Network


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

Hotspot


Dive into the research topics where Alan J. Buckler is active.

Publication


Featured researches published by Alan J. Buckler.


Cell | 1993

A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes

Marcy E. MacDonald; Christine Ambrose; Mabel P. Duyao; Richard H. Myers; Carol Lin; Lakshmi Srinidhi; Glenn Barnes; Sherryl A. M. Taylor; Marianne James; Nicolet Groot; Heather MacFarlane; Barbara Jenkins; Mary Anne Anderson; Nancy S. Wexler; James F. Gusella; Gillian P. Bates; Sarah Baxendale; Holger Hummerich; Susan Kirby; Mike North; Sandra Youngman; Richard Mott; Günther Zehetner; Zdenek Sedlacek; Annemarie Poustka; Anna-Maria Frischauf; Hans Lehrach; Alan J. Buckler; Deanna Church; Lynn Doucette-Stamm

The Huntingtons disease (HD) gene has been mapped in 4p16.3 but has eluded identification. We have used haplotype analysis of linkage disequilibrium to spotlight a small segment of 4p16.3 as the likely location of the defect. A new gene, IT15, isolated using cloned trapped exons from the target area contains a polymorphic trinucleotide repeat that is expanded and unstable on HD chromosomes. A (CAG)n repeat longer than the normal range was observed on HD chromosomes from all 75 disease families examined, comprising a variety of ethnic backgrounds and 4p16.3 haplotypes. The (CAG)n repeat appears to be located within the coding sequence of a predicted approximately 348 kd protein that is widely expressed but unrelated to any known gene. Thus, the HD mutation involves an unstable DNA segment, similar to those described in fragile X syndrome, spino-bulbar muscular atrophy, and myotonic dystrophy, acting in the context of a novel 4p16.3 gene to produce a dominant phenotype.


Cell | 1993

A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor

James A. Trofatter; Mia MacCollin; Joni L. Rutter; Jill R. Murrell; Mabel P. Duyao; Dilys M. Parry; Roswell Eldridge; Nikolai Kley; Anil G. Menon; Karen Pulaski; Volker H. Haase; Christine Ambrose; David J. Munroe; Catherine Bove; Jonathan L. Haines; Robert L. Martuza; Marcy E. MacDonald; Bernd R. Seizinger; M. Priscilla Short; Alan J. Buckler; James F. Gusella

Neurofibromatosis 2 (NF2) is a dominantly inherited disorder characterized by the occurrence of bilateral vestibular schwannomas and other central nervous system tumors including multiple meningiomas. Genetic linkage studies and investigations of both sporadic and familial tumors suggest that NF2 is caused by inactivation of a tumor suppressor gene in chromosome 22q12. We have identified a candidate gene for the NF2 tumor suppressor that has suffered nonoverlapping deletions in DNA from two independent NF2 families and alterations in meningiomas from two unrelated NF2 patients. The candidate gene encodes a 587 amino acid protein with striking similarity to several members of a family of proteins proposed to link cytoskeletal components with proteins in the cell membrane. The NF2 gene may therefore constitute a novel class of tumor suppressor gene.


Nature Genetics | 1997

The early-onset torsion dystonia gene (DYT1) encodes an ATP-binding protein

Laurie J. Ozelius; Jeffrey W. Hewett; Curtis E. Page; Susan B. Bressman; Patricia L. Kramer; Christo Shalish; Deborah de Leon; Mitchell F. Brin; Deborah Raymond; David P. Corey; Stanley Fahn; Neil Risch; Alan J. Buckler; James F. Gusella; Xandra O. Breakefield

Early-onset torsion dystonia is a movement disorder, characterized by twisting muscle contractures, that begins in childhood. Symptoms are believed to result from altered neuronal communication in the basal ganglia. This study identifies the DYT1 gene on human chromosome 9q34 as being responsible for this dominant disease. Almost all cases of early-onset dystonia have a unique 3-bp deletion that appears to have arisen independently in different ethnic populations. This deletion results in loss of one of a pair of glutamic-acid residues in a conserved region of a novel ATP-binding protein, termed torsinA. This protein has homologues in nematode, rat, mouse and humans, with some resemblance to the family of heat-shock proteins and Clp proteases.


Nature Genetics | 1997

Holt-oram syndrome is caused by mutations in TBX5, a member of the Brachyury (T) gene family

Quan Yi Li; Ruth Newbury-Ecob; Jonathan Alexander Terrett; David I. Wilson; Andrew R.J. Curtis; Cheong Ho Yi; Tom Gebuhr; Philip J. Bullen; Stephen C. Robson; Tom Strachan; Damien Bonnet; Stanislas Lyonnet; Ian D. Young; J. Alexander Raeburn; Alan J. Buckler; David J. Law; J. David Brook

Holt-Oram syndrome is a developmental disorder affecting the heart and upper limb, the gene for which was mapped to chromosome 12 two years ago. We have now identified a gene for this disorder (HOS1). The gene (TBX5) is a member of the Brachyury (T) family corresponding to the mouse TbxS gene. We have identified six mutations, three in HOS families and three in sporadic HOS cases. Each of the mutations introduces a premature stop codon in the TBXS gene product. Tissue in situ hybridization studies on human embryos from days 26 to 52 of gestation reveal expression of TBXS in heart and limb, consistent with a role in human embryonic development.


Cell | 1994

The diastrophic dysplasia gene encodes a novel sulfate transporter: Positional cloning by fine-structure linkage disequilibrium mapping

Johanna Hästbacka; Albert de la Chapelle; Melanie M. Mahtani; Greg Clines; Mary Pat Reeve-Daly; Mark J. Daly; Bruce A. Hamilton; Kenro Kusumi; Bijal Trivedi; Alix Weaver; Antonio Coloma; Michael Lovett; Alan J. Buckler; Ilkka Kaitila; Eric S. Lander

Diastrophic dysplasia (DTD) is a well-characterized autosomal recessive osteochondrodysplasia with clinical features including dwarfism, spinal deformation, and specific joint abnormalities. The disease occurs in most populations, but is particularly prevalent in Finland owing to an apparent founder effect. DTD maps to distal chromosome 5q and, based on linkage disequilibrium studies in the Finnish population, we had previously predicted that the DTD gene should lie about 64 kb away from the CSF1R locus. Here, we report the positional cloning of the DTD gene by fine-structure linkage disequilibrium mapping. The gene lies in the predicted location, approximately 70 kb proximal to CSF1R, and encodes a novel sulfate transporter. Impaired function of its product is likely to lead to undersulfation of proteoglycans in cartilage matrix and thereby to cause the clinical phenotype of the disease. These results demonstrate the power of linkage disequilibrium mapping in isolated populations for positional cloning.


Cell | 1995

ISOLATION OF A NOVEL GENE UNDERLYING BATTEN-DISEASE, CLN3

Terry J. Lerner; Rose-Mary Boustany; John W. Anderson; Kenneth L. D'Arigo; Karen Schlumpf; Alan J. Buckler; James F. Gusella; Jonathan L. Haines

Batten disease (also known as juvenile neuronal ceroid lipofuscinosis) is a recessively inherited neurodegenerative disorder of childhood characterized by progressive loss of vision, seizures, and psychomotor disturbances. The Batten disease gene, CLN3, maps to chromosome 16p12.1. The so-called 56 chromosome haplotype defined by alleles at the D16S299 and D16S298 loci is shared by 73% of Batten disease chromosomes. Exon amplification of a cosmid containing D16S298 has yielded a candidate gene that is disrupted by a 1 kb genomic deletion in all patients carrying the 56 chromosome. Two separate deletions and a point mutation altering a splice site in three unrelated families have confirmed the candidate as the CLN3 gene. The disease gene encodes a novel 438 amino acid protein of unknown function.


Somatic Cell and Molecular Genetics | 1994

Structure and expression of the Huntington's disease gene: Evidence against simple inactivation due to an expanded CAG repeat

Christine Ambrose; Mabel P. Duyao; Glenn Barnes; Gillian P. Bates; Carol Lin; Jayalakshmi Srinidhi; Sarah Baxendale; Holger Hummerich; Hans Lehrach; Michael R. Altherr; John J. Wasmuth; Alan J. Buckler; Deanna Church; David E. Housman; M. Berks; Gos Micklem; Richard Durbin; Alan Dodge; Andrew P. Read; James F. Gusella; Marcy E. MacDonald

Huntingtons disease, a neurodegenerative disorder characterized by loss of striatal neurons, is caused by an expanded, unstable trinucleotide repeat in a novel 4p16.3 gene. To lay the foundation for exploring the pathogenic mechanism in HD, we have determined the structure of the disease gene and examined its expression. TheHD locus spans 180 kb and consists of 67 exons ranging in size from 48 bp to 341 bp with an average of 138 bp. Scanning of theHD transcript failed to reveal any additional sequence alterations characteristic of HD chromosomes. A codon loss polymorphism in linkage disequilibrium with the disorder revealed that both normal and HD alleles are represented in the mRNA population in HD heterozygotes, indicating that the defect does not eliminate transcription. The gene is ubiquitously expressed as two alternatively polyadenylated forms displaying different relative abundance in various fetal and adult tissues, suggesting the operation of interacting factors in determining specificity of cell loss. TheHD gene was disrupted in a female carrying a balanced translocation with a breakpoint between exons 40 and 41. The absence of any abnormal phenotype in this individual argues against simple inactivation of the gene as the mechanism by which the expanded trinucleotide repeat causes HD. Taken together, these observations suggest that the dominant HD mutation either confers a new property on the mRNA or, more likely, alters an interaction at the protein level.


Breast Cancer Research | 2008

NVP-AUY922: a small molecule HSP90 inhibitor with potent antitumor activity in preclinical breast cancer models

Michael Rugaard Jensen; Joseph Schoepfer; Thomas Radimerski; Andrew Massey; Chantale T. Guy; Josef Brueggen; Cornelia Quadt; Alan J. Buckler; Robert Cozens; Martin J. Drysdale; Carlos Garcia-Echeverria; Patrick Chène

IntroductionHeat shock protein 90 (HSP90) is a key component of a multichaperone complex involved in the post-translational folding of a large number of client proteins, many of which play essential roles in tumorigenesis. HSP90 has emerged in recent years as a promising new target for anticancer therapies.MethodsThe concentrations of the HSP90 inhibitor NVP-AUY922 required to reduce cell numbers by 50% (GI50 values) were established in a panel of breast cancer cell lines and patient-derived human breast tumors. To investigate the properties of the compound in vivo, the pharmacokinetic profile, antitumor effect, and dose regimen were established in a BT-474 breast cancer xenograft model. The effect on HSP90-p23 complexes, client protein degradation, and heat shock response was investigated in cell culture and breast cancer xenografts by immunohistochemistry, Western blot analysis, and immunoprecipitation.ResultsWe show that the novel small molecule HSP90 inhibitor NVP-AUY922 potently inhibits the proliferation of human breast cancer cell lines with GI50 values in the range of 3 to 126 nM. NVP-AUY922 induced proliferative inhibition concurrent with HSP70 upregulation and client protein depletion – hallmarks of HSP90 inhibition. Intravenous acute administration of NVP-AUY922 to athymic mice (30 mg/kg) bearing subcutaneous BT-474 breast tumors resulted in drug levels in excess of 1,000 times the cellular GI50 value for about 2 days. Significant growth inhibition and good tolerability were observed when the compound was administered once per week. Therapeutic effects were concordant with changes in pharmacodynamic markers, including HSP90-p23 dissociation, decreases in ERBB2 and P-AKT, and increased HSP70 protein levels.ConclusionNVP-AUY922 is a potent small molecule HSP90 inhibitor showing significant activity against breast cancer cells in cellular and in vivo settings. On the basis of its mechanism of action, preclinical activity profile, tolerability, and pharmaceutical properties, the compound recently has entered clinical phase I breast cancer trials.


Cancer Discovery | 2012

Rescue Screens with Secreted Proteins Reveal Compensatory Potential of Receptor Tyrosine Kinases in Driving Cancer Growth

Fred Harbinski; Vanessa J. Craig; Sneha Sanghavi; Douglas Jeffery; Lijuan Liu; Kelly Ann Sheppard; Sabrina Wagner; Christelle Stamm; Andreas Buness; Christian Chatenay-Rivauday; Yao Yao; Feng He; Chris Lu; Vito Guagnano; Thomas Metz; Peter Finan; Francesco Hofmann; William R. Sellers; Jeffrey A. Porter; Vic E. Myer; Diana Graus-Porta; Christopher J. Wilson; Alan J. Buckler; Ralph Tiedt

The overall power of kinase inhibitors is substantially overshadowed by the acquisition of drug resistance. To address this issue, we systematically assessed the potential of secreted proteins to induce resistance to kinase inhibitors. To this end, we developed a high-throughput platform for screening a cDNA library encoding 3,432 secreted proteins in cellular assays. Using cancer cells originally dependent on either MET, FGFR2, or FGFR3, we observed a bypass of dependence through ligand-mediated activation of alternative receptor tyrosine kinases (RTK). Our findings indicate a broad and versatile potential for RTKs from the HER and FGFR families as well as MET to compensate for loss of each other. We further provide evidence that combined inhibition of simultaneously active RTKs can lead to an added anticancer effect.


Journal of Proteome Research | 2008

Product ion monitoring assay for prostate-specific antigen in serum using a linear ion-trap.

Vathany Kulasingam; Christopher R. Smith; Ihor Batruch; Alan J. Buckler; Douglas Jeffery; Eleftherios P. Diamandis

While numerous strategies exist for biomarker discovery, the bottleneck to product development and routine use at the clinic is in the verification phase of candidate biomarkers. The aim of this study was to establish a robust and high-throughput product ion monitoring (PIM) assay that is potentially capable of rapidly verifying candidates from discovery phase experiments. Using prostate-specific antigen (PSA), a model biomarker, and a routinely used mass spectrometer for discovery platforms, an ion trap (LTQ, Thermo), the utility of this instrument to perform PIM was explored. The proteotypic doubly charged intact peptide LSEPAELTDAVK ( m/ z 637) fragmenting to m/ z 943 (PAELTDAVK) was monitored. A limit of detection of 10 attomoles with a coefficient of variation (CV) of <20% was obtained for a purified recombinant PSA digest. Immunoextraction of endogenous PSA from serum using a monoclonal antibody on a 96-well microtiter plate, followed by PIM on the LTQ, enabled quantification of PSA down to less than 1 ng/mL with a limit of detection of 0.1 ng/mL and CVs < 20%. Mascot searching and ion ratio confirmation further supported the conclusion that the quantified moiety in serum was the PSA peptide. We conclude that this methodology could be adapted quickly and easily to other candidates, thus providing a much needed technology to bridge the gap between discovery and validation platforms.

Collaboration


Dive into the Alan J. Buckler's collaboration.

Top Co-Authors

Avatar

David E. Housman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge