1
|
Korleski J, Sall S, Luly KM, Johnson MK, Johnson AL, Khela H, Lal B, Taylor TC, Ashby JM, Alonso H, Li A, Zhou W, Smith-Connor K, Hughes R, Tzeng SY, Laterra J, Green JJ, Lopez-Bertoni H. Multipronged SMAD pathway targeting by lipophilic poly(β-amino ester) miR-590-3p nanomiRs inhibits mesenchymal glioblastoma growth and prolongs survival. Signal Transduct Target Ther 2025; 10:145. [PMID: 40301302 PMCID: PMC12041600 DOI: 10.1038/s41392-025-02223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/25/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025] Open
Abstract
ASBSTRACT Despite aggressive therapy, glioblastoma (GBM) recurs in almost all patients and treatment options are very limited. Despite our growing understanding of how cellular transitions associate with relapse in GBM, critical gaps remain in our ability to block these molecular changes and treat recurrent disease. In this study we combine computational biology, forward-thinking understanding of miRNA biology and cutting-edge nucleic acid delivery vehicles to advance targeted therapeutics for GBM. Computational analysis of RNA sequencing from clinical GBM specimens identified TGFβ type II receptor (TGFBR2) as a key player in the mesenchymal transition associated with worse outcome in GBM. Mechanistically, we show that elevated levels of TGFBR2 is conducive to reduced temozolomide (TMZ) sensitivity. This effect is, at least partially, induced by stem-cell driving events coordinated by the reprogramming transcription factors Oct4 and Sox2 that lead to open chromatin states. We show that blocking TGFBR2 via molecular and pharmacological approaches decreases stem cell capacity and sensitivity of clinical recurrent GBM (rGBM) isolates to TMZ in vitro. Network analysis uncovered miR-590-3p as a tumor suppressor that simultaneously inhibits multiple oncogenic nodes downstream of TGFBR2. We also developed novel biodegradable lipophilic poly(β-amino ester) nanoparticles (LiPBAEs) for in vivo microRNA (miRNAs) delivery. Following direct intra-tumoral infusion, these nanomiRs efficiently distribute through the tumors. Importantly, miR-590-3p nanomiRs inhibited the growth and extended survival of mice bearing orthotopic human rGBM xenografts, with an apparent 30% cure rate. These results show that miRNA-based targeted therapeutics provide new opportunities to treat rGBM and bypass the resistance to standard of care therapy.
Collapse
Affiliation(s)
- Jack Korleski
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota, USA
| | - Sophie Sall
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Kathryn M Luly
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maya K Johnson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Biology, Johns Hopkins University, Baltimore, USA
| | - Amanda L Johnson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Harmon Khela
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, USA
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - T C Taylor
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, USA
| | - Jean Micheal Ashby
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Hector Alonso
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Alice Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | - Russell Hughes
- Single Cell & Transcriptomics Core at the Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Departments of Materials Science & Engineering and Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, USA.
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, USA.
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA.
| | - Hernando Lopez-Bertoni
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA.
| |
Collapse
|
2
|
Bernal Astrain G, Strakhova R, Jo CH, Teszner E, Killoran RC, Smith MJ. The small GTPase MRAS is a broken switch. Nat Commun 2025; 16:647. [PMID: 39809765 PMCID: PMC11733253 DOI: 10.1038/s41467-025-55967-y] [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: 08/28/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Intense research on founding members of the RAS superfamily has defined our understanding of these critical signalling proteins, leading to the premise that small GTPases function as molecular switches dependent on differential nucleotide loading. The closest homologs of H/K/NRAS are the three-member RRAS family, and interest in the MRAS GTPase as a regulator of MAPK activity has recently intensified. We show here that MRAS does not function as a classical switch and is unable to exchange GDP-to-GTP in solution or when tethered to a lipid bilayer. The exchange defect is unaffected by inclusion of the GEF SOS1 and is conserved in a distal ortholog from nematodes. Synthetic activating mutations widely used to study the function of MRAS in a presumed GTP-loaded state do not increase exchange, but instead drive effector binding due to sampling of an activated conformation in the GDP-loaded state. This includes nucleation of the SHOC2-PP1Cα holophosphatase complex. Acquisition of NMR spectra from isotopically labeled MRAS in live cells validated the GTPase remains fully GDP-loaded, even a supposed activated mutant. These data show that RAS GTPases, including those most similar to KRAS, have disparate biochemical activities and challenge current dogma on MRAS, suggesting previous data may need reinterpretation.
Collapse
Affiliation(s)
- Gabriela Bernal Astrain
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Regina Strakhova
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Chang Hwa Jo
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Emma Teszner
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Ryan C Killoran
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
3
|
Yılmaz Uzman C, Gürsoy S, Özkan B, Vuran G, Ayyıldız Emecen D, Köprülü Ö, Bilen MM, Hazan F. Clinical features and molecular genetics of patients with RASopathies: expanding the phenotype with rare genes and novel variants. Eur J Pediatr 2024; 184:108. [PMID: 39725732 DOI: 10.1007/s00431-024-05825-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/13/2024] [Accepted: 11/01/2024] [Indexed: 12/28/2024]
Abstract
The RASopathies are a group of disorders resulting from a germline variant in the genes encoding the Ras/mitogen-activated protein kinase pathway. These disorders include Noonan syndrome (NS), cardiofaciocutaneous syndrome (CFC), Costello syndrome (CS), Legius syndrome (LS), and neurofibromatosis type 1 (NF1), and have overlapping clinical features due to RAS/MAPK dysfunction. In this study, we aimed to describe the clinical and molecular features of patients exhibiting phenotypic manifestations consistent with RASopathies. The study included 149 patients from 146 unrelated families who were admitted between 2019 and 2023 with a clinical suspicion of RASopathy spectrum disorder. Clinical and laboratory characteristics of the patients at the time of the diagnosis were obtained from hospital records. Variant analysis of twenty-four RASopathy genes was performed using a targeted next-generation sequencing (NGS) panel, and the variants were classified according to American College of Medical Genetics and Genomics Standards and Guidelines recommendations. Pathogenic/likely pathogenic variants were detected in 39 out of 149 patients (26.1%). Thirty-two patients were diagnosed as NS (32/39; 82%). The variants detected in NS patients were PTPN11 (21/32; 65.6%), LZTR1 (3/32; 9.3%), SOS1 (2/32; 6.2%), RAF1 (2/32; 6.2%), RIT1 (2/32; 6.2%), KRAS (1/32; 3.1%), and RRAS (1/32; 3.1%) genes, respectively. The remaining patients were diagnosed with CS (2/39; 5.1%), NF1 (2/39; 5.1%), NF-NS (2/39; 5.1%), and CFC (1/39; 2.5%). We observed rare clinical findings including lymphangioma circumscriptum, Meckel's diverticulum, and omphalocele in three patients with PTPN11 gene variations. Additionally, we detected corpus callosum thickness in a patient with the SOS1 gene variant, which has not been previously described in NS. We also identified three novel variants in RIT1, BRAF, and NF1 genes. CONCLUSION In this study, we described rare clinical manifestations and detected three novel variants in NF1, BRAF, and RIT1 genes. We propose that NGS technology enables the detection of variants in rare genes responsible for the etiology of RASopathies. The study, therefore, not only contributes to the existing literature but also expands the spectrum of genotype and phenotype of RASopathies. WHAT IS KNOWN • RASopathies are a group of disorders caused by germline variants in genes involved in the Ras/mitogen-activated protein kinase (RAS/MAPK) pathway. • These disorders, including Noonan syndrome (NS), Cardiofaciocutaneous syndrome (CFC), Costello syndrome (CS), Legius syndrome, and Neurofibromatosis type 1 (NF1), share overlapping clinical features due to RAS/MAPK dysfunction. Molecular diagnosis of RASopathies is crucial for understanding the genetic basis and guiding clinical management, although the phenotype-genotype relationships remain incompletely defined. WHAT IS NEW • This study provides new insights into the molecular and clinical characteristics of RASopathies by examining 149 patients from 146 families, with a focus on the genetic variants found in 24 RASopathy-related genes. Three novel variants were identified in the RIT1, BRAF, and NF1 genes, expanding the genetic spectrum of RASopathies. • Additionally, rare clinical findings, such as lymphangioma circumscriptum and corpus callosum thickness, were reported in patients with PTPN11 and SOS1 gene variations, respectively. These observations contribute new phenotypic data to the existing body of knowledge.
Collapse
Affiliation(s)
- Ceren Yılmaz Uzman
- Department of Pediatric Genetics, Dr. Behçet Uz Children's Hospital, Izmir, Turkey.
| | - Semra Gürsoy
- Department of Pediatric Genetics, Dokuz Eylül University Faculty of Medicine, Izmir, Turkey
| | - Behzat Özkan
- Department of Pediatric Endocrinology, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| | - Gamze Vuran
- Department of Pediatric Cardiology, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| | | | - Özge Köprülü
- Department of Pediatric Endocrinology, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| | - Mertkan Mustafa Bilen
- Department of Pediatric Cardiology, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| | - Filiz Hazan
- Department of Medical Genetics, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| |
Collapse
|
4
|
Kong X, Zhuo X, Huang X, Shang L, Lan T, Qin H, Chen X, Lv C, Xu Q, Wong PP. Multi-omics analysis reveals a pericyte-associated gene expression signature for predicting prognosis and therapeutic responses in solid cancers. Genomics 2024; 116:110942. [PMID: 39326641 DOI: 10.1016/j.ygeno.2024.110942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
The influence of the stroma on cancer progression has been underestimated, particularly the role of vascular pericytes in the tumor microenvironment. Herein, we identified 51 differentially expressed genes in tumor-derived pericytes (TPCs) by analyzing transcriptomic data from TCGA alongside our proteomic data. Using five key TPC-related genes, we constructed a prognostic risk model that accurately predicts prognosis and treatment responses in liver and lung cancers. Enrichment analyses linked these genes to blood vessel remodeling, function, and immune-related pathways. Single-cell RNA sequencing data from the GEO database validated these findings, showing significant upregulation of AKAP12 and RRAS in TPCs. Immunostaining confirmed increased expression of these genes in liver and lung tumors. Depletion of RRAS or AKAP12 in TPCs restored their blood vessel-supporting role. Overall, our findings suggest that TPC-related gene profiles can predict patient outcomes and therapeutic responses in solid cancers, and targeting these profiles could be an improved treatment strategy.
Collapse
Affiliation(s)
- Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xianhua Zhuo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xi Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Lihuan Shang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tianjun Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510010, China
| | - Hongquan Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaochun Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Cui Lv
- Clinical Biobank Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Qiuping Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
5
|
Mozzarelli AM, Simanshu DK, Castel P. Functional and structural insights into RAS effector proteins. Mol Cell 2024; 84:2807-2821. [PMID: 39025071 PMCID: PMC11316660 DOI: 10.1016/j.molcel.2024.06.027] [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: 03/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
RAS proteins are conserved guanosine triphosphate (GTP) hydrolases (GTPases) that act as molecular binary switches and play vital roles in numerous cellular processes. Upon GTP binding, RAS GTPases adopt an active conformation and interact with specific proteins termed RAS effectors that contain a conserved ubiquitin-like domain, thereby facilitating downstream signaling. Over 50 effector proteins have been identified in the human proteome, and many have been studied as potential mediators of RAS-dependent signaling pathways. Biochemical and structural analyses have provided mechanistic insights into these effectors, and studies using model organisms have complemented our understanding of their role in physiology and disease. Yet, many critical aspects regarding the dynamics and biological function of RAS-effector complexes remain to be elucidated. In this review, we discuss the mechanisms and functions of known RAS effector proteins, provide structural perspectives on RAS-effector interactions, evaluate their significance in RAS-mediated signaling, and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Alessandro M Mozzarelli
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter NYU Cancer Center, NYU Langone Health, New York, NY, USA
| | - Dhirendra K Simanshu
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | - Pau Castel
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter NYU Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
6
|
Arfeuille C, Vial Y, Cadenet M, Caye-Eude A, Fenneteau O, Neven Q, Bonnard AA, Pizzi S, Carpentieri G, Capri Y, Girardi K, Pedace L, Macchiaiolo M, Boudhar K, Khaled MB, Chahla WA, Lutun A, Fahd M, Drunat S, Flex E, Dalle JH, Strullu M, Locatelli F, Tartaglia M, Cavé H. Germline bi-allelic SH2B3/LNK alteration predisposes to a neonatal juvenile myelomonocytic leukemia-like disorder. Haematologica 2024; 109:2542-2554. [PMID: 37981895 PMCID: PMC11290538 DOI: 10.3324/haematol.2023.283917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare, generally aggressive myeloproliferative neoplasm affecting young children. It is characterized by granulomonocytic expansion, with monocytosis infiltrating peripheral tissues. JMML is initiated by mutations upregulating RAS signaling. Approximately 10% of cases remain without an identified driver event. Exome sequencing of two unrelated cases of familial JMML of unknown genetics and analysis of the French JMML cohort identified 11 patients with variants in SH2B3, encoding LNK, a negative regulator of the JAK-STAT pathway. All variants were absent from healthy population databases, and the mutation spectrum was consistent with a loss of function of the LNK protein. A stoploss variant was shown to affect both protein synthesis and stability. The other variants were either truncating or missense, the latter affecting the SH2 domain that interacts with activated JAK. Of the 11 patients, eight from five families inherited pathogenic bi-allelic SH2B3 germline variants from their unaffected heterozygous parents. These children represent half of the cases with no identified causal mutation in the French cohort. They displayed typical clinical and hematologic features of JMML with neonatal onset and marked thrombocytopenia. They had a hypomethylated DNA profile with fetal characteristics and did not have additional genetic alterations. All patients showed partial or complete spontaneous clinical resolution. However, progression to thrombocythemia and immunity-related pathologies may be of concern later in life. Bi-allelic SH2B3 germline mutations thus define a new condition predisposing to a JMML-like disorder, suggesting that JAK pathway deregulation is capable of initiating JMML, and opening new therapeutic options.
Collapse
Affiliation(s)
- Chloé Arfeuille
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Yoann Vial
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Margaux Cadenet
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Aurélie Caye-Eude
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Odile Fenneteau
- Service d'Hématologie Biologique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Quentin Neven
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Adeline A Bonnard
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Giovanna Carpentieri
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Yline Capri
- Département de Génétique, Unité de Génétique clinique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Katia Girardi
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Lucia Pedace
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Kamel Boudhar
- Service de réanimation néonatale, Hôpital Central de l'Armée, Alger, Algérie
| | - Monia Ben Khaled
- University of Tunis El Manar, Faculty of Medicine of Tunis, 1007, Tunisia. Pediatric Immuno- Hematology Unit, Bone Marrow Transplantation Center Tunis, Tunis, Tunisia
| | - Wadih Abou Chahla
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire de Lille, Lille
| | - Anne Lutun
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire d'Amiens, Amiens
| | - Mony Fahd
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Séverine Drunat
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome
| | - Jean-Hugues Dalle
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Marion Strullu
- INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France; Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Department of Pediatrics, Catholic University of the Sacred Hearth, 00168 Rome
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Hélène Cavé
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris.
| |
Collapse
|
7
|
Rauen KA, Tidyman WE. RASopathies - what they reveal about RAS/MAPK signaling in skeletal muscle development. Dis Model Mech 2024; 17:dmm050609. [PMID: 38847227 PMCID: PMC11179721 DOI: 10.1242/dmm.050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
Collapse
Affiliation(s)
- Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, 95817, USA
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| | - William E Tidyman
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
8
|
Pucci P, Lee LC, Han M, Matthews JD, Jahangiri L, Schlederer M, Manners E, Sorby-Adams A, Kaggie J, Trigg RM, Steel C, Hare L, James ER, Prokoph N, Ducray SP, Merkel O, Rifatbegovic F, Luo J, Taschner-Mandl S, Kenner L, Burke GAA, Turner SD. Targeting NRAS via miR-1304-5p or farnesyltransferase inhibition confers sensitivity to ALK inhibitors in ALK-mutant neuroblastoma. Nat Commun 2024; 15:3422. [PMID: 38653965 PMCID: PMC11039739 DOI: 10.1038/s41467-024-47771-x] [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: 10/28/2022] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Targeting Anaplastic lymphoma kinase (ALK) is a promising therapeutic strategy for aberrant ALK-expressing malignancies including neuroblastoma, but resistance to ALK tyrosine kinase inhibitors (ALK TKI) is a distinct possibility necessitating drug combination therapeutic approaches. Using high-throughput, genome-wide CRISPR-Cas9 knockout screens, we identify miR-1304-5p loss as a desensitizer to ALK TKIs in aberrant ALK-expressing neuroblastoma; inhibition of miR-1304-5p decreases, while mimics of this miRNA increase the sensitivity of neuroblastoma cells to ALK TKIs. We show that miR-1304-5p targets NRAS, decreasing cell viability via induction of apoptosis. It follows that the farnesyltransferase inhibitor (FTI) lonafarnib in addition to ALK TKIs act synergistically in neuroblastoma, inducing apoptosis in vitro. In particular, on combined treatment of neuroblastoma patient derived xenografts with an FTI and an ALK TKI complete regression of tumour growth is observed although tumours rapidly regrow on cessation of therapy. Overall, our data suggests that combined use of ALK TKIs and FTIs, constitutes a therapeutic approach to treat high risk neuroblastoma although prolonged therapy is likely required to prevent relapse.
Collapse
Affiliation(s)
- Perla Pucci
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Liam C Lee
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Merck & Co, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Miaojun Han
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- OncoSec, San Diego, CA, 92121, USA
| | - Jamie D Matthews
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Leila Jahangiri
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Department of Life Sciences, Birmingham City University, Birmingham, UK
- Nottingham Trent University, School of Science & Technology, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Michaela Schlederer
- Department of Pathology, Division of Experimental and Translational Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - Eleanor Manners
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Chelsea and Westminster Hospital, NHS Foundation Trust, London, SW10 9NH, UK
| | - Annabel Sorby-Adams
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Joshua Kaggie
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Ricky M Trigg
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Functional Genomics, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Christopher Steel
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Lucy Hare
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Department of Paediatric Haematology, Oncology and Palliative Care, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Emily R James
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Nina Prokoph
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Stephen P Ducray
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK
| | - Olaf Merkel
- Department of Pathology, Medical University of Vienna, Vienna, 1090, Austria
- European Research Initiative for ALK related malignancies (ERIA), Cambridge, CB2 0QQ, UK
| | - Firkret Rifatbegovic
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, 1090, Austria
- European Research Initiative for ALK related malignancies (ERIA), Cambridge, CB2 0QQ, UK
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria
| | - G A Amos Burke
- Department of Paediatric Haematology, Oncology and Palliative Care, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Suzanne D Turner
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, CB20QQ, UK.
- European Research Initiative for ALK related malignancies (ERIA), Cambridge, CB2 0QQ, UK.
- Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
9
|
Rodríguez-Martín M, Báez-Flores J, Ribes V, Isidoro-García M, Lacal J, Prieto-Matos P. Non-Mammalian Models for Understanding Neurological Defects in RASopathies. Biomedicines 2024; 12:841. [PMID: 38672195 PMCID: PMC11048513 DOI: 10.3390/biomedicines12040841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
RASopathies, a group of neurodevelopmental congenital disorders stemming from mutations in the RAS/MAPK pathway, present a unique opportunity to delve into the intricacies of complex neurological disorders. Afflicting approximately one in a thousand newborns, RASopathies manifest as abnormalities across multiple organ systems, with a pronounced impact on the central and peripheral nervous system. In the pursuit of understanding RASopathies' neurobiology and establishing phenotype-genotype relationships, in vivo non-mammalian models have emerged as indispensable tools. Species such as Danio rerio, Drosophila melanogaster, Caenorhabditis elegans, Xenopus species and Gallus gallus embryos have proven to be invaluable in shedding light on the intricate pathways implicated in RASopathies. Despite some inherent weaknesses, these genetic models offer distinct advantages over traditional rodent models, providing a holistic perspective on complex genetics, multi-organ involvement, and the interplay among various pathway components, offering insights into the pathophysiological aspects of mutations-driven symptoms. This review underscores the value of investigating the genetic basis of RASopathies for unraveling the underlying mechanisms contributing to broader neurological complexities. It also emphasizes the pivotal role of non-mammalian models in serving as a crucial preliminary step for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Vanessa Ribes
- Institut Jacques Monod, Université Paris Cité, CNRS, F-75013 Paris, France;
| | - María Isidoro-García
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Biochemistry Department, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Pablo Prieto-Matos
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Pediatrics, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Science, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
10
|
Sun C, Mahapatra KD, Elton J, Li C, Fernando W, Lohcharoenkal W, Lapins J, Homey B, Sonkoly E, Pivarcsi A. MicroRNA-23b Plays a Tumor-Suppressive Role in Cutaneous Squamous Cell Carcinoma and Targets Ras-Related Protein RRAS2. J Invest Dermatol 2023; 143:2386-2396. [PMID: 37423552 DOI: 10.1016/j.jid.2023.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/11/2023]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is one of the most common types of cancer with metastatic potential. MicroRNAs regulate gene expression at the post-transcriptional level. In this study, we report that miR-23b is downregulated in cSCCs and in actinic keratosis and that its expression is regulated by the MAPK signaling pathway. We show that miR-23b suppresses the expression of a gene network associated with key oncogenic pathways and that the miR-23b-gene signature is enriched in human cSCCs. miR-23b decreased the expression of FGF2 both at mRNA and protein levels and impaired the angiogenesis-inducing ability of cSCC cells. miR23b overexpression suppressed the capacity of cSCC cells to form colonies and spheroids, whereas the CRISPR/Cas9-mediated deletion of MIR23B resulted in increased colony and tumor sphere formation in vitro. In accordance with this, miR-23b-overexpressing cSCC cells formed significantly smaller tumors upon injection into immunocompromised mice with decreased cell proliferation and angiogenesis. Mechanistically, we verify RRAS2 as a direct target of miR-23b in cSCC. We show that RRAS2 is overexpressed in cSCC and that interference with its expression impairs angiogenesis and colony and tumorsphere formation. Taken together, our results suggest that miR-23b acts in a tumor-suppressive manner in cSCC, and its expression is decreased during squamous carcinogenesis.
Collapse
Affiliation(s)
- Chengxi Sun
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kunal Das Mahapatra
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Elton
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chen Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Winnie Fernando
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Warangkana Lohcharoenkal
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Lapins
- Unit of Dermatology, Karolinska University Hospital, Stockholm, Sweden
| | - Bernhard Homey
- Department of Dermatology, University Hospital Duesseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Enikö Sonkoly
- Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Dermatology, Karolinska University Hospital, Stockholm, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Andor Pivarcsi
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Unit of Dermatology and Venerology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Zhang XY, Yuan ZH, Li FD, Yue XP. Integrating transcriptome and metabolome to identify key genes regulating important muscular flavour precursors in sheep. Animal 2022; 16:100679. [PMID: 36481351 DOI: 10.1016/j.animal.2022.100679] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Flavour precursors are the basis of meat flavour, and their metabolism is regulated by a variety of enzymes. Thus, it is of great significance to identify the key genes related to meat flavour precursors. In this study, the difference in flavour precursors and transcriptome between Hu sheep and Dorper with different intramuscular fat (IMF) content were investigated using widely targeted metabolomics and RNA-sequencing technologies. Then, the key genes regulating the metabolism of vital precursors were explored by integrating transcriptome and metabolome. Consequently, 594 metabolites were detected in sheep longissimus dorsi, and 76 differentially abundant metabolites (DAMs) were identified between Hu sheep and Dorper. No DAMs were observed between distinct IMF content groups within each breed. A total of 10 lysophospholipids (LPs), including four lysophospholipid ethanolamines and six lysophospholipid cholines, were identified as the main differential precursors between Hu sheep and Dorper. Furthermore, the weighted gene coexpression network analysis uncovered three differentially coexpression modules that were significantly associated with the content of differential LPs in Dorper. From the three modules, GLB1, PLD3, LPCAT2, DGKE, ACOT7, and CH25H genes were identified as key genes regulating the metabolism of LPs. This work provides an insight into understanding the difference in flavour between different sheep breeds, as well as a basis for further exploring the regulatory mechanism of key genes on LPs.
Collapse
Affiliation(s)
- X Y Zhang
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, PR China
| | - Z H Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education, Yangzhou University, Yangzhou 225000, PR China
| | - F D Li
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, PR China
| | - X P Yue
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, PR China.
| |
Collapse
|
12
|
Tartaglia M, Aoki Y, Gelb BD. The molecular genetics of RASopathies: An update on novel disease genes and new disorders. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:425-439. [PMID: 36394128 PMCID: PMC10100036 DOI: 10.1002/ajmg.c.32012] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/18/2022]
Abstract
Enhanced signaling through RAS and the mitogen-associated protein kinase (MAPK) cascade underlies the RASopathies, a family of clinically related disorders affecting development and growth. In RASopathies, increased RAS-MAPK signaling can result from the upregulated activity of various RAS GTPases, enhanced function of proteins positively controlling RAS function or favoring the efficient transmission of RAS signaling to downstream transducers, functional upregulation of RAS effectors belonging to the MAPK cascade, or inefficient signaling switch-off operated by feedback mechanisms acting at different levels. The massive effort in RASopathy gene discovery performed in the last 20 years has identified more than 20 genes implicated in these disorders. It has also facilitated the characterization of several molecular activating mechanisms that had remained unappreciated due to their minor impact in oncogenesis. Here, we provide an overview on the discoveries collected during the last 5 years that have delivered unexpected insights (e.g., Noonan syndrome as a recessive disease) and allowed to profile new RASopathies, novel disease genes and new molecular circuits contributing to the control of RAS-MAPK signaling.
Collapse
Affiliation(s)
- Marco Tartaglia
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù, IRCCSRomeItaly
| | - Yoko Aoki
- Department of Medical GeneticsTohoku University School of MedicineSendaiJapan
| | - Bruce D. Gelb
- Mindich Child Health and Development InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pediatrics and GeneticsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
13
|
Fasano G, Muto V, Radio FC, Venditti M, Mosaddeghzadeh N, Coppola S, Paradisi G, Zara E, Bazgir F, Ziegler A, Chillemi G, Bertuccini L, Tinari A, Vetro A, Pantaleoni F, Pizzi S, Conti LA, Petrini S, Bruselles A, Prandi IG, Mancini C, Chandramouli B, Barth M, Bris C, Milani D, Selicorni A, Macchiaiolo M, Gonfiantini MV, Bartuli A, Mariani R, Curry CJ, Guerrini R, Slavotinek A, Iascone M, Dallapiccola B, Ahmadian MR, Lauri A, Tartaglia M. Dominant ARF3 variants disrupt Golgi integrity and cause a neurodevelopmental disorder recapitulated in zebrafish. Nat Commun 2022; 13:6841. [PMID: 36369169 PMCID: PMC9652361 DOI: 10.1038/s41467-022-34354-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Vesicle biogenesis, trafficking and signaling via Endoplasmic reticulum-Golgi network support essential developmental processes and their disruption lead to neurodevelopmental disorders and neurodegeneration. We report that de novo missense variants in ARF3, encoding a small GTPase regulating Golgi dynamics, cause a developmental disease in humans impairing nervous system and skeletal formation. Microcephaly-associated ARF3 variants affect residues within the guanine nucleotide binding pocket and variably perturb protein stability and GTP/GDP binding. Functional analysis demonstrates variably disruptive consequences of ARF3 variants on Golgi morphology, vesicles assembly and trafficking. Disease modeling in zebrafish validates further the dominant behavior of the mutants and their differential impact on brain and body plan formation, recapitulating the variable disease expression. In-depth in vivo analyses traces back impaired neural precursors' proliferation and planar cell polarity-dependent cell movements as the earliest detectable effects. Our findings document a key role of ARF3 in Golgi function and demonstrate its pleiotropic impact on development.
Collapse
Affiliation(s)
- Giulia Fasano
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Valentina Muto
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Francesca Clementina Radio
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Martina Venditti
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Niloufar Mosaddeghzadeh
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Simona Coppola
- grid.416651.10000 0000 9120 6856National Center for Rare Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Graziamaria Paradisi
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy ,grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Erika Zara
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy ,grid.7841.aDepartment of Biology and Biotechnology “Charles Darwin”, Università “Sapienza”, Rome, 00185 Italy
| | - Farhad Bazgir
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alban Ziegler
- grid.7252.20000 0001 2248 3363UFR Santé de l’Université d’Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France ,grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Giovanni Chillemi
- grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy ,grid.5326.20000 0001 1940 4177Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Centro Nazionale delle Ricerche, 70126 Bari, Italy
| | - Lucia Bertuccini
- grid.416651.10000 0000 9120 6856Servizio grandi strumentazioni e core facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonella Tinari
- grid.416651.10000 0000 9120 6856Centro di riferimento per la medicina di genere, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Annalisa Vetro
- grid.8404.80000 0004 1757 2304Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children’s Hospital, University of Florence, 50139 Florence, Italy
| | - Francesca Pantaleoni
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Simone Pizzi
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Libenzio Adrian Conti
- grid.414603.4Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Stefania Petrini
- grid.414603.4Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Alessandro Bruselles
- grid.416651.10000 0000 9120 6856Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Ingrid Guarnetti Prandi
- grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Cecilia Mancini
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Balasubramanian Chandramouli
- grid.431603.30000 0004 1757 1950Super Computing Applications and Innovation, CINECA, 40033 Casalecchio di Reno, Italy
| | - Magalie Barth
- grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Céline Bris
- grid.7252.20000 0001 2248 3363UFR Santé de l’Université d’Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France ,grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Donatella Milani
- grid.414818.00000 0004 1757 8749Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Angelo Selicorni
- grid.512106.1Mariani Center for Fragile Children Pediatric Unit, Azienda Socio Sanitaria Territoriale Lariana, 22100 Como, Italy
| | - Marina Macchiaiolo
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Michaela V. Gonfiantini
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Andrea Bartuli
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Riccardo Mariani
- grid.414603.4Department of Laboratories Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Cynthia J. Curry
- grid.266102.10000 0001 2297 6811Genetic Medicine, Dept of Pediatrics, University of California San Francisco, Ca, Fresno, Ca, San Francisco, CA 94143 USA
| | - Renzo Guerrini
- grid.8404.80000 0004 1757 2304Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children’s Hospital, University of Florence, 50139 Florence, Italy
| | - Anne Slavotinek
- grid.266102.10000 0001 2297 6811Genetic Medicine, Dept of Pediatrics, University of California San Francisco, Ca, Fresno, Ca, San Francisco, CA 94143 USA
| | - Maria Iascone
- grid.460094.f0000 0004 1757 8431Medical Genetics, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Bruno Dallapiccola
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Mohammad Reza Ahmadian
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Antonella Lauri
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Marco Tartaglia
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| |
Collapse
|
14
|
Nandi S, Chennappan S, Andrasch Y, Fidan M, Engler M, Ahmad M, Tuckermann JP, Zenker M, Cirstea IC. Increased osteoclastogenesis contributes to bone loss in the Costello syndrome Hras G12V mouse model. Front Cell Dev Biol 2022; 10:1000575. [PMID: 36330334 PMCID: PMC9624175 DOI: 10.3389/fcell.2022.1000575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
RAS GTPases are ubiquitous GDP/GTP-binding proteins that function as molecular switches in cellular signalling and control numerous signalling pathways and biological processes. Pathogenic mutations in RAS genes severely affect cellular homeostasis, leading to cancer when occurring in somatic cells and developmental disorders when the germline is affected. These disorders are generally termed as RASopathies and among them Costello syndrome (CS) is a distinctive entity that is caused by specific HRAS germline mutations. The majority of these mutations affect residues 12 and 13, the same sites as somatic oncogenic HRAS mutations. The hallmarks of the disease include congenital cardiac anomalies, impaired thriving and growth, neurocognitive impairments, distinctive craniofacial anomalies, and susceptibility to cancer. Adult patients often present signs of premature aging including reduced bone mineral density and osteoporosis. Using a CS mouse model harbouring a Hras G12V germline mutation, we aimed at determining whether this model recapitulates the patients’ bone phenotype and which bone cells are driving the phenotype when mutated. Our data revealed that Hras G12V mutation induces bone loss in mice at certain ages. In addition, we identified that bone loss correlated with an increased number of osteoclasts in vivo and Hras G12V mutations increased osteoclastogenesis in vitro. Last, but not least, mutant osteoclast differentiation was reduced by treatment in vitro with MEK and PI3K inhibitors, respectively. These results indicate that Hras is a novel regulator of bone homeostasis and an increased osteoclastogenesis due to Hras G12V mutation contributes to bone loss in the Costello syndrome.
Collapse
Affiliation(s)
- Sayantan Nandi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | | | - Yannik Andrasch
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Miray Fidan
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Melanie Engler
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Mubashir Ahmad
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Jan P. Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
- *Correspondence: Ion Cristian Cirstea,
| |
Collapse
|
15
|
Bajia D, Bottani E, Derwich K. Effects of Noonan Syndrome-Germline Mutations on Mitochondria and Energy Metabolism. Cells 2022; 11:cells11193099. [PMID: 36231062 PMCID: PMC9563972 DOI: 10.3390/cells11193099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.
Collapse
Affiliation(s)
- Donald Bajia
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Piazzale L. A. Scuro 10, 37134 Verona, Italy
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| |
Collapse
|
16
|
Wingbermühle E, Roelofs RL, Oomens W, Kramer J, Draaisma JMT, Leenders E, Kleefstra T, Kessels RPC, Egger JIM. Cognitive Phenotype and Psychopathology in Noonan Syndrome Spectrum Disorders through Various Ras/MAPK Pathway Associated Gene Variants. J Clin Med 2022; 11:jcm11164735. [PMID: 36012976 PMCID: PMC9410383 DOI: 10.3390/jcm11164735] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Cognitive difficulties are argued to be common in patients with Noonan syndrome spectrum disorders (NSSDs), but findings are based on studies in which patients with variants in PTPN11 (prevalence ~50%) were overrepresented. The current study, using a structured clinical approach, describes the cognitive phenotype and psychopathology of 100 patients (aged 6 to 61 years) with nine different gene variants in the Ras/MAPK pathway underlying NSSDs (PTPN11n = 61, PTPN11 Noonan syndrome with multiple lentigines n = 3, SOS1n = 14, KRASn = 7, LZTR1n = 5, RAF1n = 4, SHOC2n = 2, CBLn = 2, SOS2n = 2). After weighted assessment and bootstrapping of the results of individual neuropsychological assessments and measures of psychopathology, cognitive performances in most variant groups were within the ranges of expectation. IQs were significantly lower in patients with variants in PTPN11, KRAS, RAF1, and SHOC2, but no specific cognitive impairments were found. The performances of younger participants (<16 years of age) did not differ from those of adults. Alexithymia and internalizing problems were more frequent in patients with variants in PTPN11 and SOS1, while PTPN11 patients also showed higher levels of externalizing problems. These results stress the need to take intelligence into account when interpreting lower cognitive performances in individual neuropsychological assessments, which is crucial for an adequate understanding and guidance of patients with NSSDs.
Collapse
Affiliation(s)
- Ellen Wingbermühle
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Correspondence:
| | - Renée L. Roelofs
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Wouter Oomens
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
| | - Jennifer Kramer
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
| | - Jos M. T. Draaisma
- Department of Pediatrics, Amalia Children’s Hospital, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Erika Leenders
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Roy P. C. Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Center of Excellence for Korsakoff and Alcohol-Related Cognitive Disorders, Vincent van Gogh Institute for Psychiatry, 5803 DN Venray, The Netherlands
- Department of Medical Psychology and Radboudumc Alzheimer Center, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Jos I. M. Egger
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, 5803 DM Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 GD Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Center of Excellence for Korsakoff and Alcohol-Related Cognitive Disorders, Vincent van Gogh Institute for Psychiatry, 5803 DN Venray, The Netherlands
| |
Collapse
|
17
|
Genomic and Epigenomic Landscape of Juvenile Myelomonocytic Leukemia. Cancers (Basel) 2022; 14:cancers14051335. [PMID: 35267643 PMCID: PMC8909150 DOI: 10.3390/cancers14051335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm characterized by the constitutive activation of the RAS pathway. In spite of the recent progresses in the molecular characterization of JMML, this disease is still a clinical challenge due to its heterogeneity, difficult diagnosis, poor prognosis, and the lack of curative treatment options other than hematopoietic stem cell transplantation (HSCT). In this review, we will provide a detailed overview of the genetic and epigenetic alterations occurring in JMML, and discuss their clinical relevance in terms of disease prognosis and risk of relapse after HSCT. We will also present the most recent advances on novel preclinical and clinical therapeutic approaches directed against JMML molecular targets. Finally, we will outline future research perspectives to further explore the oncogenic mechanism driving JMML leukemogenesis and progression, with special attention to the application of single-cell next-generation sequencing technologies. Abstract Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm of early childhood. Most of JMML patients experience an aggressive clinical course of the disease and require hematopoietic stem cell transplantation, which is currently the only curative treatment. JMML is characterized by RAS signaling hyperactivation, which is mainly driven by mutations in one of five genes of the RAS pathway, including PTPN11, KRAS, NRAS, NF1, and CBL. These driving mutations define different disease subtypes with specific clinico-biological features. Secondary mutations affecting other genes inside and outside the RAS pathway contribute to JMML pathogenesis and are associated with a poorer prognosis. In addition to these genetic alterations, JMML commonly presents aberrant epigenetic profiles that strongly correlate with the clinical outcome of the patients. This observation led to the recent publication of an international JMML stratification consensus, which defines three JMML clinical groups based on DNA methylation status. Although the characterization of the genomic and epigenomic landscapes in JMML has significantly contributed to better understand the molecular mechanisms driving the disease, our knowledge on JMML origin, cell identity, and intratumor and interpatient heterogeneity is still scarce. The application of new single-cell sequencing technologies will be critical to address these questions in the future.
Collapse
|
18
|
van Engelen N, Diets I, Bresters D, van den Bergen JC, Vrancken AF, Kuiper RP, Jongmans MC. In Response to: Pediatric Myelodysplastic Syndrome with Germline RRAS Mutation: Expanding the Phenotype of RASopathies. J Pediatr Hematol Oncol 2022; 44:69. [PMID: 34935735 PMCID: PMC8876415 DOI: 10.1097/mph.0000000000002373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Illja Diets
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | | | | - Marjolijn C.J. Jongmans
- Princess Máxima Center for Pediatric Oncology
- Department of Genetics, University Medical Center Utrecht, Utrecht
| |
Collapse
|
19
|
Classical RAS proteins are not essential for paradoxical ERK activation induced by RAF inhibitors. Proc Natl Acad Sci U S A 2022; 119:2113491119. [PMID: 35091470 PMCID: PMC8812530 DOI: 10.1073/pnas.2113491119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 11/21/2022] Open
Abstract
RAF inhibitors unexpectedly induce ERK activation in normal and oncogenic RAS tumor cells, making them unsuitable for treating RAS-driven cancers. The precise mechanism of this paradox is not fully understood but is believed to be RAS dependent. In this study, we discovered that classical RAS proteins are not essential for RAF inhibitor-induced ERK activation in H/N/KRAS-less mouse embryonic fibroblasts. We further showed that the MRAS/SHOC2 complex is required for the classical RAS-independent paradoxical ERK activation. Our findings provide new insights into the mechanism of paradoxical ERK activation by RAF inhibitors, and they have important therapeutic implications for developing effective RAF inhibitors. RAF inhibitors unexpectedly induce ERK signaling in normal and tumor cells with elevated RAS activity. Paradoxical activation is believed to be RAS dependent. In this study, we showed that LY3009120, a pan-RAF inhibitor, can unexpectedly cause paradoxical ERK activation in KRASG12C-dependent lung cancer cell lines, when KRAS is inhibited by ARS1620, a KRASG12C inhibitor. Using H/N/KRAS-less mouse embryonic fibroblasts, we discovered that classical RAS proteins are not essential for RAF inhibitor-induced paradoxical ERK signaling. In their absence, RAF inhibitors can induce ERK phosphorylation, ERK target gene transcription, and cell proliferation. We further showed that the MRAS/SHOC2 complex is required for this process. This study highlights the complexity of the allosteric RAF regulation by RAF inhibitors, and the importance of other RAS-related proteins in this process.
Collapse
|
20
|
Abstract
The RASopathies are a group of disorders caused by a germline mutation in one of the genes encoding a component of the RAS/MAPK pathway. These disorders, including neurofibromatosis type 1, Noonan syndrome, cardiofaciocutaneous syndrome, Costello syndrome and Legius syndrome, among others, have overlapping clinical features due to RAS/MAPK dysfunction. Although several of the RASopathies are very rare, collectively, these disorders are relatively common. In this Review, we discuss the pathogenesis of the RASopathy-associated genetic variants and the knowledge gained about RAS/MAPK signaling that resulted from studying RASopathies. We also describe the cell and animal models of the RASopathies and explore emerging RASopathy genes. Preclinical and clinical experiences with targeted agents as therapeutics for RASopathies are also discussed. Finally, we review how the recently developed drugs targeting RAS/MAPK-driven malignancies, such as inhibitors of RAS activation, direct RAS inhibitors and RAS/MAPK pathway inhibitors, might be leveraged for patients with RASopathies.
Collapse
Affiliation(s)
- Katie E Hebron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Edjay Ralph Hernandez
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Marielle E Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Wintering A, Dvorak CC, Stieglitz E, Loh ML. Juvenile myelomonocytic leukemia in the molecular era: a clinician's guide to diagnosis, risk stratification, and treatment. Blood Adv 2021; 5:4783-4793. [PMID: 34525182 PMCID: PMC8759142 DOI: 10.1182/bloodadvances.2021005117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/03/2021] [Indexed: 12/03/2022] Open
Abstract
Juvenile myelomonocytic leukemia is an overlapping myeloproliferative and myelodysplastic disorder of early childhood . It is associated with a spectrum of diverse outcomes ranging from spontaneous resolution in rare patients to transformation to acute myeloid leukemia in others that is generally fatal. This unpredictable clinical course, along with initially descriptive diagnostic criteria, led to decades of productive international research. Next-generation sequencing now permits more accurate molecular diagnoses in nearly all patients. However, curative treatment is still reliant on allogeneic hematopoietic cell transplantation for most patients, and additional advances will be required to improve risk stratification algorithms that distinguish those that can be observed expectantly from others who require swift hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Astrid Wintering
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
| | - Christopher C. Dvorak
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA; and
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| |
Collapse
|
22
|
Weinstock NI, Sadler L. The RRAS2 pathogenic variant p.Q72L produces severe Noonan syndrome with hydrocephalus: A case report. Am J Med Genet A 2021; 188:364-368. [PMID: 34648682 DOI: 10.1002/ajmg.a.62523] [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: 02/28/2021] [Revised: 08/29/2021] [Accepted: 09/10/2021] [Indexed: 11/11/2022]
Abstract
Noonan syndrome (NS) is the most common disease among RASopathies, characterized by short stature, distinctive facial features, congenital cardiac defects, and variable developmental delay. NS rarely presents with overt neurologic manifestations, in particular hydrocephalus. Recent evidence suggests that pathogenic variants in the gene RRAS2 are a rare cause of NS. Specifically, an RRAS2 pathogenic variant, p.Q72L, may be particularly severe, manifesting with lethal neurologic findings. Here, we report a NS patient with documented p.Q72L variant in RRAS2. The patient was identified in utero to have hydrocephalus and a Dandy Walker malformation. Postnatal examination revealed multiple dysmorphic features, some reminiscent of NS including low-set posteriorly rotated ears, redundant nuchal skin, widely spaced nipples, and cryptorchidism. Despite suspicion of NS, results of a 14-gene Noonan syndrome panel (Invitae) were negative. Follow-up rapid whole exome sequencing revealed a de novo p.Q72L variant in RRAS2, a poorly studied gene recently identified as a cause of NS. The patient herein reported brings to three the total number of cases reported with the RRAS2 p.Q72L pathogenic variant. All three documented patients presented with a particularly fulminant course of NS, which included hydrocephalus. RRAS2, specifically p.Q72L, should be considered in severe NS cases with neurologic manifestations.
Collapse
Affiliation(s)
- Nadav I Weinstock
- Division of Genetics, Department of Pediatrics, Oishei Children's Hospital, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, New York, USA
| | - Laurie Sadler
- Division of Genetics, Department of Pediatrics, Oishei Children's Hospital, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, New York, USA
| |
Collapse
|
23
|
Weber SM, Carroll SL. The Role of R-Ras Proteins in Normal and Pathologic Migration and Morphologic Change. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1499-1510. [PMID: 34111428 PMCID: PMC8420862 DOI: 10.1016/j.ajpath.2021.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022]
Abstract
The contributions that the R-Ras subfamily [R-Ras, R-Ras2/teratocarcinoma 21 (TC21), and M-Ras] of small GTP-binding proteins make to normal and aberrant cellular functions have historically been poorly understood. However, this has begun to change with the realization that all three R-Ras subfamily members are occasionally mutated in Noonan syndrome (NS), a RASopathy characterized by the development of hematopoietic neoplasms and abnormalities affecting the immune, cardiovascular, and nervous systems. Consistent with the abnormalities seen in NS, a host of new studies have implicated R-Ras proteins in physiological and pathologic changes in cellular morphology, adhesion, and migration in the cardiovascular, immune, and nervous systems. These changes include regulating the migration and homing of mature and immature immune cells, vascular stabilization, clotting, and axonal and dendritic outgrowth during nervous system development. Dysregulated R-Ras signaling has also been linked to the pathogenesis of cardiovascular disease, intellectual disabilities, and human cancers. This review discusses the structure and regulation of R-Ras proteins and our current understanding of the signaling pathways that they regulate. It explores the phenotype of NS patients and their implications for the R-Ras subfamily functions. Next, it covers recent discoveries regarding physiological and pathologic R-Ras functions in key organ systems. Finally, it discusses how R-Ras signaling is dysregulated in cancers and mechanisms by which this may promote neoplasia.
Collapse
Affiliation(s)
- Shannon M Weber
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
24
|
Motta M, Sagi-Dain L, Krumbach OHF, Hahn A, Peleg A, German A, Lissewski C, Coppola S, Pantaleoni F, Kocherscheid L, Altmüller F, Schanze D, Logeswaran T, Chahrokh-Zadeh S, Munzig A, Nakhaei-Rad S, Cavé H, Ahmadian MR, Tartaglia M, Zenker M. Activating MRAS mutations cause Noonan syndrome associated with hypertrophic cardiomyopathy. Hum Mol Genet 2021; 29:1772-1783. [PMID: 31108500 DOI: 10.1093/hmg/ddz108] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
The RASopathies are a group of genetic syndromes caused by upregulated RAS signaling. Noonan syndrome (NS), the most common entity among the RASopathies, is characterized mainly by short stature, cardiac anomalies and distinctive facial features. Mutations in multiple RAS-MAPK pathway-related genes have been associated with NS and related phenotypes. We describe two unrelated patients presenting with hypertrophic cardiomyopathy (HCM) and dysmorphic features suggestive of NS. One of them died in the neonatal period because of cardiac failure. Targeted sequencing revealed de novo MRAS variants, c.203C > T (p.Thr68Ile) and c.67G > C (p.Gly23Arg) as causative events. MRAS has only recently been related to NS based on the observation of two unrelated affected individuals with de novo variants involving the same codons here found mutated. Gly23 and Thr68 are highly conserved residues, and the corresponding codons are known hotspots for RASopathy-associated mutations in other RAS proteins. Functional analyses documented high level of activation of MRAS mutants due to impaired GTPase activity, which was associated with constitutive plasma membrane targeting, prolonged localization in non-raft microdomains, enhanced binding to PPP1CB and SHOC2 protein, and variably increased MAPK and PI3K-AKT activation. This report provides additional evidence that a narrow spectrum of activating mutations in MRAS represents another rare cause of NS, and that MRAS has to be counted among the RASopathy genes predisposing to HCM. Moreover, our findings further emphasize the relevance of the MRAS-SHOC2-PPP1CB axis in the control of MAPK signaling, and the contribution of both MAPK and PI3K-AKT pathways in MRAS functional upregulation.
Collapse
Affiliation(s)
- Marialetizia Motta
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Lena Sagi-Dain
- The Human Genetic institute, Carmel Medical Center, Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel
| | - Oliver H F Krumbach
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Andreas Hahn
- Department of Child Neurology, University Hospital, Gießen, Germany
| | - Amir Peleg
- The Human Genetic institute, Carmel Medical Center, Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel
| | - Alina German
- Pediatric Department, Bnai-Zion Medical Center and Clalit Health Maintenance Organization, Haifa, Israel
| | | | - Simona Coppola
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pantaleoni
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | | | - Denny Schanze
- Institute of Human Genetics, University Hospital, Magdeburg, Germany
| | | | | | - Anna Munzig
- Center of Human Genetics and Laboratory Diagnostics, Martinsried, Germany
| | - Saeideh Nakhaei-Rad
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Hélène Cavé
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Robert Debré, Département de Génétique, 75019 Paris, France.,INSERM UMR 1131, Institut de Recherche Saint-Louis, Université de Paris, 75010 Paris, France
| | - Mohammad R Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Magdeburg, Germany
| |
Collapse
|
25
|
Longo JF, Carroll SL. The RASopathies: Biology, genetics and therapeutic options. Adv Cancer Res 2021; 153:305-341. [PMID: 35101235 DOI: 10.1016/bs.acr.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The RASopathies are a group of genetic diseases in which the Ras/MAPK signaling pathway is inappropriately activated as a result of mutations in genes encoding proteins within this pathway. As their causative mutations have been identified, this group of diseases has expanded to include neurofibromatosis type 1 (NF1), Legius syndrome, Noonan syndrome, CBL syndrome, Noonan syndrome-like disorder with loose anagen hair, Noonan syndrome with multiple lentigines, Costello syndrome, cardiofaciocutaneous syndrome, gingival fibromatosis and capillary malformation-arteriovenous malformation syndrome. Many of these genetic disorders share clinical features in common such as abnormal facies, short stature, varying degrees of cognitive impairment, cardiovascular abnormalities, skeletal abnormalities and a predisposition to develop benign and malignant neoplasms. Others are more dissimilar, even though their mutations are in the same gene that is mutated in a different RASopathy. Here, we describe the clinical features of each RASopathy and contrast them with the other RASopathies. We discuss the genetics of these disorders, including the causative mutations for each RASopathy, the impact that these mutations have on the function of an individual protein and how this dysregulates the Ras/MAPK signaling pathway. As several of these individual disorders are genetically heterogeneous, we also consider the different genes that can be mutated to produce disease with the same phenotype. We also discuss how our growing understanding of dysregulated Ras/MAPK signaling had led to the development of new therapeutic agents and what work will be critically important in the future to improve the lives of patients with RASopathies.
Collapse
Affiliation(s)
- Jody Fromm Longo
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
26
|
Mariani RA, Jennings L, Zhang S, Bhat R, Gong S. Morphologic and Immunophenotypic Differences in Juvenile Myelomonocytic Leukemias With CBL and Other Canonical RAS-pathway Gene Mutations: A Single Institutional Experience. J Pediatr Hematol Oncol 2021; 43:e819-e825. [PMID: 33769390 DOI: 10.1097/mph.0000000000002149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022]
Abstract
The diagnostic criteria for juvenile myelomonocytic leukemia have recently been revised to include clinical findings and RAS-pathway gene mutations per the 2016 World Health Organization Classification of Tumors of Hematopoietic and Lymphoid Tissues. Differing clinical behaviors have been observed in cases with CBL versus other RAS-pathway gene (RAS-p) mutations, notably the patients with CBL mutations can be self-limiting with spontaneous resolution. Additional clinical characteristics and histopathologic findings between these subsets are less well-described. We performed a retrospective search and identified cases with either CBL or RAS-p mutations, as per targeted and/or massively parallel sequencing. Eight patients had sufficient material for review, including cytogenetic studies and peripheral blood, bone marrow aspirate, and/or biopsy with flow cytometry analyses. Three patients showed CBL mutations and lower percentages of hemoglobin F and peripheral blood absolute monocyte counts, lesser degrees of leukocytosis compared with the RAS-p cohort, and normal megakaryocyte morphology and myeloblast immunophenotypes. Two of these patients were managed with observation only and experienced resolution of their disease. The patients with RAS-p mutations had severe thrombocytopenia, moderate to severe anemia, and experienced variable clinical outcomes. Abnormal megakaryocyte morphology and decreased numbers of megakaryocytes were seen in cases with RAS-p mutations. In addition, 3 of 4 cases with flow cytometry data demonstrated aberrant CD7 expression in myeloblasts. Our study is the first to identify morphologic and immunophenotypic differences between juvenile myelomonocytic leukemia cases with CBL or RAS-p mutations, and further supports previous reports of significantly different clinical behaviors between these subsets of patients.
Collapse
Affiliation(s)
| | | | - Shanxiang Zhang
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN
| | - Rukhmi Bhat
- Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital, Chicago, IL
| | | |
Collapse
|
27
|
Mayerhofer C, Niemeyer CM, Flotho C. Current Treatment of Juvenile Myelomonocytic Leukemia. J Clin Med 2021; 10:3084. [PMID: 34300250 PMCID: PMC8305558 DOI: 10.3390/jcm10143084] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric leukemia characterized by mutations in five canonical RAS pathway genes. The diagnosis is made by typical clinical and hematological findings associated with a compatible mutation. Although this is sufficient for clinical decision-making in most JMML cases, more in-depth analysis can include DNA methylation class and panel sequencing analysis for secondary mutations. NRAS-initiated JMML is heterogeneous and adequate management ranges from watchful waiting to allogeneic hematopoietic stem cell transplantation (HSCT). Upfront azacitidine in KRAS patients can achieve long-term remissions without HSCT; if HSCT is required, a less toxic preparative regimen is recommended. Germline CBL patients often experience spontaneous resolution of the leukemia or exhibit stable mixed chimerism after HSCT. JMML driven by PTPN11 or NF1 is often rapidly progressive, requires swift HSCT and may benefit from pretransplant therapy with azacitidine. Because graft-versus-leukemia alloimmunity is central to cure high risk patients, the immunosuppressive regimen should be discontinued early after HSCT.
Collapse
Affiliation(s)
- Christina Mayerhofer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
| | - Charlotte M. Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| |
Collapse
|
28
|
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: 0] [Impact Index Per Article: 0] [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
|
29
|
Osswald L, Hamarsheh S, Uhl FM, Andrieux G, Klein C, Dierks C, Duquesne S, Braun LM, Schmitt-Graeff A, Duyster J, Boerries M, Brummer T, Zeiser R. Oncogenic KrasG12D Activation in the Nonhematopoietic Bone Marrow Microenvironment Causes Myelodysplastic Syndrome in Mice. Mol Cancer Res 2021; 19:1596-1608. [PMID: 34088868 DOI: 10.1158/1541-7786.mcr-20-0275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/10/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022]
Abstract
The bone marrow microenvironment (BMME) is key player in regulation and maintenance of hematopoiesis. Oncogenic RAS mutations, causing constitutive activation of multiple tumor-promoting pathways, are frequently found in human cancer. So far in hematologic malignancies, RAS mutations have only been reported to occur in hematopoietic cells. In this study, we investigated the effect of oncogenic Kras expression in the BMME in a chimeric mouse model. We observed that an activating mutation of Kras in the nonhematopoietic system leads to a phenotype resembling myelodysplastic syndrome (MDS) characterized by peripheral cytopenia, marked dysplasia within the myeloid lineage as well as impaired proliferation and differentiation capacity of hematopoietic stem and progenitor cells. The phenotypic changes could be reverted when the BM was re-isolated and transferred into healthy recipients, indicating that the KrasG12D -activation in the nonhematopoietic BMME was essential for the MDS phenotype. Gene expression analysis of sorted nonhematopoietic BM niche cells from KrasG12D mice revealed upregulation of multiple inflammation-related genes including IL1-superfamily members (Il1α, Il1β, Il1f9) and the NLPR3 inflammasome. Thus, pro-inflammatory IL1-signaling in the BMME may contribute to MDS development. Our findings show that a single genetic change in the nonhematopoietic BMME can cause an MDS phenotype. Oncogenic Kras activation leads to pro-inflammatory signaling in the BMME which impairs HSPCs function. IMPLICATIONS: These findings may help to identify new therapeutic targets for MDS.
Collapse
Affiliation(s)
- Lena Osswald
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shaima'a Hamarsheh
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Maria Uhl
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudius Klein
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christine Dierks
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Duquesne
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas M Braun
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Justus Duyster
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tilman Brummer
- German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center- University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Zhang H, Cao X, Wang J, Li Q, Zhao Y, Jin X. LZTR1: A promising adaptor of the CUL3 family. Oncol Lett 2021; 22:564. [PMID: 34113392 PMCID: PMC8185703 DOI: 10.3892/ol.2021.12825] [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: 10/22/2020] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
The study of the disorders of ubiquitin-mediated proteasomal degradation may unravel the molecular basis of human diseases, such as cancer (prostate cancer, lung cancer and liver cancer, etc.) and nervous system disease (Parkinson's disease, Alzheimer's disease and Huntington's disease, etc.) and help in the design of new therapeutic methods. Leucine zipper-like transcription regulator 1 (LZTR1) is an important substrate recognition subunit of cullin-RING E3 ligase that plays an important role in the regulation of cellular functions. Mutations in LZTR1 and dysregulation of associated downstream signaling pathways contribute to the pathogenesis of Noonan syndrome (NS), glioblastoma and chronic myeloid leukemia. Understanding the molecular mechanism of the normal function of LZTR1 is thus critical for its eventual therapeutic targeting. In the present review, the structure and function of LZTR1 are described. Moreover, recent advances in the current knowledge of the functions of LZTR1 in NS, glioblastoma (GBM), chronic myeloid leukemia (CML) and schwannomatosis and the influence of LZTR1 mutations are also discussed, providing insight into how LZTR1 may be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xinyi Cao
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jian Wang
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Qian Li
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yiting Zhao
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology; Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
31
|
Lauri A, Fasano G, Venditti M, Dallapiccola B, Tartaglia M. In vivo Functional Genomics for Undiagnosed Patients: The Impact of Small GTPases Signaling Dysregulation at Pan-Embryo Developmental Scale. Front Cell Dev Biol 2021; 9:642235. [PMID: 34124035 PMCID: PMC8194860 DOI: 10.3389/fcell.2021.642235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
While individually rare, disorders affecting development collectively represent a substantial clinical, psychological, and socioeconomic burden to patients, families, and society. Insights into the molecular mechanisms underlying these disorders are required to speed up diagnosis, improve counseling, and optimize management toward targeted therapies. Genome sequencing is now unveiling previously unexplored genetic variations in undiagnosed patients, which require functional validation and mechanistic understanding, particularly when dealing with novel nosologic entities. Functional perturbations of key regulators acting on signals' intersections of evolutionarily conserved pathways in these pathological conditions hinder the fine balance between various developmental inputs governing morphogenesis and homeostasis. However, the distinct mechanisms by which these hubs orchestrate pathways to ensure the developmental coordinates are poorly understood. Integrative functional genomics implementing quantitative in vivo models of embryogenesis with subcellular precision in whole organisms contribute to answering these questions. Here, we review the current knowledge on genes and mechanisms critically involved in developmental syndromes and pediatric cancers, revealed by genomic sequencing and in vivo models such as insects, worms and fish. We focus on the monomeric GTPases of the RAS superfamily and their influence on crucial developmental signals and processes. We next discuss the effectiveness of exponentially growing functional assays employing tractable models to identify regulatory crossroads. Unprecedented sophistications are now possible in zebrafish, i.e., genome editing with single-nucleotide precision, nanoimaging, highly resolved recording of multiple small molecules activity, and simultaneous monitoring of brain circuits and complex behavioral response. These assets permit accurate real-time reporting of dynamic small GTPases-controlled processes in entire organisms, owning the potential to tackle rare disease mechanisms.
Collapse
Affiliation(s)
- Antonella Lauri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | | | | | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
32
|
Pediatric Myelodysplastic Syndrome With Germline RRAS Mutation: Expanding the Phenotype of RASopathies. J Pediatr Hematol Oncol 2021; 43:e517-e520. [PMID: 32815881 DOI: 10.1097/mph.0000000000001910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 07/10/2020] [Indexed: 12/31/2022]
Abstract
The RAS/mitogen-activated protein kinase pathway plays a significant role in cell cycle regulation. Germline mutation of this pathway leads to overlapping genetic disorders, RASopathies, and is also an important component of tumorigenesis. Here we describe a rare case of myelodysplastic syndrome with monosomy 7 in a pediatric patient with a germline RRAS mutation. RRAS mutations have been implicated in the development of juvenile myelomonocytic leukemia, but our case suggests RRAS mutations display a broader malignant potential. Our case supports the recommendation that genetic testing should include RRAS in suspected RASopathy patients and if identified, these patients undergo surveillance for hematologic malignancy.
Collapse
|
33
|
Palomo L, Acha P, Solé F. Genetic Aspects of Myelodysplastic/Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13092120. [PMID: 33925681 PMCID: PMC8124412 DOI: 10.3390/cancers13092120] [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: 03/26/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are clonal myeloid neoplasms characterized, at the time of their presentation, by the simultaneous presence of both myelodysplastic and myeloproliferative features. In MDS/MPN, the karyotype is often normal but mutations in genes that are common across myeloid neoplasms can be detected in a high proportion of cases by targeted sequencing. In this review, we intend to summarize the main genetic findings across all MDS/MPN overlap syndromes and discuss their relevance in the management of patients. Abstract Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are myeloid neoplasms characterized by the presentation of overlapping features from both myelodysplastic syndromes and myeloproliferative neoplasms. Although the classification of MDS/MPN relies largely on clinical features and peripheral blood and bone marrow morphology, studies have demonstrated that a large proportion of patients (~90%) with this disease harbor somatic mutations in a group of genes that are common across myeloid neoplasms. These mutations play a role in the clinical heterogeneity of these diseases and their clinical evolution. Nevertheless, none of them is specific to MDS/MPN and current diagnostic criteria do not include molecular data. Even when such alterations can be helpful for differential diagnosis, they should not be used alone as proof of neoplasia because some of these mutations may also occur in healthy older people. Here, we intend to review the main genetic findings across all MDS/MPN overlap syndromes and discuss their relevance in the management of the patients.
Collapse
Affiliation(s)
- Laura Palomo
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (L.P.); (P.A.)
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Pamela Acha
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (L.P.); (P.A.)
| | - Francesc Solé
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (L.P.); (P.A.)
- Correspondence: ; Tel.: +34-93-557-2806
| |
Collapse
|
34
|
Greenmyer JR, Kohorst M. Pediatric Neoplasms Presenting with Monocytosis. Curr Hematol Malig Rep 2021; 16:235-246. [PMID: 33630234 DOI: 10.1007/s11899-021-00611-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Juvenile myelomonocytic leukemia (JMML) is a rare but severe pediatric neoplasm with hematopoietic stem cell transplant as its only established curative option. The development of targeted therapeutics for JMML is being guided by an understanding of the pathobiology of this condition. Here, we review JMML with an emphasis on genetics in order to (i) demonstrate the relationship between JMML genotype and clinical phenotype and (ii) explore potential genetic targets of novel JMML therapies. RECENT FINDINGS DNA hypermethylation studies have demonstrated consistently that methylation is related to disease severity. Increasing understanding of methylation in JMML may open the door to novel therapies, such as DNA methyltransferase inhibitors. The PI3K/AKT/MTOR, JAK/STAT, and RAF/MEK/ERK pathways are being investigated as therapeutic targets for JMML. Future therapy for JMML will be driven by an increased understanding of pathobiology. Targeted therapeutic approaches hold potential for improving outcomes in patients with JMML.
Collapse
Affiliation(s)
| | - Mira Kohorst
- Pediatric Hematology and Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
35
|
Klco JM, Mullighan CG. Advances in germline predisposition to acute leukaemias and myeloid neoplasms. Nat Rev Cancer 2021; 21:122-137. [PMID: 33328584 PMCID: PMC8404376 DOI: 10.1038/s41568-020-00315-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Although much work has focused on the elucidation of somatic alterations that drive the development of acute leukaemias and other haematopoietic diseases, it has become increasingly recognized that germline mutations are common in many of these neoplasms. In this Review, we highlight the different genetic pathways impacted by germline mutations that can ultimately lead to the development of familial and sporadic haematological malignancies, including acute lymphoblastic leukaemia, acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS). Many of the genes disrupted by somatic mutations in these diseases (for example, TP53, RUNX1, IKZF1 and ETV6) are the same as those that harbour germline mutations in children and adolescents who develop these malignancies. Moreover, the presumption that familial leukaemias only present in childhood is no longer true, in large part due to the numerous studies demonstrating germline DDX41 mutations in adults with MDS and AML. Lastly, we highlight how different cooperating events can influence the ultimate phenotype in these different familial leukaemia syndromes.
Collapse
Affiliation(s)
- Jeffery M Klco
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Charles G Mullighan
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
36
|
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
|
37
|
Lores J, Prada CE, Ramírez-Montaño D, Nastasi-Catanese JA, Pachajoa H. Clinical and molecular analysis of 26 individuals with Noonan syndrome in a reference institution in Colombia. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:1042-1051. [PMID: 33300679 DOI: 10.1002/ajmg.c.31869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 11/06/2022]
Abstract
Our aim was to characterize the phenotype and genotype of individuals with Noonan syndrome in Colombia. There are published cohorts of Noonan individuals from several countries in Latin America including Brazil, Chile, and Argentina, but none from Colombia. We described 26 individuals with NS from a single large referral center in the South West of Colombia using an established database in the genetics department and hospital records search using ICD-10 codes. All patients included in this study were evaluated by a medical geneticist and have molecular confirmation of NS diagnosis. The median age at referral was 3.5 years (range, 0-39), and at molecular diagnosis was 5 years (range, 0-40). Patients mostly originated from the southwest region of Colombia (19/26, 73%). Pathogenic variants in PTPN11 are the most common cause of NS in Colombian individuals followed by SHOC2 and SOS1 variants. The prevalence of cardiomyopathy was low in this population compared to other populations. Further research is needed with a larger sample size and including different regions of Colombia to correlate our findings. This study provides new information about time to diagnosis of NS in Colombia, genotypes, and provides important information to help develop guidelines for diagnosis and management of this disease in the region.
Collapse
Affiliation(s)
- Juliana Lores
- Department of Genetics, Fundación Valle del Lili, Cali, Colombia.,Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| | - Carlos E Prada
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Fundación Cardiovascular de Colombia, Bucaramanga, Colombia
| | - Diana Ramírez-Montaño
- Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| | | | - Harry Pachajoa
- Department of Genetics, Fundación Valle del Lili, Cali, Colombia.,Centro de Investigaciones en Anomalías Congénitas y Enfermedades Raras (CIACER), Universidad Icesi, Cali, Colombia
| |
Collapse
|
38
|
Hanses U, Kleinsorge M, Roos L, Yigit G, Li Y, Barbarics B, El-Battrawy I, Lan H, Tiburcy M, Hindmarsh R, Lenz C, Salinas G, Diecke S, Müller C, Adham I, Altmüller J, Nürnberg P, Paul T, Zimmermann WH, Hasenfuss G, Wollnik B, Cyganek L. Intronic CRISPR Repair in a Preclinical Model of Noonan Syndrome-Associated Cardiomyopathy. Circulation 2020; 142:1059-1076. [PMID: 32623905 DOI: 10.1161/circulationaha.119.044794] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Noonan syndrome (NS) is a multisystemic developmental disorder characterized by common, clinically variable symptoms, such as typical facial dysmorphisms, short stature, developmental delay, intellectual disability as well as cardiac hypertrophy. The underlying mechanism is a gain-of-function of the RAS-mitogen-activated protein kinase signaling pathway. However, our understanding of the pathophysiological alterations and mechanisms, especially of the associated cardiomyopathy, remains limited and effective therapeutic options are lacking. METHODS Here, we present a family with two siblings displaying an autosomal recessive form of NS with massive hypertrophic cardiomyopathy as clinically the most prevalent symptom caused by biallelic mutations within the leucine zipper-like transcription regulator 1 (LZTR1). We generated induced pluripotent stem cell-derived cardiomyocytes of the affected siblings and investigated the patient-specific cardiomyocytes on the molecular and functional level. RESULTS Patients' induced pluripotent stem cell-derived cardiomyocytes recapitulated the hypertrophic phenotype and uncovered a so-far-not-described causal link between LZTR1 dysfunction, RAS-mitogen-activated protein kinase signaling hyperactivity, hypertrophic gene response and cellular hypertrophy. Calcium channel blockade and MEK inhibition could prevent some of the disease characteristics, providing a molecular underpinning for the clinical use of these drugs in patients with NS, but might not be a sustainable therapeutic option. In a proof-of-concept approach, we explored a clinically translatable intronic CRISPR (clustered regularly interspaced short palindromic repeats) repair and demonstrated a rescue of the hypertrophic phenotype. CONCLUSIONS Our study revealed the human cardiac pathogenesis in patient-specific induced pluripotent stem cell-derived cardiomyocytes from NS patients carrying biallelic variants in LZTR1 and identified a unique disease-specific proteome signature. In addition, we identified the intronic CRISPR repair as a personalized and in our view clinically translatable therapeutic strategy to treat NS-associated hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Ulrich Hanses
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Mandy Kleinsorge
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Lennart Roos
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Gökhan Yigit
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Yun Li
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Boris Barbarics
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Ibrahim El-Battrawy
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Huan Lan
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany (I.E-B., H.L.)
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Robin Hindmarsh
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Christof Lenz
- Institute for Clinical Chemistry (C.L.), University Medical Center Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany (C.L.)
| | - Gabriela Salinas
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Sebastian Diecke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Stem Cell Core Facility, Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.D.).,Berlin Institute of Health, Germany (S.D.)
| | - Christian Müller
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Ibrahim Adham
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.)
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Germany (J.A., P.N.)
| | - Thomas Paul
- Clinic for Pediatric Cardiology and Intensive Care Medicine (B.B., T.P.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology (M.T., W-H.Z.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Gerd Hasenfuss
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Bernd Wollnik
- Institute of Human Genetics (G.Y., Y.L., G.S., C.M., I.A., B.W.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.).,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W-H.Z., G.H., B.W.)
| | - Lukas Cyganek
- Clinic for Cardiology and Pneumology (U.H., M.K., L.R., R.H., G.H., L.C.).,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Mannheim and Berlin, Germany (U.H., M.K., L.R., G.Y., B.B., I.E-B., M.T., R.H., S.D., T.P., W.-H.Z., G.H., B.W., L.C.)
| |
Collapse
|
39
|
Lasho T, Patnaik MM. Juvenile myelomonocytic leukemia – A bona fide RASopathy syndrome. Best Pract Res Clin Haematol 2020; 33:101171. [DOI: 10.1016/j.beha.2020.101171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022]
|
40
|
Marcu A, Colita A, Radu LE, Jercan CG, Bica AM, Asan M, Coriu D, Tanase AD, Diaconu CC, Mambet C, Botezatu A, Pasca S, Teodorescu P, Anton G, Gurban P, Colita A. Single-Center Experience With Epigenetic Treatment for Juvenile Myelomonocytic Leukemia. Front Oncol 2020; 10:484. [PMID: 32328464 PMCID: PMC7161089 DOI: 10.3389/fonc.2020.00484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm diagnosed in young children, characterized by somatic or germline mutations that lead to hyperactive RAS signaling. The only curative option is hematopoietic stem cell transplantation (HSCT). Recent data showing that aberrant DNA methylation plays a significant role in pathogenesis and correlates with clinical risk suggest a possible benefit of hypomethylating agents (HMA) in JMML treatment. Aim: The aim is to report the results of HMA-based therapy with 5-azacytidine (AZA) in three JMML patients treated in a single center, non-participating in EWOG-MDS study. Methods: The diagnosis and treatment response were evaluated according to international consensus criteria. AZA 75 mg/m2 intravenous (i.v.) was administered once daily on days 1-7 of each 28-day cycle. All patients were monitored for hematologic response, spleen size, and evolution of extramedullary disease. Targeted next generation sequencing (NGS) were performed after the 3rd AZA cycle and before SCT to evaluate the molecular alterations and genetic response. Results: Three patients diagnosed with JMML were treated with AZA (off-label indication) in Pediatric Department of Fundeni Clinical Institute, Bucharest, Romania between 2017 and 2019. There were two females and one male with median age 11 months, range 2-16 months. The cytogenetic analysis showed normal karyotype in all patients. Molecular analysis confirmed KRAS G13D mutation in two patients and NRAS G12D mutation in one patient. The clinical evaluation showed important splenomegaly and hepatomegaly in all 3 pts. One patient received AZA for early relapse after haploidentical HSCT and the other two patients received upfront AZA, as bridging therapy before HSCT. After HMA therapy, 2/3 patients achieved clinical partial response (cPR), 1/3 had clinical stable disease (cSD) and all had genetic stable disease (gSD) after 3 cycles and were able to receive the planned HSTC. One patient achieved clinical and genetic complete response before HSCT. During 22 cycles of AZA there were only four adverse events but only one determined dose reduction and treatment delay. Conclusion: Our data show that AZA monotherapy is safe and effective in controlling disease both in upfront and relapsed patients in order to proceed to HSCT.
Collapse
Affiliation(s)
- Andra Marcu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Stem Cell Transplantation, Coltea Hospital, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Letitia Elena Radu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Cristina Georgiana Jercan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana Maria Bica
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Minodora Asan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Daniel Coriu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Alina Daniela Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Titu Maiorescu University of Medicine, Bucharest, Romania
| | - Carmen C. Diaconu
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Cristina Mambet
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Gabriela Anton
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Petruta Gurban
- Personal Genetics-Medical Genetics Center, Bucharest, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
41
|
Marcu A, Colita A, Radu LE, Jercan CG, Bica AM, Asan M, Coriu D, Tanase AD, Diaconu CC, Mambet C, Botezatu A, Pasca S, Teodorescu P, Anton G, Gurban P, Colita A. Single-Center Experience With Epigenetic Treatment for Juvenile Myelomonocytic Leukemia. Front Oncol 2020; 10:484. [PMID: 32328464 PMCID: PMC7161089 DOI: 10.3389/fonc.2020.00484,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/17/2020] [Indexed: 01/27/2025] Open
Abstract
Background: Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm diagnosed in young children, characterized by somatic or germline mutations that lead to hyperactive RAS signaling. The only curative option is hematopoietic stem cell transplantation (HSCT). Recent data showing that aberrant DNA methylation plays a significant role in pathogenesis and correlates with clinical risk suggest a possible benefit of hypomethylating agents (HMA) in JMML treatment. Aim: The aim is to report the results of HMA-based therapy with 5-azacytidine (AZA) in three JMML patients treated in a single center, non-participating in EWOG-MDS study. Methods: The diagnosis and treatment response were evaluated according to international consensus criteria. AZA 75 mg/m2 intravenous (i.v.) was administered once daily on days 1-7 of each 28-day cycle. All patients were monitored for hematologic response, spleen size, and evolution of extramedullary disease. Targeted next generation sequencing (NGS) were performed after the 3rd AZA cycle and before SCT to evaluate the molecular alterations and genetic response. Results: Three patients diagnosed with JMML were treated with AZA (off-label indication) in Pediatric Department of Fundeni Clinical Institute, Bucharest, Romania between 2017 and 2019. There were two females and one male with median age 11 months, range 2-16 months. The cytogenetic analysis showed normal karyotype in all patients. Molecular analysis confirmed KRAS G13D mutation in two patients and NRAS G12D mutation in one patient. The clinical evaluation showed important splenomegaly and hepatomegaly in all 3 pts. One patient received AZA for early relapse after haploidentical HSCT and the other two patients received upfront AZA, as bridging therapy before HSCT. After HMA therapy, 2/3 patients achieved clinical partial response (cPR), 1/3 had clinical stable disease (cSD) and all had genetic stable disease (gSD) after 3 cycles and were able to receive the planned HSTC. One patient achieved clinical and genetic complete response before HSCT. During 22 cycles of AZA there were only four adverse events but only one determined dose reduction and treatment delay. Conclusion: Our data show that AZA monotherapy is safe and effective in controlling disease both in upfront and relapsed patients in order to proceed to HSCT.
Collapse
Affiliation(s)
- Andra Marcu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Stem Cell Transplantation, Coltea Hospital, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Letitia Elena Radu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Cristina Georgiana Jercan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana Maria Bica
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Minodora Asan
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Daniel Coriu
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Alina Daniela Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Hematology, Titu Maiorescu University of Medicine, Bucharest, Romania
| | - Carmen C. Diaconu
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Cristina Mambet
- Cellular and Molecular Pathology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Sergiu Pasca
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Gabriela Anton
- Molecular Virology Department, Stefan S Nicolau Institute of Virology, Bucharest, Romania
| | - Petruta Gurban
- Personal Genetics-Medical Genetics Center, Bucharest, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
42
|
Malaquias AC, Noronha RM, Souza TTO, Homma TK, Funari MFA, Yamamoto GL, Silva FV, Moraes MB, Honjo RS, Kim CA, Nesi-França S, Carvalho JAR, Quedas EPS, Bertola DR, Jorge AAL. Impact of Growth Hormone Therapy on Adult Height in Patients with PTPN11 Mutations Related to Noonan Syndrome. Horm Res Paediatr 2020; 91:252-261. [PMID: 31132774 DOI: 10.1159/000500264] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 04/10/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The aim of this study was to evaluate the response to recombinant human growth hormone (rhGH) treatment in patients with Noonan syndrome (NS). MATERIALS AND METHODS Forty-two patients (35 PTPN11+) were treated with rhGH, and 17 were followed-up until adult height. The outcomes were changes in growth velocity (GV) and height standard deviation scores (SDS) for normal (height-CDC SDS) and Noonan standards (height-NS SDS). RESULTS The pretreatment chronological age was 10.3 ± 3.5 years. Height-CDC SDS and height-NS SDS were -3.1 ± 0.7 and -0.5 ± 0.6, respectively. PTPN11+ patients had a better growth response than PTPN11- patients. GV SDS increased from -1.2 ± 1.8 to 3.1 ± 2.8 after the first year of therapy in PTPN11+ patients, and from -1.9 ± 2.6 to -0.1 ± 2.6 in PTPN11- patients. The gain in height-CDC SDS during the first year was higher in PTPN11+ than PTPN11- (0.6 ± 0.4 vs. 0.1 ± 0.2, p = 0.008). Similarly, the gain was observed in height-NS SDS (0.6 ± 0.3 vs. 0.2 ± 0.2, respectively, p < 0.001). Among the patients that reached adult height (n = 17), AH-CDC SDS and AH-NS SDS were -2.1 ± 0.7 and 0.7 ± 0.8, respectively. The total increase in height SDS was 1.3 ± 0.7 and 1.5 ± 0.6 for normal and NS standards, respectively. CONCLUSIONS This study supports the advantage of rhGH therapy on adult height in PTPN11+ patients. In comparison, PTPN11- patients showed a poor response to rhGH. However, this PTPN11- group was small, preventing an adequate comparison among different genotypes and no guarantee of response to therapy in genes besides PTPN11.
Collapse
Affiliation(s)
- Alexsandra C Malaquias
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil, .,Departamento de Pediatria, Faculdade de Ciencias Medicas da Santa Casa de Sao Paulo, Sao Paulo, Brazil,
| | - Renata M Noronha
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Departamento de Pediatria, Faculdade de Ciencias Medicas da Santa Casa de Sao Paulo, Sao Paulo, Brazil
| | - Thaiana T O Souza
- Departamento de Pediatria, Faculdade de Ciencias Medicas da Santa Casa de Sao Paulo, Sao Paulo, Brazil
| | - Thais K Homma
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Laboratorio de Hormonios e Genetica Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Hospital das Clinicas, FMUSP, Sao Paulo, Brazil
| | - Mariana F A Funari
- Laboratorio de Hormonios e Genetica Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Hospital das Clinicas, FMUSP, Sao Paulo, Brazil
| | | | - Fernanda Viana Silva
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | | | - Rachel S Honjo
- Unidade de Genetica, Instituto da Crianca, FMUSP, Sao Paulo, Brazil
| | - Chong A Kim
- Unidade de Genetica, Instituto da Crianca, FMUSP, Sao Paulo, Brazil
| | - Suzana Nesi-França
- Unidade de Endocrinologia Pediatrica, Departamento de Pediatria, Universidade Federal do Parana, Curitiba, Brazil
| | - Julienne A R Carvalho
- Unidade de Endocrinologia Pediatrica, Departamento de Pediatria, Universidade Federal do Parana, Curitiba, Brazil
| | - Elisangela P S Quedas
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Debora R Bertola
- Unidade de Genetica, Instituto da Crianca, FMUSP, Sao Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia-Genetica, LIM/25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Laboratorio de Hormonios e Genetica Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Hospital das Clinicas, FMUSP, Sao Paulo, Brazil
| |
Collapse
|
43
|
After 95 years, it's time to eRASe JMML. Blood Rev 2020; 43:100652. [PMID: 31980238 DOI: 10.1016/j.blre.2020.100652] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/07/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Juvenile myelomonocytic leukaemia (JMML) is a rare clonal disorder of early childhood. Constitutive activation of the RAS pathway is the initial event in JMML. Around 90% of patients diagnosed with JMML carry a mutation in the PTPN11, NRAS, KRAS, NF1 or CBL genes. It has been demonstrated that after this first genetic event, an additional somatic mutation or epigenetic modification is involved in disease progression. The available genetic and clinical data have enabled researchers to establish relationships between JMML and several clinical conditions, including Noonan syndrome, Ras-associated lymphoproliferative disease, and Moyamoya disease. Despite scientific progress and the development of more effective treatments, JMML is still a deadly disease: the 5-year survival rate is ~50%. Here, we report on recent research having led to a better understanding of the genetic and molecular mechanisms involved in JMML.
Collapse
|
44
|
Gross AM, Frone M, Gripp KW, Gelb BD, Schoyer L, Schill L, Stronach B, Biesecker LG, Esposito D, Hernandez ER, Legius E, Loh ML, Martin S, Morrison DK, Rauen KA, Wolters PL, Zand D, McCormick F, Savage SA, Stewart DR, Widemann BC, Yohe ME. Advancing RAS/RASopathy therapies: An NCI-sponsored intramural and extramural collaboration for the study of RASopathies. Am J Med Genet A 2020; 182:866-876. [PMID: 31913576 DOI: 10.1002/ajmg.a.61485] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/22/2019] [Indexed: 12/18/2022]
Abstract
RASopathies caused by germline pathogenic variants in genes that encode RAS pathway proteins. These disorders include neurofibromatosis type 1 (NF1), Noonan syndrome (NS), cardiofaciocutaneous syndrome (CFC), and Costello syndrome (CS), and others. RASopathies are characterized by heterogenous manifestations, including congenital heart disease, failure to thrive, and increased risk of cancers. Previous work led by the NCI Pediatric Oncology Branch has altered the natural course of one of the key manifestations of the RASopathy NF1. Through the conduct of a longitudinal cohort study and early phase clinical trials, the MEK inhibitor selumetinib was identified as the first active therapy for the NF1-related peripheral nerve sheath tumors called plexiform neurofibromas (PNs). As a result, selumetinib was granted breakthrough therapy designation by the FDA for the treatment of PN. Other RASopathy manifestations may also benefit from RAS targeted therapies. The overall goal of Advancing RAS/RASopathy Therapies (ART), a new NCI initiative, is to develop effective therapies and prevention strategies for the clinical manifestations of the non-NF1 RASopathies and for tumors characterized by somatic RAS mutations. This report reflects discussions from a February 2019 initiation meeting for this project, which had broad international collaboration from basic and clinical researchers and patient advocates.
Collapse
Affiliation(s)
- Andrea M Gross
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Megan Frone
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Karen W Gripp
- Department of Genetics, Division of Pediatrics, Al duPont Hospital for Children, Wilmington, Delaware
| | - Bruce D Gelb
- Department of Pediatrics, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Leslie G Biesecker
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, Maryland
| | - Dominic Esposito
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Edjay Ralph Hernandez
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Eric Legius
- Laboratory for Neurofibromatosis Research, Department of Human Genetics, KU Leuven University Hospital, Leuven, Belgium
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Staci Martin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Deborah K Morrison
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, California
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Dina Zand
- Center for Drug Evaluation and Research, Food and Drug Administration, Rockville, Maryland
| | - Frank McCormick
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marielle E Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
45
|
Castellanos E, Rosas I, Negro A, Gel B, Alibés A, Baena N, Pineda M, Pi G, Pintos G, Salvador H, Lázaro C, Blanco I, Vilageliu L, Brems H, Grinberg D, Legius E, Serra E. Mutational spectrum by phenotype: panel‐based NGS testing of patients with clinical suspicion of RASopathy and children with multiple café‐au‐lait macules. Clin Genet 2019; 97:264-275. [DOI: 10.1111/cge.13649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Elisabeth Castellanos
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Inma Rosas
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Alex Negro
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Bernat Gel
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Andreu Alibés
- Cancer Genetics and Epigenetics GroupProgram of Predictive and Personalized Medicine of Cancer (PMPPC), Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
| | - Neus Baena
- Genetics Laboratory of the UDIAT‐CDParc Tauli Health Corporation Barcelona Spain
| | - Mercè Pineda
- Neuropaediatrics UnitHospital Sant Joan de Déu Barcelona Spain
| | - Graciela Pi
- Neuropaediatrics UnitLa Ribera Hospital Valencia Spain
| | - Guillem Pintos
- Department of PediatricsGermans Trias i Pujol University Hospital and Research Institute (IGTP), Universitat de Barcelona Barcelona Spain
| | - Hector Salvador
- Paediatrics Oncology UnitHospital Sant Joan de Déu Barcelona Spain
| | - Conxi Lázaro
- Hereditary Cancer ProgramCatalan Institute of Oncology (ICO‐IDIBELL‐ONCOBELL), L'Hospitalet de Llobregat Barcelona Spain
- Centro de Investigación Biomédica en RED (CIBERONC)Instituto de Salud Carlos III Madrid Spain
| | - Ignacio Blanco
- Clinical Genetics and Genetic Counseling ProgramGermans Trias i Pujol Hospital Barcelona Spain
| | - Lluïsa Vilageliu
- Department of Genetics, Microbiology and Statistics, Facultat de BiologiaUniversitat de Barcelona (UB), IBUB, IRSJD, CIBERER Barcelona Spain
| | - Hilde Brems
- Laboratory for Neurofibromatosis Research, Department of Human GeneticsKU Leuven University Hospital Leuven Belgium
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Facultat de BiologiaUniversitat de Barcelona (UB), IBUB, IRSJD, CIBERER Barcelona Spain
| | - Eric Legius
- Laboratory for Neurofibromatosis Research, Department of Human GeneticsKU Leuven University Hospital Leuven Belgium
| | - Eduard Serra
- Hereditary Cancer Group, Program of Predictive and Personalized Medicine of Cancer (PMPPC)Germans Trias & Pujol Research Institute (IGTP) Barcelona Spain
- Centro de Investigación Biomédica en RED (CIBERONC)Instituto de Salud Carlos III Madrid Spain
| |
Collapse
|
46
|
Shallis RM, Zeidan AM. Myelodysplastic/myeloproliferative neoplasm, unclassifiable (MDS/MPN-U): More than just a "catch-all" term? Best Pract Res Clin Haematol 2019; 33:101132. [PMID: 32460977 DOI: 10.1016/j.beha.2019.101132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
The clinicopathology of MDS and MPN are not mutually exclusive and for this reason the category of myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) exists. Several sub-entities have been included under the MDS/MPN umbrella, including MDS/MPN-unclassifiable (MDS/MPN-U) for those cases whose morphologic and clinical phenotype do not meet criteria to be classified as any other MDS/MPN sub-entity. Though potentially regarded as a wastebasket diagnosis, since its integration into myeloid disease classification, MDS/MPN-U has been refined with increasing understanding of the mutational and genomic events that drive particular clinicopathologic phenotypes, even within MDS/MPN-U. The prototypical example is the identification of SF3B1 mutations and its durable association with MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T), an entity previously buried within, but now a separate category outside of MDS/MPN-U. Continued and enhanced study of those entities under MDS/MPN-U, a perhaps provisional category itself, is likely to progressively identify commonality between many "unclassifiables" to establish a new classifiable diagnosis.
Collapse
Affiliation(s)
- Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA; Yale Cancer Center, New Haven, USA.
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA; Yale Cancer Center, New Haven, USA
| |
Collapse
|
47
|
A Statistical Test for Differential Network Analysis Based on Inference of Gaussian Graphical Model. Sci Rep 2019; 9:10863. [PMID: 31350445 PMCID: PMC6659630 DOI: 10.1038/s41598-019-47362-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/15/2019] [Indexed: 11/09/2022] Open
Abstract
Differential network analysis investigates how the network of connected genes changes from one condition to another and has become a prevalent tool to provide a deeper and more comprehensive understanding of the molecular etiology of complex diseases. Based on the asymptotically normal estimation of large Gaussian graphical model (GGM) in the high-dimensional setting, we developed a computationally efficient test for differential network analysis through testing the equality of two precision matrices, which summarize the conditional dependence network structures of the genes. Additionally, we applied a multiple testing procedure to infer the differential network structure with false discovery rate (FDR) control. Through extensive simulation studies with different combinations of parameters including sample size, number of vertices, level of heterogeneity and graph structure, we demonstrated that our method performed much better than the current available methods in terms of accuracy and computational time. In real data analysis on lung adenocarcinoma, we revealed a differential network with 3503 nodes and 2550 edges, which consisted of 50 clusters with an FDR threshold at 0.05. Many of the top gene pairs in the differential network have been reported relevant to human cancers. Our method represents a powerful tool of network analysis for high-dimensional biological data.
Collapse
|
48
|
Capri Y, Flex E, Krumbach OH, Carpentieri G, Cecchetti S, Lißewski C, Rezaei Adariani S, Schanze D, Brinkmann J, Piard J, Pantaleoni F, Lepri FR, Goh ESY, Chong K, Stieglitz E, Meyer J, Kuechler A, Bramswig NC, Sacharow S, Strullu M, Vial Y, Vignal C, Kensah G, Cuturilo G, Kazemein Jasemi NS, Dvorsky R, Monaghan KG, Vincent LM, Cavé H, Verloes A, Ahmadian MR, Tartaglia M, Zenker M. Activating Mutations of RRAS2 Are a Rare Cause of Noonan Syndrome. Am J Hum Genet 2019; 104:1223-1232. [PMID: 31130282 DOI: 10.1016/j.ajhg.2019.04.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/18/2019] [Indexed: 01/18/2023] Open
Abstract
Aberrant signaling through pathways controlling cell response to extracellular stimuli constitutes a central theme in disorders affecting development. Signaling through RAS and the MAPK cascade controls a variety of cell decisions in response to cytokines, hormones, and growth factors, and its upregulation causes Noonan syndrome (NS), a developmental disorder whose major features include a distinctive facies, a wide spectrum of cardiac defects, short stature, variable cognitive impairment, and predisposition to malignancies. NS is genetically heterogeneous, and mutations in more than ten genes have been reported to underlie this disorder. Despite the large number of genes implicated, about 10%-20% of affected individuals with a clinical diagnosis of NS do not have mutations in known RASopathy-associated genes, indicating that additional unidentified genes contribute to the disease, when mutated. By using a mixed strategy of functional candidacy and exome sequencing, we identify RRAS2 as a gene implicated in NS in six unrelated subjects/families. We show that the NS-causing RRAS2 variants affect highly conserved residues localized around the nucleotide binding pocket of the GTPase and are predicted to variably affect diverse aspects of RRAS2 biochemical behavior, including nucleotide binding, GTP hydrolysis, and interaction with effectors. Additionally, all pathogenic variants increase activation of the MAPK cascade and variably impact cell morphology and cytoskeletal rearrangement. Finally, we provide a characterization of the clinical phenotype associated with RRAS2 mutations.
Collapse
|
49
|
Qu L, Pan C, He SM, Lang B, Gao GD, Wang XL, Wang Y. The Ras Superfamily of Small GTPases in Non-neoplastic Cerebral Diseases. Front Mol Neurosci 2019; 12:121. [PMID: 31213978 PMCID: PMC6555388 DOI: 10.3389/fnmol.2019.00121] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
The small GTPases from the Ras superfamily play crucial roles in basic cellular processes during practically the entire process of neurodevelopment, including neurogenesis, differentiation, gene expression, membrane and protein traffic, vesicular trafficking, and synaptic plasticity. Small GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Different subfamilies of small GTPases have been linked to a number of non-neoplastic cerebral diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), intellectual disability, epilepsy, drug addiction, Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and a large number of idiopathic cerebral diseases. Here, we attempted to make a clearer illustration of the relationship between Ras superfamily GTPases and non-neoplastic cerebral diseases, as well as their roles in the neural system. In future studies, potential treatments for non-neoplastic cerebral diseases which are based on small GTPase related signaling pathways should be explored further. In this paper, we review all the available literature in support of this possibility.
Collapse
Affiliation(s)
- Liang Qu
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Chao Pan
- Beijing Institute of Biotechnology, Beijing, China
| | - Shi-Ming He
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China.,Department of Neurosurgery, Xi'an International Medical Center, Xi'an, China
| | - Bing Lang
- The School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
50
|
Castel P, Cheng A, Cuevas-Navarro A, Everman DB, Papageorge AG, Simanshu DK, Tankka A, Galeas J, Urisman A, McCormick F. RIT1 oncoproteins escape LZTR1-mediated proteolysis. Science 2019; 363:1226-1230. [PMID: 30872527 PMCID: PMC6986682 DOI: 10.1126/science.aav1444] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 02/17/2019] [Indexed: 12/11/2022]
Abstract
RIT1 oncoproteins have emerged as an etiologic factor in Noonan syndrome and cancer. Despite the resemblance of RIT1 to other members of the Ras small guanosine triphosphatases (GTPases), mutations affecting RIT1 are not found in the classic hotspots but rather in a region near the switch II domain of the protein. We used an isogenic germline knock-in mouse model to study the effects of RIT1 mutation at the organismal level, which resulted in a phenotype resembling Noonan syndrome. By mass spectrometry, we detected a RIT1 interactor, leucine zipper-like transcription regulator 1 (LZTR1), that acts as an adaptor for protein degradation. Pathogenic mutations affecting either RIT1 or LZTR1 resulted in incomplete degradation of RIT1. This led to RIT1 accumulation and dysregulated growth factor signaling responses. Our results highlight a mechanism of pathogenesis that relies on impaired protein degradation of the Ras GTPase RIT1.
Collapse
Affiliation(s)
- Pau Castel
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Alice Cheng
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Antonio Cuevas-Navarro
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Alex G Papageorge
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dhirendra K Simanshu
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Alexandra Tankka
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline Galeas
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Anatoly Urisman
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Frank McCormick
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|