Network


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

Hotspot


Dive into the research topics where Christiane M. Robbins is active.

Publication


Featured researches published by Christiane M. Robbins.


Nature | 2003

Recurrent de novo point mutations in lamin A cause Hutchinson–Gilford progeria syndrome

Maria Eriksson; W Ted Brown; Leslie B. Gordon; Michael W. Glynn; Joel Singer; Laura J. Scott; Michael R. Erdos; Christiane M. Robbins; Tracy Moses; Peter Berglund; Amalia Dutra; Evgenia Pak; Sandra G. Durkin; Antonei B. Csoka; Michael Boehnke; Thomas W. Glover; Francis S. Collins

Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder characterized by features reminiscent of marked premature ageing. Here, we present evidence of mutations in lamin A (LMNA) as the cause of this disorder. The HGPS gene was initially localized to chromosome 1q by observing two cases of uniparental isodisomy of 1q—the inheritance of both copies of this material from one parent—and one case with a 6-megabase paternal interstitial deletion. Sequencing of LMNA, located in this interval and previously implicated in several other heritable disorders, revealed that 18 out of 20 classical cases of HGPS harboured an identical de novo (that is, newly arisen and not inherited) single-base substitution, G608G(GGC > GGT), within exon 11. One additional case was identified with a different substitution within the same codon. Both of these mutations result in activation of a cryptic splice site within exon 11, resulting in production of a protein product that deletes 50 amino acids near the carboxy terminus. Immunofluorescence of HGPS fibroblasts with antibodies directed against lamin A revealed that many cells show visible abnormalities of the nuclear membrane. The discovery of the molecular basis of this disease may shed light on the general phenomenon of human ageing.


Nature Genetics | 2003

High frequency of BRAF mutations in nevi.

Pamela M. Pollock; Ursula Harper; Katherine S. Hansen; Laura M. Yudt; Mitchell S. Stark; Christiane M. Robbins; Tracy Moses; Galen Hostetter; Urs Wagner; John W. Kakareka; Ghadi Salem; Tom Pohida; Peter J. Heenan; Paul H. Duray; Olli Kallioniemi; Nicholas K. Hayward; Jeffrey M. Trent; Paul S. Meltzer

To evaluate the timing of mutations in BRAF (v-raf murine sarcoma viral oncogene homolog B1) during melanocytic neoplasia, we carried out mutation analysis on microdissected melanoma and nevi samples. We observed mutations resulting in the V599E amino-acid substitution in 41 of 60 (68%) melanoma metastases, 4 of 5 (80%) primary melanomas and, unexpectedly, in 63 of 77 (82%) nevi. These data suggest that mutational activation of the RAS/RAF/MAPK pathway in nevi is a critical step in the initiation of melanocytic neoplasia but alone is insufficient for melanoma tumorigenesis.


Nature | 2007

A Transforming Mutation in the Pleckstrin Homology Domain of Akt1 in Cancer.

John D. Carpten; Andrew L. Faber; Candice Horn; Gregory P. Donoho; Stephen L. Briggs; Christiane M. Robbins; Galen Hostetter; Sophie Boguslawski; Tracy Moses; Stephanie Savage; Mark Uhlik; Aimin Lin; Jian Du; Yue-Wei Qian; Douglas J. Zeckner; Greg Tucker-Kellogg; Jeffrey W. Touchman; Ketan Patel; Spyro Mousses; Michael L. Bittner; Richard W. Schevitz; Mei-Huei T. Lai; Kerry Blanchard; James E. Thomas

Although AKT1 (v-akt murine thymoma viral oncogene homologue 1) kinase is a central member of possibly the most frequently activated proliferation and survival pathway in cancer, mutation of AKT1 has not been widely reported. Here we report the identification of a somatic mutation in human breast, colorectal and ovarian cancers that results in a glutamic acid to lysine substitution at amino acid 17 (E17K) in the lipid-binding pocket of AKT1. Lys 17 alters the electrostatic interactions of the pocket and forms new hydrogen bonds with a phosphoinositide ligand. This mutation activates AKT1 by means of pathological localization to the plasma membrane, stimulates downstream signalling, transforms cells and induces leukaemia in mice. This mechanism indicates a direct role of AKT1 in human cancer, and adds to the known genetic alterations that promote oncogenesis through the phosphatidylinositol-3-OH kinase/AKT pathway. Furthermore, the E17K substitution decreases the sensitivity to an allosteric kinase inhibitor, so this mutation may have important clinical utility for AKT drug development.


Nature Genetics | 2002

HRPT2, encoding parafibromin, is mutated in hyperparathyroidism-jaw tumor syndrome.

John D. Carpten; Christiane M. Robbins; Andrea Villablanca; Lars Forsberg; S. Presciuttini; Joan E. Bailey-Wilson; William F. Simonds; Elizabeth M. Gillanders; A.M. Kennedy; Jindong Chen; Sunita K. Agarwal; Raman Sood; Mary Pat Jones; Tracy Moses; Carola J. Haven; David Petillo; P.D. Leotlela; B. Harding; D. Cameron; A.A. Pannett; Anders Höög; H. Heath; L.A. James-Newton; Bruce G. Robinson; R.J. Zarbo; Branca Cavaco; W. Wassif; Nancy D. Perrier; I.B. Rosen; U. Kristoffersson

We report here the identification of a gene associated with the hyperparathyroidism–jaw tumor (HPT–JT) syndrome. A single locus associated with HPT–JT (HRPT2) was previously mapped to chromosomal region 1q25–q32. We refined this region to a critical interval of 12 cM by genotyping in 26 affected kindreds. Using a positional candidate approach, we identified thirteen different heterozygous, germline, inactivating mutations in a single gene in fourteen families with HPT–JT. The proposed role of HRPT2 as a tumor suppressor was supported by mutation screening in 48 parathyroid adenomas with cystic features, which identified three somatic inactivating mutations, all located in exon 1. None of these mutations were detected in normal controls, and all were predicted to cause deficient or impaired protein function. HRPT2 is a ubiquitously expressed, evolutionarily conserved gene encoding a predicted protein of 531 amino acids, for which we propose the name parafibromin. Our findings suggest that HRPT2 is a tumor-suppressor gene, the inactivation of which is directly involved in predisposition to HPT–JT and in development of some sporadic parathyroid tumors.


Nature Genetics | 2002

Germline mutations in the ribonuclease L gene in families showing linkage with HPC1.

John D. Carpten; Nina N. Nupponen; Sarah D. Isaacs; Raman Sood; Christiane M. Robbins; Jun Xu; Mezbah U. Faruque; Tracy Moses; C. Ewing; Elizabeth M. Gillanders; P. Hu; P. Bujnovszky; Izabela Makalowska; Agnes Baffoe-Bonnie; D. Faith; Jennifer A. Smith; Dietrich A. Stephan; Kathy E. Wiley; Michael J. Brownstein; Derek Gildea; B. Kelly; R. Jenkins; Galen Hostetter; M. Matikainen; J. Schleutker; K. Klinger; T. Connors; Yong Bing Xiang; Zhining Wang; A. De Marzo

Although prostate cancer is the most common non-cutaneous malignancy diagnosed in men in the United States, little is known about inherited factors that influence its genetic predisposition. Here we report that germline mutations in the gene encoding 2′-5′-oligoadenylate(2-5A)–dependent RNase L (RNASEL) segregate in prostate cancer families that show linkage to the HPC1 (hereditary prostate cancer 1) region at 1q24–25 (ref. 9). We identified RNASEL by a positional cloning/candidate gene method, and show that a nonsense mutation and a mutation in an initiation codon of RNASEL segregate independently in two HPC1-linked families. Inactive RNASEL alleles are present at a low frequency in the general population. RNASEL regulates cell proliferation and apoptosis through the interferon-regulated 2-5A pathway and has been suggested to be a candidate tumor suppressor gene. We found that microdissected tumors with a germline mutation showed loss of heterozygosity and loss of RNase L protein, and that RNASEL activity was reduced in lymphoblasts from heterozyogous individuals compared with family members who were homozygous with respect to the wildtype allele. Thus, germline mutations in RNASEL may be of diagnostic value, and the 2-5A pathway might provide opportunities for developing therapies for those with prostate cancer.


Nature Genetics | 1999

Determination of ancestral alleles for human single-nucleotide polymorphisms using high-density oligonucleotide arrays.

Joseph G. Hacia; Jian-Bing Fan; Oliver A. Ryder; Li Jin; Keith Edgemon; G. Ghandour; R. A. Mayer; Bryan K. Sun; Linda Hsie; Christiane M. Robbins; Lawrence C. Brody; Dong Wang; Eric S. Lander; Robert J. Lipshutz; Stephen P. A. Fodor; Francis S. Collins

Here we report the application of high-density oligonucleotide array (DNA chip)-based analysis to determine the distant history of single nucleotide polymorphisms (SNPs) in current human populations. We analysed orthologues for 397 human SNP sites (identified in CEPH pedigrees from Amish, Venezuelan and Utah populations) from 23 common chimpanzee, 19 pygmy chimpanzee and 11 gorilla genomic DNA samples. From this data we determined 214 proposed ancestral alleles (the sequence found in the last common ancestor of humans and chimpanzees). In a diverse human population set, we found that SNP alleles with higher frequencies were more likely to be ancestral than less frequently occurring alleles. There were, however, exceptions. We also found three shared human/pygmy chimpanzee polymorphisms, all involving CpG dinucleotides, and two shared human/gorilla polymorphisms, one involving a CpG dinucleotide. We demonstrate that microarray-based assays allow rapid comparative sequence analysis of intra- and interspecies genetic variation.


The New England Journal of Medicine | 2012

Germline Mutations in HOXB13 and Prostate-Cancer Risk

Charles M. Ewing; Anna M. Ray; Ethan M. Lange; Kimberly A. Zuhlke; Christiane M. Robbins; Waibhav Tembe; Kathleen E. Wiley; Sarah D. Isaacs; Dorhyun Johng; Yunfei Wang; Chris Bizon; Guifang Yan; Marta Gielzak; Alan W. Partin; Vijayalakshmi Shanmugam; Tyler Izatt; Shripad Sinari; David Craig; S. Lilly Zheng; Patrick C. Walsh; James E. Montie; Jianfeng Xu; John D. Carpten; William B. Isaacs; Kathleen A. Cooney

BACKGROUND Family history is a significant risk factor for prostate cancer, although the molecular basis for this association is poorly understood. Linkage studies have implicated chromosome 17q21-22 as a possible location of a prostate-cancer susceptibility gene. METHODS We screened more than 200 genes in the 17q21-22 region by sequencing germline DNA from 94 unrelated patients with prostate cancer from families selected for linkage to the candidate region. We tested family members, additional case subjects, and control subjects to characterize the frequency of the identified mutations. RESULTS Probands from four families were discovered to have a rare but recurrent mutation (G84E) in HOXB13 (rs138213197), a homeobox transcription factor gene that is important in prostate development. All 18 men with prostate cancer and available DNA in these four families carried the mutation. The carrier rate of the G84E mutation was increased by a factor of approximately 20 in 5083 unrelated subjects of European descent who had prostate cancer, with the mutation found in 72 subjects (1.4%), as compared with 1 in 1401 control subjects (0.1%) (P=8.5x10(-7)). The mutation was significantly more common in men with early-onset, familial prostate cancer (3.1%) than in those with late-onset, nonfamilial prostate cancer (0.6%) (P=2.0x10(-6)). CONCLUSIONS The novel HOXB13 G84E variant is associated with a significantly increased risk of hereditary prostate cancer. Although the variant accounts for a small fraction of all prostate cancers, this finding has implications for prostate-cancer risk assessment and may provide new mechanistic insights into this common cancer. (Funded by the National Institutes of Health and others.).


Nature Genetics | 2003

Melanoma mouse model implicates metabotropic glutamate signaling in melanocytic neoplasia.

Pamela M. Pollock; Karine A. Cohen-Solal; Raman Sood; Jin Namkoong; Jeffrey J. Martino; Aruna Koganti; Hua Zhu; Christiane M. Robbins; Izabela Makalowska; Seung Shick Shin; Yarí E. Marín; Kathleen G. Roberts; Laura M. Yudt; Amy Chen; Jun Cheng; Arturo Incao; Heather W. Pinkett; Christopher L. Graham; Karen J. Dunn; Steven M. Crespo-Carbone; Kerine R. Mackason; Kevin Ryan; Daniel Sinsimer; James S. Goydos; Kenneth R. Reuhl; Michael Eckhaus; Paul S. Meltzer; William J. Pavan; Jeffrey M. Trent; Suzie Chen

To gain insight into melanoma pathogenesis, we characterized an insertional mouse mutant, TG3, that is predisposed to develop multiple melanomas. Physical mapping identified multiple tandem insertions of the transgene into intron 3 of Grm1 (encoding metabotropic glutamate receptor 1) with concomitant deletion of 70 kb of intronic sequence. To assess whether this insertional mutagenesis event results in alteration of transcriptional regulation, we analyzed Grm1 and two flanking genes for aberrant expression in melanomas from TG3 mice. We observed aberrant expression of only Grm1. Although we did not detect its expression in normal mouse melanocytes, Grm1 was ectopically expressed in the melanomas from TG3 mice. To confirm the involvement of Grm1 in melanocytic neoplasia, we created an additional transgenic line with Grm1 expression driven by the dopachrome tautomerase promoter. Similar to the original TG3, the Tg(Grm1)EPv line was susceptible to melanoma. In contrast to human melanoma, these transgenic mice had a generalized hyperproliferation of melanocytes with limited transformation to fully malignant metastasis. We detected expression of GRM1 in a number of human melanoma biopsies and cell lines but not in benign nevi and melanocytes. This study provides compelling evidence for the importance of metabotropic glutamate signaling in melanocytic neoplasia.


Nature Genetics | 2004

Nonsense-mediated decay microarray analysis identifies mutations of EPHB2 in human prostate cancer

Pia Huusko; Damaris Ponciano-Jackson; Maija Wolf; Jeff Kiefer; David O. Azorsa; Sukru Tuzmen; Don Weaver; Christiane M. Robbins; Tracy Moses; Minna Allinen; Sampsa Hautaniemi; Yidong Chen; Abdel G. Elkahloun; Mark Basik; G. Steven Bova; Lukas Bubendorf; Alessandro Lugli; Guido Sauter; Johanna Schleutker; Hilmi Ozcelik; Sabine Elowe; Tony Pawson; Jeffrey M. Trent; John D. Carpten; Olli Kallioniemi; Spyro Mousses

The identification of tumor-suppressor genes in solid tumors by classical cancer genetics methods is difficult and slow. We combined nonsense-mediated RNA decay microarrays and array-based comparative genomic hybridization for the genome-wide identification of genes with biallelic inactivation involving nonsense mutations and loss of the wild-type allele. This approach enabled us to identify previously unknown mutations in the receptor tyrosine kinase gene EPHB2. The DU 145 prostate cancer cell line, originating from a brain metastasis, carries a truncating mutation of EPHB2 and a deletion of the remaining allele. Additional frameshift, splice site, missense and nonsense mutations are present in clinical prostate cancer samples. Transfection of DU 145 cells, which lack functional EphB2, with wild-type EPHB2 suppresses clonogenic growth. Taken together with studies indicating that EphB2 may have an essential role in cell migration and maintenance of normal tissue architecture, our findings suggest that mutational inactivation of EPHB2 may be important in the progression and metastasis of prostate cancer.


Genome Research | 2011

Copy number and targeted mutational analysis reveals novel somatic events in metastatic prostate tumors

Christiane M. Robbins; Waibov A. Tembe; Angela Baker; Shripad Sinari; Tracy Moses; Stephen M. Beckstrom-Sternberg; James S. Beckstrom-Sternberg; Michael T. Barrett; James Long; Arul M. Chinnaiyan; James Lowey; Edward Suh; John V. Pearson; David Craig; David B. Agus; Kenneth J. Pienta; John D. Carpten

Advanced prostate cancer can progress to systemic metastatic tumors, which are generally androgen insensitive and ultimately lethal. Here, we report a comprehensive genomic survey for somatic events in systemic metastatic prostate tumors using both high-resolution copy number analysis and targeted mutational survey of 3508 exons from 577 cancer-related genes using next generation sequencing. Focal homozygous deletions were detected at 8p22, 10q23.31, 13q13.1, 13q14.11, and 13q14.12. Key genes mapping within these deleted regions include PTEN, BRCA2, C13ORF15, and SIAH3. Focal high-level amplifications were detected at 5p13.2-p12, 14q21.1, 7q22.1, and Xq12. Key amplified genes mapping within these regions include SKP2, FOXA1, and AR. Furthermore, targeted mutational analysis of normal-tumor pairs has identified somatic mutations in genes known to be associated with prostate cancer including AR and TP53, but has also revealed novel somatic point mutations in genes including MTOR, BRCA2, ARHGEF12, and CHD5. Finally, in one patient where multiple independent metastatic tumors were available, we show common and divergent somatic alterations that occur at both the copy number and point mutation level, supporting a model for a common clonal progenitor with metastatic tumor-specific divergence. Our study represents a deep genomic analysis of advanced metastatic prostate tumors and has revealed candidate somatic alterations, possibly contributing to lethal prostate cancer.

Collaboration


Dive into the Christiane M. Robbins's collaboration.

Top Co-Authors

Avatar

John D. Carpten

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Jeffrey M. Trent

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Raman Sood

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Izabela Makalowska

Adam Mickiewicz University in Poznań

View shared research outputs
Top Co-Authors

Avatar

Tracy Moses

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joan E. Bailey-Wilson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dietrich A. Stephan

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William B. Isaacs

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge