity in patients with Noonan syndrome and other RASopathies, as well as their molecular basis.
Collapse
Affiliation(s)
- Alexsandra C Malaquias
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Unidade de Endocrinologia Pediátrica, Departamento de Pediatria, Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
21
|
Lee TL, Lin PH, Chen PL, Hong JB, Wu CC. Hereditary Hearing Impairment with Cutaneous Abnormalities. Genes (Basel) 2020; 12:43. [PMID: 33396879 PMCID: PMC7823799 DOI: 10.3390/genes12010043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/25/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Syndromic hereditary hearing impairment (HHI) is a clinically and etiologically diverse condition that has a profound influence on affected individuals and their families. As cutaneous findings are more apparent than hearing-related symptoms to clinicians and, more importantly, to caregivers of affected infants and young individuals, establishing a correlation map of skin manifestations and their underlying genetic causes is key to early identification and diagnosis of syndromic HHI. In this article, we performed a comprehensive PubMed database search on syndromic HHI with cutaneous abnormalities, and reviewed a total of 260 relevant publications. Our in-depth analyses revealed that the cutaneous manifestations associated with HHI could be classified into three categories: pigment, hyperkeratosis/nail, and connective tissue disorders, with each category involving distinct molecular pathogenesis mechanisms. This outline could help clinicians and researchers build a clear atlas regarding the phenotypic features and pathogenetic mechanisms of syndromic HHI with cutaneous abnormalities, and facilitate clinical and molecular diagnoses of these conditions.
Collapse
Affiliation(s)
- Tung-Lin Lee
- Department of Medical Education, National Taiwan University Hospital, Taipei City 100, Taiwan;
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 11556, Taiwan;
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei City 100, Taiwan;
| | - Pei-Lung Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei City 100, Taiwan;
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei City 100, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10041, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10041, Taiwan
| | - Jin-Bon Hong
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei City 100, Taiwan
- Department of Dermatology, National Taiwan University Hospital, Taipei City 100, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 11556, Taiwan;
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei City 100, Taiwan;
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10041, Taiwan
- Department of Medical Research, National Taiwan University Biomedical Park Hospital, Hsinchu City 300, Taiwan
| |
Collapse
|
22
|
Exploring the mechanism of the potent allosteric inhibitor compound2 on SHP2 WT and SHP2 F285S by molecular dynamics study. J Mol Graph Model 2020; 103:107807. [PMID: 33338846 DOI: 10.1016/j.jmgm.2020.107807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/23/2022]
Abstract
Abnormal activation of Ras/MAPK signaling pathway could trigger excessive cell division. Src-homology 2 (SH2) domain-containing protein tyrosine phosphatase (SHP2) could promote Ras/MAPK activation by integrating growth factor signals. Thus, SHP2 inhibitors had become a hot topic in the treatment of cancer. SHP2F285S, mutation in SHP2, was detected in leukemia variants. The compound 2 (3-benzyl-8-chloro-2-hydroxy-4H-benzo[4,5]thiazolo[3,2-a]pyrimidin-4-one) had been reported that it was a potent allosteric inhibitor of both SHP2 wild type (SHP2WT) and the F285S mutant (SHP2F285S). However, the mechanism of inhibition remained to be further discovered. Herein, molecular docking and molecular dynamic (MD) simulation were performed to explain the inhibition mechanism of compound 2 on SHP2WT and SHP2F285S. Overall, the molecular docking analysis revealed that compound 2 maintained the "close" structure of SHP2 protein probably by locking the C-SH2 and PTP domain. Next, post-analysis demonstrated that compound 2 might make TYR66-GLU76 of D'E-loop in N-SH2 and GLU258-LYS266 of B'C-loop, HIS458-ARG465 of P-loop, VAL497-THR507 of Q-loop in PTP domain regions tightly connect and much easier maintain "self-inhibited" conformation of SHP2F285S-compound2 than that of SHP2WT-compound2. Importantly, GLU76 of D'E-loop could play a vital role in inhibition of SHP2WT-compound2 and SHP2F285S-compound2. This work provided a reliable clue to develop novel inhibitors for leukemia related to SHP2F285S.
Collapse
|
23
|
Proteome activity landscapes of tumor cell lines determine drug responses. Nat Commun 2020; 11:3639. [PMID: 32686665 PMCID: PMC7371697 DOI: 10.1038/s41467-020-17336-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 06/22/2020] [Indexed: 01/02/2023] Open
Abstract
Integrated analysis of genomes, transcriptomes, proteomes and drug responses of cancer cell lines (CCLs) is an emerging approach to uncover molecular mechanisms of drug action. We extend this paradigm to measuring proteome activity landscapes by acquiring and integrating quantitative data for 10,000 proteins and 55,000 phosphorylation sites (p-sites) from 125 CCLs. These data are used to contextualize proteins and p-sites and predict drug sensitivity. For example, we find that Progesterone Receptor (PGR) phosphorylation is associated with sensitivity to drugs modulating estrogen signaling such as Raloxifene. We also demonstrate that Adenylate kinase isoenzyme 1 (AK1) inactivates antimetabolites like Cytarabine. Consequently, high AK1 levels correlate with poor survival of Cytarabine-treated acute myeloid leukemia patients, qualifying AK1 as a patient stratification marker and possibly as a drug target. We provide an interactive web application termed ATLANTiC (http://atlantic.proteomics.wzw.tum.de), which enables the community to explore the thousands of novel functional associations generated by this work.
Collapse
|
24
|
Ho D, Quake SR, McCabe ERB, Chng WJ, Chow EK, Ding X, Gelb BD, Ginsburg GS, Hassenstab J, Ho CM, Mobley WC, Nolan GP, Rosen ST, Tan P, Yen Y, Zarrinpar A. Enabling Technologies for Personalized and Precision Medicine. Trends Biotechnol 2020; 38:497-518. [PMID: 31980301 PMCID: PMC7924935 DOI: 10.1016/j.tibtech.2019.12.021] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Individualizing patient treatment is a core objective of the medical field. Reaching this objective has been elusive owing to the complex set of factors contributing to both disease and health; many factors, from genes to proteins, remain unknown in their role in human physiology. Accurately diagnosing, monitoring, and treating disorders requires advances in biomarker discovery, the subsequent development of accurate signatures that correspond with dynamic disease states, as well as therapeutic interventions that can be continuously optimized and modulated for dose and drug selection. This work highlights key breakthroughs in the development of enabling technologies that further the goal of personalized and precision medicine, and remaining challenges that, when addressed, may forge unprecedented capabilities in realizing truly individualized patient care.
Collapse
Affiliation(s)
- Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore; The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore; Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, CA, USA; Department of Applied Physics, Stanford University, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Wee Joo Chng
- Department of Haematology and Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Edward K Chow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xianting Ding
- Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, NC, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis, MO, USA; Psychological & Brain Sciences, Washington University in St. Louis, MO, USA
| | - Chih-Ming Ho
- Department of Mechanical Engineering, University of California, Los Angeles, CA, USA
| | - William C Mobley
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University, CA, USA
| | - Steven T Rosen
- Comprehensive Cancer Center and Beckman Research Institute, City of Hope, CA, USA
| | - Patrick Tan
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yun Yen
- College of Medical Technology, Center of Cancer Translational Research, Taipei Cancer Center of Taipei Medical University, Taipei, Taiwan
| | - Ali Zarrinpar
- Department of Surgery, Division of Transplantation & Hepatobiliary Surgery, University of Florida, FL, USA
| |
Collapse
|
25
|
Ranza E, Guimier A, Verloes A, Capri Y, Marques C, Auclair M, Mathieu-Dramard M, Morin G, Thevenon J, Faivre L, Thauvin-Robinet C, Innes AM, Dyment DA, Vigouroux C, Amiel J. Overlapping phenotypes between SHORT and Noonan syndromes in patients with PTPN11 pathogenic variants. Clin Genet 2020; 98:10-18. [PMID: 32233106 DOI: 10.1111/cge.13746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
Overlapping syndromes such as Noonan, Cardio-Facio-Cutaneous, Noonan syndrome (NS) with multiple lentigines and Costello syndromes are genetically heterogeneous conditions sharing a dysregulation of the RAS/mitogen-activated protein kinase (MAPK) pathway and are known collectively as the RASopathies. PTPN11 was the first disease-causing gene identified in NS and remains the more prevalent. We report seven patients from three families presenting heterozygous missense variants in PTPN11 probably responsible for a disease phenotype distinct from the classical Noonan syndrome. The clinical presentation and common features of these seven cases overlap with the SHORT syndrome. The latter is the consequence of PI3K/AKT signaling deregulation with the predominant disease-causing gene being PIK3R1. Our data suggest that the phenotypic spectrum associated with pathogenic variants of PTPN11 could be wider than previously described, and this could be due to the dual activity of SHP2 (ie, PTPN11 gene product) on the RAS/MAPK and PI3K/AKT signaling.
Collapse
Affiliation(s)
- Emmanuelle Ranza
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Anne Guimier
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Alain Verloes
- Department of Genetics, APHP- Robert Debré University Hospital & INSERM UMR1141, Paris, France
| | - Yline Capri
- Department of Genetics, APHP- Robert Debré University Hospital & INSERM UMR1141, Paris, France
| | - Charles Marques
- Faculdade de Medicina, Centro Universitario Estacio, Ribeirao Preto, São Paulo, Brazil
| | - Martine Auclair
- Centre de Recherche Saint-Antoine, et Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM UMR_S 938, Paris, France
| | - Michèle Mathieu-Dramard
- Service de Génétique Clinique, Centre de référence maladies rares, CHU d'Amiens-site Sud, Amiens, France
| | - Gilles Morin
- Service de Génétique Clinique, Centre de référence maladies rares, CHU d'Amiens-site Sud, Amiens, France
| | - Julien Thevenon
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - Laurence Faivre
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Centre de Référence maladies rares « Anomalies du Développement et syndrome malformatifs » de l'Est et Centre de Génétique, FHU TRANSLAD, Hôpital d'Enfants, CHU, Dijon, France.,Equipe d'Accueil 4271, Génétique des Anomalies du Développement, FHU TRANSLAD, Université de Bourgogne, Dijon, France
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David A Dyment
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Corinne Vigouroux
- Centre de Recherche Saint-Antoine, et Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM UMR_S 938, Paris, France.,AP-HP, Hôpital Saint-Antoine, Centre de Référence des Pathologies Rares de l'Insulino-Sécrétion et de l'Insulino-Sensibilité (PRISIS), Service d'Endocrinologie, Diabétologie et Endocrinologie de la Reproduction, and Laboratoire Commun de Biologie et Génétique Moléculaires, Paris, France
| | - Jeanne Amiel
- Service de Génétique, Hôpital Necker-Enfants Malades, Paris, France.,Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
26
|
Athota JP, Bhat M, Nampoothiri S, Gowrishankar K, Narayanachar SG, Puttamallesh V, Farooque MO, Shetty S. Molecular and clinical studies in 107 Noonan syndrome affected individuals with PTPN11 mutations. BMC MEDICAL GENETICS 2020; 21:50. [PMID: 32164556 PMCID: PMC7068896 DOI: 10.1186/s12881-020-0986-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/25/2020] [Indexed: 02/08/2023]
Abstract
Background Noonan syndrome (NS), an autosomal dominant developmental genetic disorder, is caused by germline mutations in genes associated with the RAS / mitogen-activated protein kinase (MAPK) pathway. In several studies PTPN11 is one of the genes with a significant number of pathogenic variants in NS-affected patients. Therefore, clinically diagnosed NS individuals are initially tested for pathogenic variants in PTPN11 gene to confirm the relationship before studying genotype–phenotype correlation. Methods Individuals (363) with clinically diagnosed NS from four hospitals in South India were recruited and the exons of PTPN11 gene were sequenced. Results Thirty-two previously described pathogenic variants in eight different exons in PTPN11 gene were detected in 107 patients, of whom 10 were familial cases. Exons 3, 8 and 13 had the highest number of pathogenic variants. The most commonly identified pathogenic variants in this series were in exon 8 (c.922A > G, c.923A > G), observed in 22 of the affected. Congenital cardiac anomalies were present in 84% of the mutation-positive cohort, the majority being defects in the right side of the heart. The most common facial features were downward-slanting palpebral fissures, hypertelorism and low-set posteriorly rotated ears. Other clinical features included short stature (40%), pectus excavatum (54%) and, in males, unilateral or bilateral cryptorchidism (44%). Conclusion The clinical features and mutational spectrum observed in our cohort are similar to those reported in other large studies done worldwide. This is the largest case series of NS-affected individuals with PTPN11 mutations described till date from India.
Collapse
Affiliation(s)
| | - Meenakshi Bhat
- Molecular Genetics, Centre for Human Genetics, Bengaluru, 560100, India.,Pediatric Genetics, Indira Gandhi Institute of Child Health, Bengaluru, 560029, India
| | - Sheela Nampoothiri
- Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre (AIMS), Kochi, 682041, India
| | | | | | | | | | - Swathi Shetty
- Molecular Genetics, Centre for Human Genetics, Bengaluru, 560100, India.
| |
Collapse
|
27
|
Martinelli S, Pannone L, Lissewski C, Brinkmann J, Flex E, Schanze D, Calligari P, Anselmi M, Pantaleoni F, Canale VC, Radio FC, Ioannides A, Rahner N, Schanze I, Josifova D, Bocchinfuso G, Ryten M, Stella L, Tartaglia M, Zenker M. Pathogenic PTPN11 variants involving the poly-glutamine Gln 255 -Gln 256 -Gln 257 stretch highlight the relevance of helix B in SHP2's functional regulation. Hum Mutat 2020; 41:1171-1182. [PMID: 32112654 DOI: 10.1002/humu.24007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/14/2020] [Accepted: 02/27/2020] [Indexed: 01/28/2023]
Abstract
Germline PTPN11 mutations cause Noonan syndrome (NS), the most common disorder among RASopathies. PTPN11 encodes SHP2, a protein tyrosine-phosphatase controlling signaling through the RAS-MAPK and PI3K-AKT pathways. Generally, NS-causing PTPN11 mutations are missense changes destabilizing the inactive conformation of the protein or enhancing its binding to signaling partners. Here, we report on two PTPN11 variants resulting in the deletion or duplication of one of three adjacent glutamine residues (Gln255 -to-Gln257 ). While p.(Gln257dup) caused a typical NS phenotype in carriers of a first family, p.(Gln257del) had incomplete penetrance in a second family. Missense mutations involving Gln256 had previously been reported in NS. This poly-glutamine stretch is located on helix B of the PTP domain, a region involved in stabilizing SHP2 in its autoinhibited state. Molecular dynamics simulations predicted that changes affecting this motif perturb the SHP2's catalytically inactive conformation and/or substrate recognition. Biochemical data showed that duplication and deletion of Gln257 variably enhance SHP2's catalytic activity, while missense changes involving Gln256 affect substrate specificity. Expression of mutants in HEK293T cells documented their activating role on MAPK signaling, uncoupling catalytic activity and modulation of intracellular signaling. These findings further document the relevance of helix B in the regulation of SHP2's function.
Collapse
Affiliation(s)
- Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Pannone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Christina Lissewski
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Julia Brinkmann
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Denny Schanze
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Paolo Calligari
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Massimiliano Anselmi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Francesca Pantaleoni
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Viviana Claudia Canale
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | | | - Adonis Ioannides
- Clinical Genetics, University of Nicosia Medical School, Nicosia, Cyprus.,South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Nils Rahner
- Medical Faculty, Institute of Human Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Ina Schanze
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Dragana Josifova
- South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Mina Ryten
- South East Thames Regional Genetics Service, Guy's Hospital, London, UK
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Martin Zenker
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
28
|
Kanavy DM, McNulty SM, Jairath MK, Brnich SE, Bizon C, Powell BC, Berg JS. Comparative analysis of functional assay evidence use by ClinGen Variant Curation Expert Panels. Genome Med 2019; 11:77. [PMID: 31783775 PMCID: PMC6884856 DOI: 10.1186/s13073-019-0683-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The 2015 American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) guidelines for clinical sequence variant interpretation state that "well-established" functional studies can be used as evidence in variant classification. These guidelines articulated key attributes of functional data, including that assays should reflect the biological environment and be analytically sound; however, details of how to evaluate these attributes were left to expert judgment. The Clinical Genome Resource (ClinGen) designates Variant Curation Expert Panels (VCEPs) in specific disease areas to make gene-centric specifications to the ACMG/AMP guidelines, including more specific definitions of appropriate functional assays. We set out to evaluate the existing VCEP guidelines for functional assays. METHODS We evaluated the functional criteria (PS3/BS3) of six VCEPs (CDH1, Hearing Loss, Inherited Cardiomyopathy-MYH7, PAH, PTEN, RASopathy). We then established criteria for evaluating functional studies based on disease mechanism, general class of assay, and the characteristics of specific assay instances described in the primary literature. Using these criteria, we extensively curated assay instances cited by each VCEP in their pilot variant classification to analyze VCEP recommendations and their use in the interpretation of functional studies. RESULTS Unsurprisingly, our analysis highlighted the breadth of VCEP-approved assays, reflecting the diversity of disease mechanisms among VCEPs. We also noted substantial variability between VCEPs in the method used to select these assays and in the approach used to specify strength modifications, as well as differences in suggested validation parameters. Importantly, we observed discrepancies between the parameters VCEPs specified as required for approved assay instances and the fulfillment of these requirements in the individual assays cited in pilot variant interpretation. CONCLUSIONS Interpretation of the intricacies of functional assays often requires expert-level knowledge of the gene and disease, and current VCEP recommendations for functional assay evidence are a useful tool to improve the accessibility of functional data by providing a starting point for curators to identify approved functional assays and key metrics. However, our analysis suggests that further guidance is needed to standardize this process and ensure consistency in the application of functional evidence.
Collapse
Affiliation(s)
- Dona M Kanavy
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shannon M McNulty
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Meera K Jairath
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah E Brnich
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chris Bizon
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bradford C Powell
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan S Berg
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
29
|
Baraban E, Sadigh S, Rosenbaum J, Van Arnam J, Bogusz AM, Mehr C, Bagg A. Cyclin D1 expression and novel mutational findings in Rosai-Dorfman disease. Br J Haematol 2019; 186:837-844. [PMID: 31172509 DOI: 10.1111/bjh.16006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Rosai-Dorfman disease (RDD) is an enigmatic histiocytic disorder classically diagnosed by a distinctive combination of pathological features: emperipolesis, or migration of intact haematological cells through the voluminous cytoplasm of lesional histiocytes, and expression of S100 by these histiocytes. The pathogenesis has long been elusive until the recent detection of recurrent and mutually exclusive mutations in several oncogenes in the mitogen-activated protein kinase (MAPK) pathway. Based on these findings, we investigated a cohort of 21 RDD patients and found that the lesional histiocytes in 86% (18/21) of patients exhibited strong and diffuse nuclear Cyclin D1 expression, which not only may provide a diagnostic marker for this sometimes pathologically challenging disease, but also probably reflects constitutive MAPK pathway activation because we additionally identified phosphorylated-ERK expression in 90% (19/21) of cases. Further, we performed massively parallel sequencing on a subset (6/18) of the CyclinD1 positive cases, identifying several mutations that have not been previously reported in RDD. Taken together, our findings bolster the concept of RDD as a disease of MAPK activation in a substantial percentage of cases and enhance the current understanding of the pathogenesis of RDD.
Collapse
Affiliation(s)
- Ezra Baraban
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sam Sadigh
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jason Rosenbaum
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John Van Arnam
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Agata M Bogusz
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsea Mehr
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Freire BL, Homma TK, Funari MFA, Lerario AM, Vasques GA, Malaquias AC, Arnhold IJP, Jorge AAL. Multigene Sequencing Analysis of Children Born Small for Gestational Age With Isolated Short Stature. J Clin Endocrinol Metab 2019; 104:2023-2030. [PMID: 30602027 DOI: 10.1210/jc.2018-01971] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/27/2018] [Indexed: 02/04/2023]
Abstract
CONTEXT Patients born small for gestational age (SGA) who present with persistent short stature could have an underlying genetic etiology that will account for prenatal and postnatal growth impairment. We applied a unique massive parallel sequencing approach in cohort of patients with exclusively nonsyndromic SGA to simultaneously interrogate for clinically substantial genetic variants. OBJECTIVE To perform a genetic investigation of children with isolated short stature born SGA. DESIGN Screening by exome (n = 16) or targeted gene panel (n = 39) sequencing. SETTING Tertiary referral center for growth disorders. PATIENTS AND METHODS We selected 55 patients born SGA with persistent short stature without an identified cause of short stature. MAIN OUTCOME MEASURES Frequency of pathogenic findings. RESULTS We identified heterozygous pathogenic or likely pathogenic genetic variants in 8 of 55 patients, all in genes already associated with growth disorders. Four of the genes are associated with growth plate development, IHH (n = 2), NPR2 (n = 2), SHOX (n = 1), and ACAN (n = 1), and two are involved in the RAS/MAPK pathway, PTPN11 (n = 1) and NF1 (n = 1). None of these patients had clinical findings that allowed for a clinical diagnosis. Seven patients were SGA only for length and one was SGA for both length and weight. CONCLUSION These genomic approaches identified pathogenic or likely pathogenic genetic variants in 8 of 55 patients (15%). Six of the eight patients carried variants in genes associated with growth plate development, indicating that mild forms of skeletal dysplasia could be a cause of growth disorders in this group of patients.
Collapse
Affiliation(s)
- Bruna L Freire
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Thais K Homma
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Mariana F A Funari
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Antônio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Gabriela A Vasques
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Alexsandra C Malaquias
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia Pediátrica, Departamento de Pediatria, Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Ivo J P Arnhold
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| |
Collapse
|
31
|
Bessis D, Miquel J, Bourrat E, Chiaverini C, Morice-Picard F, Abadie C, Manna F, Baumann C, Best M, Blanchet P, Bursztejn AC, Capri Y, Coubes C, Giuliano F, Guillaumont S, Hadj-Rabia S, Jacquemont ML, Jeandel C, Lacombe D, Mallet S, Mazereeuw-Hautier J, Molinari N, Pallure V, Pernet C, Philip N, Pinson L, Sarda P, Sigaudy S, Vial Y, Willems M, Geneviève D, Verloes A, Cavé H. Dermatological manifestations in Noonan syndrome: a prospective multicentric study of 129 patients positive for mutation. Br J Dermatol 2019; 180:1438-1448. [PMID: 30417923 DOI: 10.1111/bjd.17404] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Data on dermatological manifestations of Noonan syndrome (NS) remain heterogeneous and are based on limited dermatological expertise. OBJECTIVES To describe the dermatological manifestations of NS, compare them with the literature findings, and test for dermatological phenotype-genotype correlations with or without the presence of PTPN11 mutations. METHODS We performed a large 4-year, prospective, multicentric, collaborative dermatological and genetic study. RESULTS Overall, 129 patients with NS were enrolled, including 65 patients with PTPN11-NS, 34 patients with PTPN11-NS with multiple lentigines (NSML), and 30 patients with NS who had a mutation other than PTPN11. Easy bruising was the most frequent dermatological finding in PTPN11-NS, present in 53·8% of patients. Multiple lentigines and café-au-lait macules (n ≥ 3) were present in 94% and 80% of cases of NSML linked to specific mutations of PTPN11, respectively. Atypical forms of NSML could be associated with NS with RAF1 or NRAS mutations. In univariate analysis, patients without a PTPN11 mutation showed (i) a significantly higher frequency of keratinization disorders (P = 0·001), including keratosis pilaris (P = 0·005), ulerythema ophryogenes (P = 0·0001) and palmar and/or plantar hyperkeratosis (P = 0·06, trend association), and (ii) a significantly higher frequency of scarce scalp hair (P = 0·035) and scarce or absent eyelashes (P = 0·06, trend association) than those with PTPN11 mutations. CONCLUSIONS The cutaneous phenotype of NS with a PTPN11 mutation is generally mild and nonspecific, whereas the absence of a PTPN11 mutation is associated with a high frequency of keratinization disorders and hair abnormalities.
Collapse
Affiliation(s)
- D Bessis
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France.,University of Montpellier, Montpellier, France.,INSERM U1058, Montpellier, France
| | - J Miquel
- Department of Paediatric Dermatology, Femme-Mère-Enfant Hospital, University of South Réunion, Saint-Pierre Réunion, France.,Department of Dermatology, University of Rennes, Rennes, France
| | - E Bourrat
- Department of Paediatric Dermatology, Robert-Debré Hospital, AP-HP, Paris, France
| | - C Chiaverini
- Department of Dermatology, L'Archet 2 Hospital, Nice, France.,University of Nice, Nice, France
| | - F Morice-Picard
- Department of Paediatric Dermatology, Pellegrin University Hospital of Bordeaux, Bordeaux, France
| | - C Abadie
- Department of Clinical Genetics, Sud Hospital, Rennes, France.,University Hospital of Rennes, Rennes, France
| | - F Manna
- University of Montpellier, Montpellier, France.,Department of Medical Information, Epidemiological and Clinical Research Unit, La Colombière Hospital, Montpellier, France
| | - C Baumann
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - M Best
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France.,University of Montpellier, Montpellier, France
| | - P Blanchet
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - A-C Bursztejn
- Department of Dermatology, Brabois Hospital, Nancy, France.,University of Nancy, Nancy, France
| | - Y Capri
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - C Coubes
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - F Giuliano
- University of Nice, Nice, France.,Department of Clinical Genetics, L'Archet 2 Hospital, Nice, France
| | - S Guillaumont
- University of Montpellier, Montpellier, France.,Department of Paediatric Cardiology, Arnaud de Villeneuve Hospital, Montpellier, France
| | - S Hadj-Rabia
- Department of Paediatric Dermatology, Reference Centre for Rare Skin Diseases, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - M-L Jacquemont
- Department of Clinical Genetics, Femme-Mère-Enfant Hospital, University of South Réunion, Saint-Pierre Réunion, France
| | - C Jeandel
- University of Montpellier, Montpellier, France.,Department of Paediatric Endocrinology, Arnaud de Villeneuve Hospital, Montpellier, France
| | - D Lacombe
- Department of Clinical Genetics, Pellegrin University Hospital of Bordeaux, AP-HP, Paris, France
| | - S Mallet
- Department of Dermatology, La Timone Hospital, AP-HM, Marseille, France.,University of Marseille, Marseille, France
| | - J Mazereeuw-Hautier
- Department of Dermatology, Larrey Hospital, Reference Centre for Rare Skin Diseases, Toulouse, France.,University of Toulouse, Toulouse, France
| | - N Molinari
- University of Montpellier, Montpellier, France.,Department of Medical Information, Epidemiological and Clinical Research Unit, La Colombière Hospital, Montpellier, France
| | - V Pallure
- Department of Dermatology, CH, Perpignan, Perpignan, France
| | - C Pernet
- Department of Dermatology, Saint-Eloi Hospital, Competence Centre for Rare Skin Diseases, Montpellier, France
| | - N Philip
- University of Marseille, Marseille, France.,Department of Clinical Genetics, La Timone Hospital, AP-HM, Marseille, France
| | - L Pinson
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - P Sarda
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - S Sigaudy
- University of Marseille, Marseille, France.,Department of Clinical Genetics, La Timone Hospital, AP-HM, Marseille, France
| | - Y Vial
- University of Paris-Diderot, Paris, France.,Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP, Paris, France
| | - M Willems
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France
| | - D Geneviève
- Department of Clinical Genetics, Arnaud de Villeneuve Hospital, Montpellier, France.,INSERM U1183, Montpellier, France
| | - A Verloes
- Department of Clinical Genetics, Robert-Debré Hospital, AP-HP, Paris, France.,University of Paris-Diderot, Paris, France
| | - H Cavé
- University of Paris-Diderot, Paris, France.,Department of Genetic Biochemistry, Robert-Debré Hospital, AP-HP, Paris, France
| |
Collapse
|
32
|
Hornbeck PV, Kornhauser JM, Latham V, Murray B, Nandhikonda V, Nord A, Skrzypek E, Wheeler T, Zhang B, Gnad F. 15 years of PhosphoSitePlus®: integrating post-translationally modified sites, disease variants and isoforms. Nucleic Acids Res 2019; 47:D433-D441. [PMID: 30445427 PMCID: PMC6324072 DOI: 10.1093/nar/gky1159] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/26/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022] Open
Abstract
For 15 years the mission of PhosphoSitePlus® (PSP, https://www.phosphosite.org) has been to provide comprehensive information and tools for the study of mammalian post-translational modifications (PTMs). The number of unique PTMs in PSP is now more than 450 000 from over 22 000 articles and thousands of MS datasets. The most important areas of growth in PSP are in disease and isoform informatics. Germline mutations associated with inherited diseases and somatic cancer mutations have been added to the database and can now be viewed along with PTMs and associated quantitative information on novel 'lollipop' plots. These plots enable researchers to interactively visualize the overlap between disease variants and PTMs, and to identify mutations that may alter phenotypes by rewiring signaling networks. We are expanding the sequence space to include over 30 000 human and mouse isoforms to enable researchers to explore the important but understudied biology of isoforms. This represents a necessary expansion of sequence space to accommodate the growing precision and depth of coverage enabled by ongoing advances in mass spectrometry. Isoforms are aligned using a new algorithm. Exploring the worlds of PTMs and disease mutations in the entire isoform space will hopefully lead to new biomarkers, therapeutic targets, and insights into isoform biology.
Collapse
Affiliation(s)
- Peter V Hornbeck
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Jon M Kornhauser
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Vaughan Latham
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Beth Murray
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Vidhisha Nandhikonda
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Alex Nord
- University of Montana, Missoula, MT, USA
| | - Elżbieta Skrzypek
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | | | - Bin Zhang
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| | - Florian Gnad
- Department of Bioinformatics and Computational Biology, Cell Signaling Technology Inc., Danvers, MA, USA
| |
Collapse
|
33
|
Furlan V, Konc J, Bren U. Inverse Molecular Docking as a Novel Approach to Study Anticarcinogenic and Anti-Neuroinflammatory Effects of Curcumin. Molecules 2018; 23:E3351. [PMID: 30567342 PMCID: PMC6321024 DOI: 10.3390/molecules23123351] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/17/2018] [Indexed: 11/16/2022] Open
Abstract
Research efforts are placing an ever increasing emphasis on identifying signal transduction pathways related to the chemopreventive activity of curcumin. Its anticarcinogenic effects are presumably mediated by the regulation of signaling cascades, including nuclear factor κB (NF-κB), activator protein 1 (AP-1), and mitogen-activated protein kinases (MAPK). By modulating signal transduction pathways, curcumin induces apoptosis in malignant cells, thus inhibiting cancer development and progression. Due to the lack of mechanistic insight in the scientific literature, we developed a novel inverse molecular docking protocol based on the CANDOCK algorithm. For the first time, we performed inverse molecular docking of curcumin into a collection of 13,553 available human protein structures from the Protein Data Bank resulting in prioritized target proteins of curcumin. Our predictions were in agreement with the scientific literature and confirmed that curcumin binds to folate receptor β, DNA (cytosine-5)-methyltransferase 3A, metalloproteinase-2, mitogen-activated protein kinase 9, epidermal growth factor receptor and apoptosis-inducing factor 1. We also identified new potential protein targets of curcumin, namely deoxycytidine kinase, NAD-dependent protein deacetylase sirtuin-1 and -2, ecto-5'-nucleotidase, core histone macro-H2A.1, tyrosine-protein phosphatase non-receptor type 11, macrophage colony-stimulating factor 1 receptor, GTPase HRas, aflatoxin B1 aldehyde reductase member 3, aldo-keto reductase family 1 member C3, amiloride-sensitive amine oxidase, death-associated protein kinase 2 and tryptophan-tRNA ligase, that may all play a crucial role in its observed anticancer effects. Moreover, our inverse docking results showed that curcumin potentially binds also to the proteins cAMP-specific 3',5'-cyclic phosphodiesterase 4D and 17-β-hydroxysteroid dehydrogenase type 10, which provides a new explanation for its efficiency in the treatment of Alzheimer's disease. We firmly believe that our computational results will complement and direct future experimental studies on curcumin's anticancer activity as well as on its therapeutic effects against Alzheimer's disease.
Collapse
Affiliation(s)
- Veronika Furlan
- Faculty of Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia.
| | - Janez Konc
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia.
| | - Urban Bren
- Faculty of Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia.
- National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
34
|
Bustelo XR, Crespo P, Fernández-Pisonero I, Rodríguez-Fdez S. RAS GTPase-dependent pathways in developmental diseases: old guys, new lads, and current challenges. Curr Opin Cell Biol 2018; 55:42-51. [PMID: 30007125 PMCID: PMC7615762 DOI: 10.1016/j.ceb.2018.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/14/2018] [Accepted: 06/14/2018] [Indexed: 10/28/2022]
Abstract
Deregulated RAS signaling is associated with increasing numbers of congenital diseases usually referred to as RASopathies. The spectrum of genes and mutant alleles causing these diseases has been significantly expanded in recent years. This progress has triggered new challenges, including the origin and subsequent selection of the mutations driving these diseases, the specific pathobiological programs triggered by those mutations, the type of correlations that exist between the genotype and the clinical features of patients, and the ancillary genetic factors that influence the severity of the disease in patients. These issues also directly impinge on the feasibility of using RAS pathway drugs to treat RASopathy patients. Here, we will review the main developments and pending challenges in this research topic.
Collapse
Affiliation(s)
- Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain.
| | - Piero Crespo
- CIBERONC, CSIC-University of Cantabria, 39011 Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-University of Cantabria, 39011 Santander, Spain
| | - Isabel Fernández-Pisonero
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
35
|
Py C, Christinat Y, Kreutzfeldt M, McKee TA, Dietrich PY, Tsantoulis P. Response of NF1-Mutated Melanoma to an MEK Inhibitor. JCO Precis Oncol 2018; 2:1-11. [DOI: 10.1200/po.18.00028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Céline Py
- All authors: University Hospital of Geneva, Geneva, Switzerland
| | - Yann Christinat
- All authors: University Hospital of Geneva, Geneva, Switzerland
| | | | - Thomas A. McKee
- All authors: University Hospital of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
36
|
Tajan M, Paccoud R, Branka S, Edouard T, Yart A. The RASopathy Family: Consequences of Germline Activation of the RAS/MAPK Pathway. Endocr Rev 2018; 39:676-700. [PMID: 29924299 DOI: 10.1210/er.2017-00232] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Noonan syndrome [NS; Mendelian Inheritance in Men (MIM) #163950] and related syndromes [Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome; MIM #151100), Noonan-like syndrome with loose anagen hair (MIM #607721), Costello syndrome (MIM #218040), cardio-facio-cutaneous syndrome (MIM #115150), type I neurofibromatosis (MIM #162200), and Legius syndrome (MIM #611431)] are a group of related genetic disorders associated with distinctive facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was clinically described more than 50 years ago, and disease genes have been identified throughout the last 3 decades, providing a molecular basis to better understand their physiopathology and identify targets for therapeutic strategies. Most of these genes encode proteins belonging to or regulating the so-called RAS/MAPK signaling pathway, so these syndromes have been gathered under the name RASopathies. In this review, we provide a clinical overview of RASopathies and an update on their genetics. We then focus on the functional and pathophysiological effects of RASopathy-causing mutations and discuss therapeutic perspectives and future directions.
Collapse
Affiliation(s)
- Mylène Tajan
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Romain Paccoud
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Sophie Branka
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Armelle Yart
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| |
Collapse
|
37
|
Gelb BD, Cavé H, Dillon MW, Gripp KW, Lee JA, Mason-Suares H, Rauen KA, Williams B, Zenker M, Vincent LM. ClinGen's RASopathy Expert Panel consensus methods for variant interpretation. Genet Med 2018; 20:1334-1345. [PMID: 29493581 PMCID: PMC6119537 DOI: 10.1038/gim.2018.3] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/02/2018] [Indexed: 01/06/2023] Open
Abstract
Purpose Standardized and accurate variant assessment is essential for effective medical care. To that end, Clinical Genome (ClinGen) Resource clinical domain working groups (CDWG) are systematically reviewing disease-associated genes for sufficient evidence to support disease causality and creating disease-specific specifications of ACMG-AMP guidelines for consistent and accurate variant classification. Methods The ClinGen RASopathy CDWG established an expert panel (EP) to curate gene information and generate gene and disease-specific specifications to ACMG-AMP variant classification framework. These specifications were tested by classifying 37 exemplar pathogenic variants plus an additional 66 variants in ClinVar distributed across nine RASopathy genes. Results RASopathy-related specifications were applied to sixteen ACMG-AMP criteria, with five also having adjustable strength with availability of additional evidence. Another five criteria were deemed not applicable. Key adjustments to minor allele frequency thresholds, multiple de novo occurrence events and/or segregation, and strength adjustments impacted 60% of variant classifications. Unpublished case-level data from participating laboratories impacted 45% of classifications supporting the need for data sharing. Conclusions RAS-specific ACMG-AMP specifications optimized the utility of available clinical evidence and Ras/MAPK pathway-specific characteristics to consistently classify RASopathy-associated variants. These specifications highlight how grouping genes by shared features promotes rapid multi-genic variant assessment without sacrificing specificity and accuracy.
Collapse
Affiliation(s)
- Bruce D Gelb
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hélène Cavé
- Département de Génétique, Hôpital Robert Debré and Institut Universitaire d'Hématologie, Université Paris Diderot, Paris-Sorbonne-Cité, Paris, France
| | - Mitchell W Dillon
- Icahn School of Medicine at Mount Sinai, Molecular Genetic Testing Laboratory, New York, New York, USA
| | - Karen W Gripp
- Division of Medical Genetics, A.I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Jennifer A Lee
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Heather Mason-Suares
- Laboratory for Molecular Medicine, Partners Healthcare, Cambridge, Massachusetts, USA
| | - Katherine A Rauen
- Department of Pediatrics, University of California Davis, UC Davis MIND Institute, Sacramento, California, USA
| | | | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | | | | |
Collapse
|
38
|
Abstract
Adult height and growth patterns are largely genetically programmed. Studies in twins have indicated that the heritability of height is high (>80%), suggesting that genetic variation is the main determinant of stature. Height exhibits a normal (Gaussian) distribution according to sex, age, and ancestry. Short stature is usually defined as a height which is 2 standard deviations (S.D.) less than the mean height of a specific population. This definition includes 2.3% of the population and usually includes healthy individuals. In this group of short stature non-syndromic conditions, the genetic influence occurs polygenically or oligogenically. As a rule, each common genetic variant accounts for a small effect (1mm) on individual height variation. Recently, several studies demonstrated that some rare variants can cause greater effect on height, without causing a syndromic condition. In more extreme cases, height SDS below 2.5 or 3 (which would comprise approximately 0.6 and 0.1% of the population, respectively) is frequently associated with syndromic conditions and are usually caused by a monogenic defect. More than 1,000 inherited/genetic diseases have growth disorder as an important phenotype. These conditions are usually responsible for syndromic short stature. In the coming years, we expect to discover several genetic causes of short stature, thereby explaining the phenotype of what we currently classify as short stature of unknown cause. These discoveries will have a profound impact on the follow-up and treatment of these children.
Collapse
Affiliation(s)
- Michelle Grunauer
- Pediatric Intensive Care Unit, Hospital de los Valles, Escuela de Medicina, Universidad San Francisco de Quito (USFQ), Quito, Ecuador
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genetica (LIM25), Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), Sao Paulo, Brazil.
| |
Collapse
|
39
|
LaRochelle JR, Fodor M, Ellegast JM, Liu X, Vemulapalli V, Mohseni M, Stams T, Buhrlage SJ, Stegmaier K, LaMarche MJ, Acker MG, Blacklow SC. Identification of an allosteric benzothiazolopyrimidone inhibitor of the oncogenic protein tyrosine phosphatase SHP2. Bioorg Med Chem 2017; 25:6479-6485. [DOI: 10.1016/j.bmc.2017.10.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
|
40
|
Elson A. Stepping out of the shadows: Oncogenic and tumor-promoting protein tyrosine phosphatases. Int J Biochem Cell Biol 2017; 96:135-147. [PMID: 28941747 DOI: 10.1016/j.biocel.2017.09.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/18/2022]
Abstract
Protein tyrosine phosphorylation is critical for proper function of cells and organisms. Phosphorylation is regulated by the concerted but generically opposing activities of tyrosine kinases (PTKs) and tyrosine phosphatases (PTPs), which ensure its proper regulation, reversibility, and ability to respond to changing physiological situations. Historically, PTKs have been associated mainly with oncogenic and pro-tumorigenic activities, leading to the generalization that protein dephosphorylation is anti-oncogenic and hence that PTPs are tumor-suppressors. In many cases PTPs do suppress tumorigenesis. However, a growing body of evidence indicates that PTPs act as dominant oncogenes and drive cell transformation in a number of contexts, while in others PTPs support transformation that is driven by other oncogenes. This review summarizes the known transforming and tumor-promoting activities of the classical, tyrosine specific PTPs and highlights their potential as drug targets for cancer therapy.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|