Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bruce D. Gelb is active.

Publication


Featured researches published by Bruce D. Gelb.


Nature Genetics | 2001

Mutations in PTPN11 , encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome

Marco Tartaglia; Ernest L. Mehler; Rosalie Goldberg; Giuseppe Zampino; Han G. Brunner; Hannie Kremer; Ineke van der Burgt; Andrew H. Crosby; Andra Ion; Steve Jeffery; Kamini Kalidas; Michael A. Patton; Raju Kucherlapati; Bruce D. Gelb

Noonan syndrome (MIM 163950) is an autosomal dominant disorder characterized by dysmorphic facial features, proportionate short stature and heart disease (most commonly pulmonic stenosis and hypertrophic cardiomyopathy). Webbed neck, chest deformity, cryptorchidism, mental retardation and bleeding diatheses also are frequently associated with this disease. This syndrome is relatively common, with an estimated incidence of 1 in 1,000–2,500 live births. It has been mapped to a 5-cM region (N-SH2) on chromosome 12q24.1, and genetic heterogeneity has also been documented. Here we show that missense mutations in PTPN11 (MIM 176876)—a gene encoding the nonreceptor protein tyrosine phosphatase SHP-2, which contains two Src homology 2 (SH2) domains—cause Noonan syndrome and account for more than 50% of the cases that we examined. All PTPN11 missense mutations cluster in interacting portions of the amino N-SH2 domain and the phosphotyrosine phosphatase domains, which are involved in switching the protein between its inactive and active conformations. An energetics-based structural analysis of two N-SH2 mutants indicates that in these mutants there may be a significant shift of the equilibrium favoring the active conformation. This implies that they are gain-of-function changes and that the pathogenesis of Noonan syndrome arises from excessive SHP-2 activity.


Science | 1996

Pycnodysostosis, a lysosomal disease caused by cathepsin K deficiency

Bruce D. Gelb; Guo-Ping Shi; Harold A. Chapman; Robert J. Desnick

Pycnodysostosis, an autosomal recessive osteochondrodysplasia characterized by osteosclerosis and short stature, maps to chromosome 1q21. Cathepsin K, a cysteine protease gene that is highly expressed in osteoclasts, localized to the pycnodysostosis region. Nonsense, missense, and stop codon mutations in the gene encoding cathepsin K were identified in patients. Transient expression of complementary DNA containing the stop codon mutation resulted in messenger RNA but no immunologically detectable protein. Thus, pycnodysostosis results from gene defects in a lysosomal protease with highest expression in osteoclasts. These findings suggest that cathepsin K is a major protease in bone resorption, providing a possible rationale for the treatment of disorders such as osteoporosis and certain forms of arthritis.


Nature Genetics | 2003

Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia

Marco Tartaglia; Charlotte M. Niemeyer; Alessandra Fragale; Xiaoling Song; Jochen Buechner; Andreas Jung; Karel Hählen; Henrik Hasle; Jonathan D. Licht; Bruce D. Gelb

We report here that individuals with Noonan syndrome and juvenile myelomonocytic leukemia (JMML) have germline mutations in PTPN11 and that somatic mutations in PTPN11 account for 34% of non-syndromic JMML. Furthermore, we found mutations in PTPN11 in a small percentage of individuals with myelodysplastic syndrome (MDS) and de novo acute myeloid leukemia (AML). Functional analyses documented that the two most common mutations in PTPN11 associated with JMML caused a gain of function.


American Journal of Human Genetics | 2002

PTPN11 mutations in Noonan syndrome: molecular spectrum, genotype-phenotype correlation, and phenotypic heterogeneity

Marco Tartaglia; Kamini Kalidas; Adam Shaw; Xiaoling Song; Dan L. Musat; Ineke van der Burgt; Han G. Brunner; Débora Romeo Bertola; Andrew H. Crosby; Andra Ion; Raju Kucherlapati; Steve Jeffery; Michael A. Patton; Bruce D. Gelb

Noonan syndrome (NS) is a developmental disorder characterized by facial dysmorphia, short stature, cardiac defects, and skeletal malformations. We recently demonstrated that mutations in PTPN11, the gene encoding the non-receptor-type protein tyrosine phosphatase SHP-2 (src homology region 2-domain phosphatase-2), cause NS, accounting for approximately 50% of cases of this genetically heterogeneous disorder in a small cohort. All mutations were missense changes and clustered at the interacting portions of the amino-terminal src-homology 2 (N-SH2) and protein tyrosine phosphatase (PTP) domains. A gain of function was postulated as a mechanism for the disease. Here, we report the spectrum and distribution of PTPN11 mutations in a large, well-characterized cohort with NS. Mutations were found in 54 of 119 (45%) unrelated individuals with sporadic or familial NS. There was a significantly higher prevalence of mutations among familial cases than among sporadic ones. All defects were missense, and several were recurrent. The vast majority of mutations altered amino acid residues located in or around the interacting surfaces of the N-SH2 and PTP domains, but defects also affected residues in the C-SH2 domain, as well as in the peptide linking the N-SH2 and C-SH2 domains. Genotype-phenotype analysis revealed that pulmonic stenosis was more prevalent among the group of subjects with NS who had PTPN11 mutations than it was in the group without them (70.6% vs. 46.2%; P<.01), whereas hypertrophic cardiomyopathy was less prevalent among those with PTPN11 mutations (5.9% vs. 26.2%; P<.005). The prevalence of other congenital heart malformations, short stature, pectus deformity, cryptorchidism, and developmental delay did not differ between the two groups. A PTPN11 mutation was identified in a family inheriting Noonan-like/multiple giant-cell lesion syndrome, extending the phenotypic range of disease associated with this gene.


Circulation | 2007

Genetic Basis for Congenital Heart Defects: Current Knowledge A Scientific Statement From the American Heart Association Congenital Cardiac Defects Committee, Council on Cardiovascular Disease in the Young: Endorsed by the American Academy of Pediatrics

Mary Ella Pierpont; Craig T. Basson; D. Woodrow Benson; Bruce D. Gelb; Therese M. Giglia; Elizabeth Goldmuntz; Glenn McGee; Craig Sable; Deepak Srivastava; Catherine L. Webb

The intent of this review is to provide the clinician with a summary of what is currently known about the contribution of genetics to the origin of congenital heart disease. Techniques are discussed to evaluate children with heart disease for genetic alterations. Many of these techniques are now available on a clinical basis. Information on the genetic and clinical evaluation of children with cardiac disease is presented, and several tables have been constructed to aid the clinician in the assessment of children with different types of heart disease. Genetic algorithms for cardiac defects have been constructed and are available in an appendix. It is anticipated that this summary will update a wide range of medical personnel, including pediatric cardiologists and pediatricians, adult cardiologists, internists, obstetricians, nurses, and thoracic surgeons, about the genetic aspects of congenital heart disease and will encourage an interdisciplinary approach to the child and adult with congenital heart disease.


Nature | 2010

Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome

Xonia Carvajal-Vergara; Ana Sevilla; Sunita L. D'Souza; Yen Sin Ang; Christoph Schaniel; Dung Fang Lee; Lei Yang; Aaron D. Kaplan; Eric D. Adler; Roye Rozov; Yongchao Ge; Ninette Cohen; Lisa Edelmann; Betty Y. Chang; Avinash Waghray; Jie Su; Sherly Pardo; Klaske D. Lichtenbelt; Marco Tartaglia; Bruce D. Gelb; Ihor R. Lemischka

The generation of reprogrammed induced pluripotent stem cells (iPSCs) from patients with defined genetic disorders holds the promise of increased understanding of the aetiologies of complex diseases and may also facilitate the development of novel therapeutic interventions. We have generated iPSCs from patients with LEOPARD syndrome (an acronym formed from its main features; that is, lentigines, electrocardiographic abnormalities, ocular hypertelorism, pulmonary valve stenosis, abnormal genitalia, retardation of growth and deafness), an autosomal-dominant developmental disorder belonging to a relatively prevalent class of inherited RAS–mitogen-activated protein kinase signalling diseases, which also includes Noonan syndrome, with pleomorphic effects on several tissues and organ systems. The patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase. The iPSCs have been extensively characterized and produce multiple differentiated cell lineages. A major disease phenotype in patients with LEOPARD syndrome is hypertrophic cardiomyopathy. We show that in vitro-derived cardiomyocytes from LEOPARD syndrome iPSCs are larger, have a higher degree of sarcomeric organization and preferential localization of NFATC4 in the nucleus when compared with cardiomyocytes derived from human embryonic stem cells or wild-type iPSCs derived from a healthy brother of one of the LEOPARD syndrome patients. These features correlate with a potential hypertrophic state. We also provide molecular insights into signalling pathways that may promote the disease phenotype.


Nature Genetics | 2007

Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy

Bhaswati Pandit; Anna Sarkozy; Len A. Pennacchio; Claudio Carta; Kimihiko Oishi; Simone Martinelli; Edgar A. Pogna; Wendy Schackwitz; Anna Ustaszewska; Andrew P. Landstrom; J. Martijn Bos; Steve R. Ommen; Giorgia Esposito; Francesca Lepri; Christian Faul; Peter Mundel; Juan Pedro López Siguero; Romano Tenconi; Angelo Selicorni; Cesare Rossi; Laura Mazzanti; Isabella Torrente; Bruno Marino; Maria Cristina Digilio; Giuseppe Zampino; Michael J. Ackerman; Bruno Dallapiccola; Marco Tartaglia; Bruce D. Gelb

Noonan and LEOPARD syndromes are developmental disorders with overlapping features, including cardiac abnormalities, short stature and facial dysmorphia. Increased RAS signaling owing to PTPN11, SOS1 and KRAS mutations causes ∼60% of Noonan syndrome cases, and PTPN11 mutations cause 90% of LEOPARD syndrome cases. Here, we report that 18 of 231 individuals with Noonan syndrome without known mutations (corresponding to 3% of all affected individuals) and two of six individuals with LEOPARD syndrome without PTPN11 mutations have missense mutations in RAF1, which encodes a serine-threonine kinase that activates MEK1 and MEK2. Most mutations altered a motif flanking Ser259, a residue critical for autoinhibition of RAF1 through 14-3-3 binding. Of 19 subjects with a RAF1 mutation in two hotspots, 18 (or 95%) showed hypertrophic cardiomyopathy (HCM), compared with the 18% prevalence of HCM among individuals with Noonan syndrome in general. Ectopically expressed RAF1 mutants from the two HCM hotspots had increased kinase activity and enhanced ERK activation, whereas non–HCM-associated mutants were kinase impaired. Our findings further implicate increased RAS signaling in pathological cardiomyocyte hypertrophy.


Nature Genetics | 2007

Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome

Marco Tartaglia; Len A. Pennacchio; Chen Zhao; Kamlesh K. Yadav; Valentina Fodale; Anna Sarkozy; Bhaswati Pandit; Kimihiko Oishi; Simone Martinelli; Wendy Schackwitz; Anna Ustaszewska; Joel Martin; James Bristow; Claudio Carta; Francesca Lepri; Cinzia Neri; Isabella Vasta; Kate Gibson; Cynthia J. Curry; Juan Pedro López Siguero; Maria Cristina Digilio; Giuseppe Zampino; Bruno Dallapiccola; Dafna Bar-Sagi; Bruce D. Gelb

Noonan syndrome is a developmental disorder characterized by short stature, facial dysmorphia, congenital heart defects and skeletal anomalies. Increased RAS-mitogen-activated protein kinase (MAPK) signaling due to PTPN11 and KRAS mutations causes 50% of cases of Noonan syndrome. Here, we report that 22 of 129 individuals with Noonan syndrome without PTPN11 or KRAS mutation have missense mutations in SOS1, which encodes a RAS-specific guanine nucleotide exchange factor. SOS1 mutations cluster at codons encoding residues implicated in the maintenance of SOS1 in its autoinhibited form. In addition, ectopic expression of two Noonan syndrome–associated mutants induces enhanced RAS and ERK activation. The phenotype associated with SOS1 defects lies within the Noonan syndrome spectrum but is distinctive, with a high prevalence of ectodermal abnormalities but generally normal development and linear growth. Our findings implicate gain-of-function mutations in a RAS guanine nucleotide exchange factor in disease for the first time and define a new mechanism by which upregulation of the RAS pathway can profoundly change human development.


Nature | 2013

De novo mutations in histone-modifying genes in congenital heart disease.

Samir Zaidi; Murim Choi; Hiroko Wakimoto; Lijiang Ma; Jianming Jiang; John D. Overton; Angela Romano-Adesman; Robert D. Bjornson; Roger E. Breitbart; Kerry K. Brown; Nicholas Carriero; Yee Him Cheung; John Deanfield; Steve Depalma; Khalid A. Fakhro; Joseph T. Glessner; Hakon Hakonarson; Jonathan R. Kaltman; Juan P. Kaski; Richard Kim; Jennie Kline; Teresa Lee; Jeremy Leipzig; Alexander E. Lopez; Shrikant Mane; Laura E. Mitchell; Jane W. Newburger; Michael Parfenov; Itsik Pe'er; George A. Porter

Congenital heart disease (CHD) is the most frequent birth defect, affecting 0.8% of live births. Many cases occur sporadically and impair reproductive fitness, suggesting a role for de novo mutations. Here we compare the incidence of de novo mutations in 362 severe CHD cases and 264 controls by analysing exome sequencing of parent–offspring trios. CHD cases show a significant excess of protein-altering de novo mutations in genes expressed in the developing heart, with an odds ratio of 7.5 for damaging (premature termination, frameshift, splice site) mutations. Similar odds ratios are seen across the main classes of severe CHD. We find a marked excess of de novo mutations in genes involved in the production, removal or reading of histone 3 lysine 4 (H3K4) methylation, or ubiquitination of H2BK120, which is required for H3K4 methylation. There are also two de novo mutations in SMAD2, which regulates H3K27 methylation in the embryonic left–right organizer. The combination of both activating (H3K4 methylation) and inactivating (H3K27 methylation) chromatin marks characterizes ‘poised’ promoters and enhancers, which regulate expression of key developmental genes. These findings implicate de novo point mutations in several hundreds of genes that collectively contribute to approximately 10% of severe CHD.


Journal of Experimental Medicine | 2008

Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia

Elisabetta Flex; Valentina Petrangeli; Lorenzo Stella; Sabina Chiaretti; Tekla Hornakova; Laurent Knoops; Cristina Ariola; Valentina Fodale; Emmanuelle Clappier; Francesca Paoloni; Simone Martinelli; Alessandra Fragale; Massimo Sanchez; Simona Tavolaro; Monica Messina; Giovanni Cazzaniga; Andrea Camera; Giovanni Pizzolo; Assunta Tornesello; Marco Vignetti; Angela Battistini; Hélène Cavé; Bruce D. Gelb; Jean-Christophe Renauld; Andrea Biondi; Stefan N. Constantinescu; Robin Foà; Marco Tartaglia

Aberrant signal transduction contributes substantially to leukemogenesis. The Janus kinase 1 (JAK1) gene encodes a cytoplasmic tyrosine kinase that noncovalently associates with a variety of cytokine receptors and plays a nonredundant role in lymphoid cell precursor proliferation, survival, and differentiation. We report that somatic mutations in JAK1 occur in individuals with acute lymphoblastic leukemia (ALL). JAK1 mutations were more prevalent among adult subjects with the T cell precursor ALL, where they accounted for 18% of cases, and were associated with advanced age at diagnosis, poor response to therapy, and overall prognosis. All mutations were missense, and some were predicted to destabilize interdomain interactions controlling the activity of the kinase. Three mutations that were studied promoted JAK1 gain of function and conferred interleukin (IL)-3–independent growth in Ba/F3 cells and/or IL-9–independent resistance to dexamethasone-induced apoptosis in T cell lymphoma BW5147 cells. Such effects were associated with variably enhanced activation of multiple downstream signaling pathways. Leukemic cells with mutated JAK1 alleles shared a gene expression signature characterized by transcriptional up-regulation of genes positively controlled by JAK signaling. Our findings implicate dysregulated JAK1 function in ALL, particularly of T cell origin, and point to this kinase as a target for the development of novel antileukemic drugs.

Collaboration


Dive into the Bruce D. Gelb's collaboration.

Top Co-Authors

Avatar

Marco Tartaglia

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Robert J. Desnick

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Zampino

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Simone Martinelli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Francesca Lepri

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

George A. Diaz

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Goldmuntz

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy E. Roberts

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kimihiko Oishi

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge