Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
2.
J Biol Chem ; 295(29): 10023-10031, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32499374

ABSTRACT

Homeostatic scaling of the synapse, such as synaptic down-scaling, has been proposed to offset deleterious effects induced by sustained synaptic strength enhancement. Proper function and subcellular distribution of Src homology 2 domain-containing nonreceptor protein tyrosine phosphatase (SHP2) are required for synaptic plasticity. However, the role of SHP2 in synaptic down-scaling remains largely unknown. Here, using biochemical assays and cell-imaging techniques, we found that synaptic SHP2 levels are temporally regulated during synaptic down-scaling in cultured hippocampal neurons. Furthermore, we observed that a Noonan syndrome-associated mutation of SHP2, resulting in a D61G substitution, prevents synaptic down-scaling. We further show that this effect is due to an inability of the SHP2-D61G variant to properly disassociate from postsynaptic density protein 95, leading to impaired SHP2 dispersion from synaptic sites after synaptic down-scaling. Our findings reveal a molecular mechanism of the Noonan syndrome-associated genetic variant SHP2-D61G that contributes to deficient synaptic down-scaling.


Subject(s)
Mutation, Missense , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Synapses/metabolism , Amino Acid Substitution , Animals , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , Mice , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Synapses/genetics , Synapses/pathology
3.
Blood ; 134(25): 2304-2317, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31562133

ABSTRACT

Src homology 2 domain-containing phosphatase 2 (SHP2), encoded by the PTPN11 gene, is a ubiquitous protein tyrosine phosphatase that is a critical regulator of signal transduction. Germ line mutations in the PTPN11 gene responsible for catalytic gain or loss of function of SHP2 cause 2 disorders with multiple organ defects: Noonan syndrome (NS) and NS with multiple lentigines (NSML), respectively. Bleeding anomalies have been frequently reported in NS, but causes remain unclear. This study investigates platelet activation in patients with NS and NSML and in 2 mouse models carrying PTPN11 mutations responsible for these 2 syndromes. Platelets from NS mice and patients displayed a significant reduction in aggregation induced by low concentrations of GPVI and CLEC-2 agonists and a decrease in thrombus growth on a collagen surface under arterial shear stress. This was associated with deficiencies in GPVI and αIIbß3 integrin signaling, platelet secretion, and thromboxane A2 generation. Similarly, arterial thrombus formation was significantly reduced in response to a local carotid injury in NS mice, associated with a significant increase in tail bleeding time. In contrast, NSML mouse platelets exhibited increased platelet activation after GPVI and CLEC-2 stimulation and enhanced platelet thrombotic phenotype on collagen matrix under shear stress. Blood samples from NSML patients also showed a shear stress-dependent elevation of platelet responses on collagen matrix. This study brings new insights into the understanding of SHP2 function in platelets, points to new thrombopathies linked to platelet signaling defects, and provides important information for the medical care of patients with NS in situations involving risk of bleeding.


Subject(s)
Blood Platelets/enzymology , Germ-Line Mutation , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Animals , Blood Platelets/pathology , Humans , Mice , Mice, Mutant Strains , Noonan Syndrome/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
4.
Mol Biol (Mosk) ; 50(1): 27-33, 2016.
Article in Russian | MEDLINE | ID: mdl-27028808

ABSTRACT

Tyrosyl phosphorylation participates in various pathological and physiological processes, which are regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). The Src homology-2 domain containing phosphatase SHP2 (encoded by PTPN11) is an important phosphatase, which was found to be implicated in the regulation of genetic disease, development, metabolic, neurological, muscle, skeletal disease and cancer. Germline mutations in PTPN11 cause the Noonan Syndrome, LEOPARD syndrome and metachondromatosis. Somatic PTPN11 mutations occur in hematologic malignancies and in solid tumors. SHP2 is also an important component in oncogenic signaling pathways. It may play different roles in different stages and positions of human cancers. Whether SHP2 is an oncogene or cancer suppressor gene remains to be elucidated. Elucidation of the regulatory mechanisms of SHP2 in human disease will provide new insights into disease and new targets for therapy. Here, we summarized the structural basis and recent research progression on SHP2 in various human disease, including genetic and cancer diseases.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Humans , LEOPARD Syndrome/enzymology , Mutation , Neoplasms/enzymology , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction
6.
PLoS One ; 9(9): e106682, 2014.
Article in English | MEDLINE | ID: mdl-25184253

ABSTRACT

Noonan syndrome (NS) and LEOPARD syndrome (LS) cause congenital afflictions such as short stature, hypertelorism and heart defects. More than 50% of NS and almost all of LS cases are caused by activating and inactivating mutations of the phosphatase Shp2, respectively. How these biochemically opposing mutations lead to similar clinical outcomes is not clear. Using zebrafish models of NS and LS and mass spectrometry-based phosphotyrosine proteomics, we identified a down-regulated peptide of Fer kinase in both NS and LS. Further investigation showed a role for Fer during development, where morpholino-based knockdown caused craniofacial defects, heart edema and short stature. During gastrulation, loss of Fer caused convergence and extension defects without affecting cell fate. Moreover, Fer knockdown cooperated with NS and LS, but not wild type Shp2 to induce developmental defects, suggesting a role for Fer in the pathogenesis of both NS and LS.


Subject(s)
LEOPARD Syndrome/enzymology , Mutation , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein-Tyrosine Kinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Gene Knockdown Techniques , LEOPARD Syndrome/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein-Tyrosine Kinases/genetics , Proteomics , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Curr Cancer Drug Targets ; 14(6): 567-88, 2014.
Article in English | MEDLINE | ID: mdl-25039348

ABSTRACT

Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), encoded by the human PTPN11 gene, is a ubiquitously expressed protein tyrosine phosphatase (PTP) that consists of two tandem Src homology (SH2) domains (N-SH2 and C-SH2), a PTP catalytic domain, and a C-terminal tail with tyrosyl phosphorylation sites. It plays critical roles in numerous cellular processes through the regulation of various signaling pathways in PTP catalytic activity-dependent and -independent manners. Dysfunction of SHP2 resulting from pathogenic mutations and aberrant expression leads to the dysregulation of multiple signaling pathways, thus contributing to different human disorders. Germline and somatic mutations in PTPN11 are involved in Noonan syndrome (NS), LEOPARD syndrome (LS), and hematological malignancies, as well as several solid tumors. In this report, we provide an overview of the current knowledge of the structure and function of SHP2, and further discuss the molecular and pathogenic mechanism of SHP2 in human diseases, with a special focus on tumorigenesis. Furthermore, we summarize that SHP2 might itself represent a potential drug target for cancer prevention and treatment. Ongoing research and development of SHP2-specific inhibitors would enhance this potential.


Subject(s)
Cell Transformation, Neoplastic/metabolism , LEOPARD Syndrome/enzymology , Neoplasms/enzymology , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Drug Design , Enzyme Inhibitors/therapeutic use , Genetic Predisposition to Disease , Humans , LEOPARD Syndrome/genetics , Models, Molecular , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Noonan Syndrome/genetics , Phenotype , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction , Structure-Activity Relationship
8.
Am J Med Genet A ; 164A(9): 2351-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24891296

ABSTRACT

The RASopathies are a relatively common group of phenotypically similar and genetically related autosomal dominant genetic syndromes caused by missense mutations affecting genes participating in the RAS/mitogen-activated protein kinase (MAPK) pathway that include Noonan syndrome (NS) and Noonan syndrome with multiple lentigines (NSML, formerly LEOPARD syndrome). NS and NSML can be difficult to differentiate during infancy, but the presence of multiple lentigines, café au lait spots, and specific cardiac defects facilitate the diagnosis. Furthermore, individual PTPN11 missense mutations are highly specific to each syndrome and engender opposite biochemical alterations on the function of SHP-2, the protein product of that gene. Here, we report on a 5-year-old male with two de novo PTPN11 mutations in cis, c.1471C>T (p.Pro491Ser), and c.1492C>T (p.Arg498Trp), which are associated with NS and NSML, respectively. This boy's phenotype is intermediate between NS and NSML with facial dysmorphism, short stature, mild global developmental delay, pulmonic stenosis, and deafness but absence of café au lait spots or lentigines. The double-mutant SHP-2 was found to be catalytically impaired. This raises the question of whether clinical differences between NS and NSML can be ascribed solely to the relative SHP-2 catalytic activity.


Subject(s)
Alleles , Biocatalysis , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Child, Preschool , Facies , Humans , Infant , Infant, Newborn , Male , Phenotype
9.
Development ; 141(9): 1961-70, 2014 May.
Article in English | MEDLINE | ID: mdl-24718990

ABSTRACT

Germline mutations in PTPN11, encoding Shp2, cause Noonan syndrome (NS) and LEOPARD syndrome (LS), two developmental disorders that are characterized by multiple overlapping symptoms. Interestingly, Shp2 catalytic activity is enhanced by NS mutations and reduced by LS mutations. Defective cardiac development is a prominent symptom of both NS and LS, but how the Shp2 variants affect cardiac development is unclear. Here, we have expressed the most common NS and LS Shp2-variants in zebrafish embryos to investigate their role in cardiac development in vivo. Heart function was impaired in embryos expressing NS and LS variants of Shp2. The cardiac anomalies first occurred during elongation of the heart tube and consisted of reduced cardiomyocyte migration, coupled with impaired leftward heart displacement. Expression of specific laterality markers was randomized in embryos expressing NS and LS variants of Shp2. Ciliogenesis and cilia function in Kupffer's vesicle was impaired, likely accounting for the left/right asymmetry defects. Mitogen-activated protein kinase (MAPK) signaling was activated to a similar extent in embryos expressing NS and LS Shp2 variants. Interestingly, inhibition of MAPK signaling prior to gastrulation rescued cilia length and heart laterality defects. These results suggest that NS and LS Shp2 variant-mediated hyperactivation of MAPK signaling leads to impaired cilia function in Kupffer's vesicle, causing left-right asymmetry defects and defective early cardiac development.


Subject(s)
Heart Defects, Congenital/genetics , LEOPARD Syndrome/genetics , Mutation/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Benzamides/pharmacology , Body Patterning/drug effects , Cell Movement/drug effects , Cilia/drug effects , Cilia/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/pathology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/physiopathology , Heart Function Tests , Humans , LEOPARD Syndrome/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
10.
Am J Med Genet A ; 164A(4): 934-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24458522

ABSTRACT

Here, we describe neurobehavioral features in patients with RASopathies (i.e., Noonan syndrome, LEOPARD syndrome, Costello syndrome, and cardiofaciocutaneous syndrome), developmental disorders caused by mutations in genes coding transducers participating in the RAS-MAPK signaling cascade. Parents of 70 individuals with a RASopathy were asked to fill out the following questionnaires: Child Behavior Checklist (CBCL), Social Communication Questionnaire version lifetime (SCQ-L), and Modified Checklist for Autism in toddlers (M-CHAT). Data analysis indicated high rates of internalizing (37%) and externalizing problems (31%) on CBCL. Scores over the cut-off were documented in 64% of patients with cardiofaciocutaneous syndrome, 44% with Costello syndrome, and 12% with Noonan syndrome on SCQ-L/M-CHAT. Our findings indicate that mutations promoting dysregulation of the RAS-MAPK cascade mark an increased psychopathological risk and highlight that autistic-like behavior could be underdiagnosed in patients with RASopathies.


Subject(s)
MAP Kinase Signaling System/genetics , Mental Disorders/enzymology , Mental Disorders/genetics , ras Proteins/genetics , Adolescent , Adult , Autistic Disorder/enzymology , Autistic Disorder/genetics , Child , Child, Preschool , Costello Syndrome/enzymology , Costello Syndrome/genetics , Developmental Disabilities/enzymology , Developmental Disabilities/genetics , Ectodermal Dysplasia/enzymology , Ectodermal Dysplasia/genetics , Facies , Failure to Thrive/enzymology , Failure to Thrive/genetics , Female , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Humans , LEOPARD Syndrome/enzymology , LEOPARD Syndrome/genetics , Male , Mutation/genetics , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Young Adult
11.
BMC Ophthalmol ; 13: 70, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24219368

ABSTRACT

BACKGROUND: Noonan syndrome is an autosomal, dominantly inherited disease; it is physically characterized by short stature, short neck, webbed neck, abnormal auricles, high arched palate, and cardiovascular malformation. Its pathological condition is thought to be due to a gain-of-function mutation in the Ras-mitogen-activated protein kinase (MAPK) signal transduction pathway. Eyelid abnormalities such as ocular hypertelorism and blepharoptosis are the most commonly observed eye complications. CASE PRESENTATION: We report a case of Noonan syndrome associated with mature cataract that required operation. A 42-year-old man was diagnosed with Noonan syndrome at the age of 1 year. He underwent an eye examination after complaining of decreased visual acuity in the right eye and was diagnosed with mature cataract, which was treated by cataract surgery. There were no intraoperative complications, and the postoperative course was uneventful. Protein analysis of lens capsule and epithelium at capsulorhexis showed MAPK cascade proteins such as ERK and p38MAPK were upregulated. An abnormality in the PTPN11 gene was also observed; a potential mechanism of cataract onset may be that opacity of the lens rapidly progressed due to abnormal activation of the Ras-MAPK signal transduction pathway. CONCLUSION: This case highlights the possible association of cataract formation with MAPK cascade protein upregulation in Noonan syndrome.


Subject(s)
Cataract/enzymology , Lens, Crystalline/enzymology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Noonan Syndrome/enzymology , Adult , Humans , Male
12.
J Proteomics ; 84: 132-47, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23584145

ABSTRACT

SHP2/PTPN11 is a key regulator of cytokine, growth factor and integrin signaling. SHP2 influences cell survival, proliferation and differentiation by regulating major signaling pathways. Mutations in PTPN11 cause severe diseases like Noonan, LEOPARD syndrome or leukemia. Whereas several of these mutations result in altered enzymatic activity due to impaired auto-inhibition, not all disease patterns can be explained by this mechanism. In this study we analyzed altered binding properties of disease-related SHP2-mutants bearing point mutations within the SH2-domain (T42A, E139D, and R138Q). Mutants were chosen according to SPR assays, which revealed different binding properties of mutated SH2 towards phosphorylated receptor peptides. To analyze global changes in mutant binding properties we applied quantitative mass spectrometry (SILAC). Using an in vitro approach we identified overall more than 1000 protein candidates, which specifically bind to the SH2-domain of SHP2. We discovered that mutations in the SH2-domain selectively affected protein enrichment by altering the binding capacity of the SH2-domain. Mutation-dependent, enhanced or reduced exposure of SHP2 to its binding partners could have an impact on the dynamics of signaling networks. Thus, disease-associated mutants of SHP2 should not only be discussed in the context of deregulated auto-inhibition but also with respect to deregulated protein targeting of the SHP2 mutants. BIOLOGICAL SIGNIFICANCE: Using quantitative mass spectrometry based proteomics we provided evidence that disease related mutations in SHP2 domains of SHP2 are able to influence SHP2 recruitment to its targets in mutation dependent manner. We discovered that mutations in the SH2-domain selectively affected protein enrichment ratios suggesting altered binding properties of the SH2-domain. We demonstrated that mutations within SHP2, which had been attributed to affect the enzymatic activity (i.e. affect the open/close status of SHP2), also differ in respect to binding properties. Our study indicates that SHP2 mutations need to be discussed not only in terms of deregulated auto-inhibition but also with respect to deregulated protein targeting properties of the SHP2 mutants. Discovery of the new binding partners for disease-related SHP2 mutants might provide a fruitful foundation for developing strategies targeting Noonan-associated leukemia.


Subject(s)
Leukemia/enzymology , Mutation, Missense , Neoplasm Proteins/metabolism , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Amino Acid Substitution , HeLa Cells , Humans , Leukemia/genetics , Leukemia/pathology , Neoplasm Proteins/genetics , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , src Homology Domains
13.
J Biol Chem ; 287(32): 27066-77, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22711529

ABSTRACT

Activating mutations in PTPN11 cause Noonan syndrome, the most common nonchromosomal disorder affecting development and growth. PTPN11 encodes SHP2, an Src homology 2 (SH2) domain-containing protein-tyrosine phosphatase that positively modulates RAS function. Here, we characterized functionally all possible amino acid substitutions arising from single-base changes affecting codons 62 and 63 to explore the molecular mechanisms lying behind the largely invariant occurrence of the Y62D and Y63C substitutions recurring in Noonan syndrome. We provide structural and biochemical data indicating that the autoinhibitory interaction between the N-SH2 and protein-tyrosine phosphatase (PTP) domains is perturbed in both mutants as a result of an extensive structural rearrangement of the N-SH2 domain. Most mutations affecting Tyr(63) exerted an unpredicted disrupting effect on the structure of the N-SH2 phosphopeptide-binding cleft mediating the interaction of SHP2 with signaling partners. Among all the amino acid changes affecting that codon, the disease-causing mutation was the only substitution that perturbed the stability of the inactive conformation of SHP2 without severely impairing proper phosphopeptide binding of N-SH2. On the other hand, the disruptive effect of the Y62D change on the autoinhibited conformation of the protein was balanced, in part, by less efficient binding properties of the mutant. Overall, our data demonstrate that the selection-by-function mechanism acting as driving force for PTPN11 mutations affecting codons 62 and 63 implies balancing of counteracting effects operating on the allosteric control of the function of SHP2.


Subject(s)
Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , src Homology Domains , Amino Acid Sequence , Humans , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
14.
Proc Natl Acad Sci U S A ; 109(11): 4257-62, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22371576

ABSTRACT

Noonan syndrome (NS), a genetic disease caused in half of cases by activating mutations of the tyrosine phosphatase SHP2 (PTPN11), is characterized by congenital cardiopathies, facial dysmorphic features, and short stature. How mutated SHP2 induces growth retardation remains poorly understood. We report here that early postnatal growth delay is associated with low levels of insulin-like growth factor 1 (IGF-1) in a mouse model of NS expressing the D61G mutant of SHP2. Conversely, inhibition of SHP2 expression in growth hormone (GH)-responsive cell lines results in increased IGF-1 release upon GH stimulation. SHP2-deficient cells display decreased ERK1/2 phosphorylation and rat sarcoma (RAS) activation in response to GH, whereas expression of NS-associated SHP2 mutants results in ERK1/2 hyperactivation in vitro and in vivo. RAS/ERK1/2 inhibition in SHP2-deficient cells correlates with impaired dephosphorylation of the adaptor Grb2-associated binder-1 (GAB1) on its RAS GTPase-activating protein (RASGAP) binding sites and is rescued by interfering with RASGAP recruitment or function. We demonstrate that inhibition of ERK1/2 activation results in an increase of IGF-1 levels in vitro and in vivo, which is associated with significant growth improvement in NS mice. In conclusion, NS-causing SHP2 mutants inhibit GH-induced IGF-1 release through RAS/ERK1/2 hyperactivation, a mechanism that could contribute to growth retardation. This finding suggests that, in addition to its previously shown beneficial effect on NS-linked cardiac and craniofacial defects, RAS/ERK1/2 modulation could also alleviate the short stature phenotype in NS caused by PTPN11 mutations.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/metabolism , Mutation/genetics , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Binding Sites , Biometry , Enzyme Activation/drug effects , Insulin-Like Growth Factor I/biosynthesis , Janus Kinase 2/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Noonan Syndrome/blood , Noonan Syndrome/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , STAT5 Transcription Factor/metabolism , ras Proteins/metabolism
15.
Development ; 139(5): 948-57, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22278918

ABSTRACT

Noonan syndrome is one of the most common causes of human congenital heart disease and is frequently associated with missense mutations in the protein phosphatase SHP-2. Interestingly, patients with acute myelogenous leukemia (AML), acute lymphoblastic leukemia (ALL), juvenile myelomonocytic leukemia (JMML) and LEOPARD syndrome frequently carry a second, somatically introduced subset of missense mutations in SHP-2. To determine the cellular and molecular mechanisms by which SHP-2 regulates heart development and, thus, understand how Noonan-associated mutations affect cardiogenesis, we introduced SHP-2 encoding the most prevalent Noonan syndrome and JMML mutations into Xenopus embryos. Resulting embryos show a direct relationship between a Noonan SHP-2 mutation and its ability to cause cardiac defects in Xenopus; embryos expressing Noonan SHP-2 mutations exhibit morphologically abnormal hearts, whereas those expressing an SHP-2 JMML-associated mutation do not. Our studies indicate that the cardiac defects associated with the introduction of the Noonan-associated SHP-2 mutations are coupled with a delay or arrest of the cardiac cell cycle in M-phase and a failure of cardiomyocyte progenitors to incorporate into the developing heart. We show that these defects are a result of an underlying malformation in the formation and polarity of cardiac actin fibers and F-actin deposition. We show that these defects can be rescued in culture and in embryos through the inhibition of the Rho-associated, coiled-coil-containing protein kinase 1 (ROCK), thus demonstrating a direct relationship between SHP-2(N308D) and ROCK activation in the developing heart.


Subject(s)
Actin Cytoskeleton/metabolism , Heart , Myocardium/metabolism , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Xenopus laevis/embryology , rho-Associated Kinases/metabolism , Animals , Enzyme Activation , Heart/anatomy & histology , Heart/embryology , Humans , Mutation, Missense , Myocardium/cytology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Xenopus laevis/anatomy & histology , rho-Associated Kinases/genetics
16.
Am J Med Genet A ; 152A(11): 2850-3, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20979190

ABSTRACT

Noonan syndrome (NS; OMIM 163950) is an autosomal dominant disorder with variable clinical expression and genetic heterogeneity. Clinical manifestations include characteristic facial features, short stature, and cardiac anomalies. Mutations in protein-tyrosine phosphatase, non-receptor-type 11 (PTPN11), encoding SHP-2, account for about half of NS patients. We report on a Moroccan family with two children with NS and apparently unaffected parents. The molecular studies showed the heterozygous mutation c.922A>G of PTPN11 gene in the two affected sibs. Neither the parents, nor the oldest brother carries this mutation in hematologic cells. The mutation was also absent in buccal epithelial cells and fingernails of both parents. We believe this is the first report of germ cell mosaicism in NS and suggest an empirical risk for recurrence of that is less than 1%.


Subject(s)
Mosaicism , Noonan Syndrome/genetics , Parents , Siblings , Adult , Child, Preschool , Female , Haplotypes/genetics , Humans , Infant , Infant, Newborn , Male , Middle Aged , Noonan Syndrome/enzymology , Pedigree , Pregnancy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 27(5): 554-8, 2010 Oct.
Article in Chinese | MEDLINE | ID: mdl-20931536

ABSTRACT

OBJECTIVE: To investigate the mutations in protein tyrosine phosphatase, nonreceptor-type 11 (PTPN11) gene in patients with Noonan syndrome (NS). METHODS: Three sporadic patients with NS were studied. Genomic DNAs were extracted from peripheral blood leukocytes. All 15 coding exons and their flanking intronic boundaries of the PTPN11 gene were amplified by polymerase chain reaction and followed by direct sequencing. DNAs from parents were sequenced in the corresponding region when the mutation was detected in their affected child. The identified mutation was screened in 100 healthy individuals for exclusion of polymorphism by restriction endonuclease digestion of the PCR products. Protein conservation analysis was performed among 10 species using an online ClustalW tool. RESULTS: Direct DNA sequence analysis identified a heterozygous 181G to A change in exon 3 of the PTPN11 gene in one patient, which resulted in the substitution of an aspartic acid residue by an asparagine at codon 61. The mutation was absent in his parents and 100 controls, and is located in a highly conserved amino acid site. No mutation in the coding region of PTPN11 gene was observed in the other two patients. CONCLUSION: The p.D61N mutation was reported previously in Caucasians and is a de-novo mutation in this patient. Our study further confirmed that the p.D61N is a pathogenic mutation for NS and consistent with the clinical diagnosis. Additional genes may be involved in the other two patients with NS, indicating high genetic heterogeneity of this disease.


Subject(s)
Noonan Syndrome/genetics , Point Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Amino Acid Sequence , Base Sequence , Case-Control Studies , Child , Exons , Female , Humans , Male , Molecular Sequence Data , Mutation, Missense , Noonan Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Sequence Alignment , Young Adult
18.
Am J Med Genet A ; 152A(11): 2768-74, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20954246

ABSTRACT

Reports on Noonan syndrome (NS) have documented multiple types of coagulation defects and bleeding diathesis, and a wide range of clinical presentations. Early studies suggested that a large proportion of NS patients have coagulation defects, whereas more recent reports indicate low rates of coagulopathy. The aim of this study was to evaluate phenotypic characteristics, PTPN11 gene mutations, and hematological and coagulation parameters in 30 clinically diagnosed cases of NS. One of the NS patients had a history of easy bruising; however, his hematological and coagulation tests were normal. None of the other patients had clinical coagulation problems. In the NS group, values for platelet count, activity of factors XI, XII, and protein C were significantly lower than the corresponding means for the control group. However, the results of coagulation tests in the NS group were diagnostically inconclusive and only one patient had clinical signs of coagulopathy. Interestingly, two NS patients had low protein C activity. One of these children had an A1517C mutation and transient myelodysplasia. The other patient had a C1528G mutation in exon 13 that has not been reported previously. Neither of these individuals experienced a thrombotic event or any complication during approximately 3 years of follow-up. For all patients clinically diagnosed with NS, a thorough history of coagulation issues should be taken and first-line coagulation testing should be done to evaluate for bleeding diathesis. However, if these assessments reveal nothing abnormal, complications related to coagulation are unlikely and extensive testing is unnecessary.


Subject(s)
Mutation/genetics , Noonan Syndrome/blood , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Adolescent , Blood Coagulation/genetics , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Male , Noonan Syndrome/enzymology
19.
Biochim Biophys Acta ; 1802(2): 275-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19835954

ABSTRACT

Noonan syndrome (NS) is an autosomal dominant disorder, and a main feature is congenital heart malformation. About 50% of cases are caused by gain-of-function mutations in the tyrosine phosphatase SHP2/PTPN11, a downstream regulator of ERK/MAPK. Recently it was reported that SHP2 also localizes to the mitochondrial intercristae/intermembrane space (IMS), but the role of SHP2 in mitochondria is unclear. The mitochondrial oxidative phosphorylation (OxPhos) system provides the vast majority of cellular energy and produces reactive oxygen species (ROS). Changes in ROS may interfere with organ development such as that observed in NS patients. Several phosphorylation sites have been found in OxPhos components including cytochrome c oxidase (CcO) and cytochrome c (Cytc), and we hypothesized that OxPhos complexes may be direct or indirect targets of SHP2. We analyzed mitochondrial function using mouse fibroblasts from wild-types, SHP2 knockdowns, and D61G SHP2 mutants leading to constitutively active SHP2, as found in NS patients. Levels of OxPhos complexes were similar except for CcO and Cytc, which were 37% and 28% reduced in the D61G cells. However, CcO activity was significantly increased, as we also found for two lymphoblast cell lines from NS patients with two independent mutations in PTPN11. D61G cells showed lower mitochondrial membrane potential and 30% lower ATP content compared to controls. ROS were significantly increased; aconitase activity, a marker for ROS-induced damage, was decreased; and catalase activity was increased in D61G cells. We propose that decreased energy levels and/or increased ROS may explain, at least in part, some of the clinical features in NS that overlap with children with mitochondrial disorders.


Subject(s)
Mitochondria/enzymology , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cytochromes c/genetics , Cytochromes c/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Kinetics , Membrane Potentials , Mice , Mice, Knockout , Mice, Mutant Strains , Mitochondria/physiology , Mitochondrial Membranes/physiology , Mutation , Noonan Syndrome/enzymology , Oxidative Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Reactive Oxygen Species/metabolism , Reference Values
20.
Horm Res ; 71 Suppl 2: 64-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19407499

ABSTRACT

Short stature, with a mean final height almost two standard deviations below the normal mean, is a major feature of Noonan syndrome. The biological basis of the growth failure is not yet clear. The recent detection of mutations in the protein tyrosine phosphatase, non-receptor type 11 gene (PTPN11) in half of all individuals with Noonan syndrome has opened up a new perspective from the endocrine point of view, since the tyrosine phosphatase SHP2 encoded by PTPN11 is implicated in the downregulation of growth hormone (GH) receptor signalling. Current data show decreased insulin-like growth factor (IGF)-I and IGF-binding protein 3 (IGFBP-3) levels in those children with Noonan syndrome who carry PTPN11 mutations. GH responsiveness seems to be reduced in the presence of PTPN11 mutations, but, so far, data are too scarce to draw any final conclusions. Children with Noonan or Noonan-related syndromes carrying mutations in components of the Ras-mitogen-activated protein kinase (MAPK) signalling pathway downstream from SHP2 also have short stature, though less frequently in the case of SOS1 mutations. Therefore, apart from the disturbance of GH signalling, there must be other relevant mechanisms that influence longitudinal growth in Noonan syndrome. In a small subgroup of patients with Noonan syndrome and Noonan-related syndromes, tumour risk is increased. This susceptibility is relevant when GH therapy is considered. Progress in the understanding of cell regulation by Ras-MAPK signalling and its interconnection with other pathways will hopefully provide evidence on which therapy might be helpful and which might be nocuous in the care of children with Noonan syndrome.


Subject(s)
Carrier Proteins/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Noonan Syndrome/enzymology , Oncogene Protein p21(ras)/metabolism , Carrier Proteins/genetics , Down-Regulation/genetics , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/therapy , Noonan Syndrome/genetics , Noonan Syndrome/therapy , Oncogene Protein p21(ras)/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL