Network


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

Hotspot


Dive into the research topics where Patricia K. Donahoe is active.

Publication


Featured researches published by Patricia K. Donahoe.


Nature Genetics | 2004

Detection of large-scale variation in the human genome

A. John Iafrate; Lars Feuk; Miguel Rivera; Marc Listewnik; Patricia K. Donahoe; Ying Qi; Stephen W. Scherer; Charles Lee

We identified 255 loci across the human genome that contain genomic imbalances among unrelated individuals. Twenty-four variants are present in > 10% of the individuals that we examined. Half of these regions overlap with genes, and many coincide with segmental duplications or gaps in the human genome assembly. This previously unappreciated heterogeneity may underlie certain human phenotypic variation and susceptibility to disease and argues for a more dynamic human genome structure.


Cell | 1986

Isolation of the bovine and human genes for müllerian inhibiting substance and expression of the human gene in animal cells

Richard L. Cate; Robert J. Mattaliano; Catherine Hession; Richard Tizard; N.M. Farber; A. Cheung; E.G. Ninfa; Alexis Z. Frey; D.J. Gash; E.P. Chow; R.A. Fisher; J.M. Bertonis; G. Torres; Barbara P. Wallner; R.C. Ragin; D.T. MacLaughlin; Patricia K. Donahoe

We have isolated the bovine and human genes for Müllerian inhibiting substance (MIS), a testicular glycoprotein that causes regression of the Müllerian duct during development of the male embryo. The mRNA sequence of bovine MIS, determined from an analysis of cDNA and genomic clones, codes for a protein of 575 amino acids containing a 24 amino acid leader peptide. The human gene has five exons that code for a protein of 560 amino acids. A comparison of the bovine and human MIS proteins reveals a highly conserved C-terminal domain that shows marked homology with human transforming growth factor-beta and the beta chain of porcine inhibin. Animal cells transfected with the human gene secrete biologically active MIS, which causes regression of the rat Müllerian duct in vitro.


Cell | 1996

The Immunophilin FKBP12 Functions as a Common Inhibitor of the TGFβ Family Type I Receptors

Tongwen Wang; Bi-Yu Li; Paul D. Danielson; Paresh C. Shah; Sybil Rockwell; R J Lechleider; Jennifer Martin; Patricia K. Donahoe

The immunophilin FKBP12 is an evolutionarily conserved abundant protein; however, its physiological roles remain poorly defined. Here we report that FKBP12 is a common cytoplasmic interactor of TGF beta family type I receptors. FKBP12 binds to ligand-free TGF beta type I receptor, from which it is released upon a ligand-induced, type II receptor mediated phosphorylation of the type I receptor. Blocking FKBP12/type I receptor interaction with FK506 nonfunctional derivatives enhances the ligand activity, indicating that FKBP12 binding is inhibitory to the signaling pathways of the TGF beta family ligands. Overexpression of a myristylated FKBP12 in Mv1Lu cell specifically inhibits two separate pathways activated by TGF beta, and two point mutations on FKBP12 (G89P, I90K) abolish the inhibitory activity of FKBP12, suggesting that FKBP12 may dock a cytoplasmic protein to the type I receptors to inhibit TGF beta family mediated signaling.


Nature Biotechnology | 2011

Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants.

Dalila Pinto; Katayoon Darvishi; Xinghua Shi; Diana Rajan; Diane Rigler; Tom Fitzgerald; Anath C. Lionel; Bhooma Thiruvahindrapuram; Jeffrey R. MacDonald; Ryan E. Mills; Aparna Prasad; Kristin M Noonan; Susan Gribble; Elena Prigmore; Patricia K. Donahoe; Richard S Smith; Ji Hyeon Park; Nigel P. Carter; Charles Lee; Stephen W. Scherer; Lars Feuk

We have systematically compared copy number variant (CNV) detection on eleven microarrays to evaluate data quality and CNV calling, reproducibility, concordance across array platforms and laboratory sites, breakpoint accuracy and analysis tool variability. Different analytic tools applied to the same raw data typically yield CNV calls with <50% concordance. Moreover, reproducibility in replicate experiments is <70% for most platforms. Nevertheless, these findings should not preclude detection of large CNVs for clinical diagnostic purposes because large CNVs with poor reproducibility are found primarily in complex genomic regions and would typically be removed by standard clinical data curation. The striking differences between CNV calls from different platforms and analytic tools highlight the importance of careful assessment of experimental design in discovery and association studies and of strict data curation and filtering in diagnostics. The CNV resource presented here allows independent data evaluation and provides a means to benchmark new algorithms.


Proceedings of the National Academy of Sciences of the United States of America | 2008

A large-scale analysis of tissue-specific pathology and gene expression of human disease genes and complexes

Kasper Lage; Niclas Tue Hansen; E. Olof Karlberg; Aron Charles Eklund; Francisco S. Roque; Patricia K. Donahoe; Zoltan Szallasi; Thomas Skøt Jensen; Søren Brunak

Heritable diseases are caused by germ-line mutations that, despite tissuewide presence, often lead to tissue-specific pathology. Here, we make a systematic analysis of the link between tissue-specific gene expression and pathological manifestations in many human diseases and cancers. Diseases were systematically mapped to tissues they affect from disease-relevant literature in PubMed to create a disease–tissue covariation matrix of high-confidence associations of >1,000 diseases to 73 tissues. By retrieving >2,000 known disease genes, and generating 1,500 disease-associated protein complexes, we analyzed the differential expression of a gene or complex involved in a particular disease in the tissues affected by the disease, compared with nonaffected tissues. When this analysis is scaled to all diseases in our dataset, there is a significant tendency for disease genes and complexes to be overexpressed in the normal tissues where defects cause pathology. In contrast, cancer genes and complexes were not overexpressed in the tissues from which the tumors emanate. We specifically identified a complex involved in XY sex reversal that is testis-specific and down-regulated in ovaries. We also identified complexes in Parkinson disease, cardiomyopathies, and muscular dystrophy syndromes that are similarly tissue specific. Our method represents a conceptual scaffold for organism-spanning analyses and reveals an extensive list of tissue-specific draft molecular pathways, both known and unexpected, that might be disrupted in disease.


Nature Genetics | 2007

Mutations in LRP2 , which encodes the multiligand receptor megalin, cause Donnai-Barrow and facio-oculo-acoustico-renal syndromes

Sibel Kantarci; Lihadh Al-Gazali; R. Sean Hill; Dian Donnai; Graeme C.M. Black; Eric Bieth; Nicolas Chassaing; Didier Lacombe; Koenraad Devriendt; Ahmad S. Teebi; Maria Loscertales; Caroline D. Robson; Tianming Liu; David T. MacLaughlin; Kristin M Noonan; Meaghan K Russell; Christopher A. Walsh; Patricia K. Donahoe; Barbara R. Pober

Donnai-Barrow syndrome is associated with agenesis of the corpus callosum, congenital diaphragmatic hernia, facial dysmorphology, ocular anomalies, sensorineural hearing loss and developmental delay. By studying multiplex families, we mapped this disorder to chromosome 2q23.3–31.1 and identified LRP2 mutations in six families with Donnai-Barrow syndrome and one family with facio-oculo-acoustico-renal syndrome. LRP2 encodes megalin, a multiligand uptake receptor that regulates levels of diverse circulating compounds. This work implicates a pathway with potential pharmacological therapeutic targets.


The New England Journal of Medicine | 1997

Measurements of serum mullerian inhibiting substance in the evaluation of children with nonpalpable gonads

Mary M. Lee; Patricia K. Donahoe; Bernard L. Silverman; Tomonobu Hasegawa; Yukihiro Hasegawa; Michael L. Gustafson; Yuchiao Chang; David T. MacLaughlin

BACKGROUND Müllerian inhibiting substance, produced constitutively by the prepubertal testes, promotes involution of the müllerian ducts during normal male sexual differentiation. In children with virilization and nonpalpable gonads, only those with testicular tissue should have detectable serum concentrations of müllerian inhibiting substance. METHODS We measured serum mullerian inhibiting substance in 65 children with virilization at birth and nonpalpable gonads (age at diagnosis, 2 days to 11 years) and serum testosterone in 54 of them either after the administration of human chorionic gonadotropin or during the physiologic rise in testosterone that occurs in normal infants. RESULTS The mean (+/-SD) serum mullerian inhibiting substance concentration in the 17 children with no testicular tissue was 0.7+/-0.5 ng per milliliter, as compared with 37.5+/-39.6 ng per milliliter in the 48 children with testes (P<0.001). In the latter group, the mean values in the 14 children with abnormal testes and the 34 with normal testes were 11.5+/-11.8 and 48.2+/-42.1 ng per milliliter, respectively (P< 0.001). The sensitivity and specificity of the serum müllerian inhibiting substance assay for detecting the absence of testicular tissue were 92 percent and 98 percent, respectively, as compared with 69 percent and 83 percent for the measurement of serum testosterone. Furthermore, measurement of serum mullerian inhibiting substance was more sensitive than serum testosterone measurement for the identification of children with abnormal testes (67 percent vs. 25 percent), whereas the specificity of the two tests was similar. CONCLUSIONS Measurements of serum mullerian inhibiting substance can be used to determine testicular status in prepubertal children with nonpalpable gonads, thus differentiating anorchia from undescended testes in boys with bilateral cryptorchidism and serving as a measure of testicular integrity in children with intersexual anomalies.


Journal of Surgical Research | 1977

A graded organ culture assay for the detection of mullerian inhibiting substance

Patricia K. Donahoe; Yasuo Ito; W. Hardy Hendren

Abstract A sensitive, reliable, semiquantitaive organ culture assay has been developed to detect the presence of Mullerian Inhibiting Substance, both in whole testicular fragments, and in cell-free extracts. This in vitro assay allows regression to occur in a manner which mimics the in vivo regression of the male fetal Mullerian duct. Timed exposure studies demonstrate that the Mullerian duct must be exposed to the testicular fragments for at least 12 hr before regression is initiated. Seventy-two hours are required for the morphological changes of regression to be fully expressed. Mullerian Inhibiting Substance is a compound of considerable importance in sexual differentiation. This assay will allow a more systematic approach to the study of this unique fetal regressor.


Human Pathology | 1982

Dysgenesis of testicular and streak gonads in the syndrome of mixed gonadal dysgenesis: Perspective derived from a clinicopathologic analysis of twenty-one cases

Stanley J. Robboy; Theodore Miller; Patricia K. Donahoe; Caren Jahre; William R. Welch; Florence P. Haseltine; Wayne A. Miller; Leonard Atkins; John D. Crawford

The clinical and pathologic aspects of 21 cases of mixed gonadal dysgenesis (MGD) were studied. The gonads in 15 patients consisted of a macroscopic testis and a streak gonad; six patients had variants, including two with bilateral testes and four with bilateral streak gonads or tumors. Functionally, the gonads were incompetent. Testes 1) failed to completely inhibit müllerian development, 2) failed to support full differentiation of mesonephric duct structures, 3) failed to adequately masculinize development of the external genitalia, or 4) often failed to mediate their own descent, resulting in asymmetry of the internal and external genitalia. None of the streak gonads mediated normal female adolescent development or fertility. Microscopic examination revealed that every gonad, regardless of its gross appearance, was morphologically abnormal. Although gonads with seminiferous tubules usually developed to a moderately advanced state, macroscopically resembling testes, the hilar zone remained architecturally disorganized; the cortex invariably lacked more than a rudimentary tunica albuginea or exhibited partial ovarian differentiation, sometimes even with a rare primordial follicle. Over time, the seminiferous tubules atrophied and hyalinized. Gonads that grossly resembled streak gonads were observed microscopically to be composed of a stroma resembling that of normal ovarian cortex. In patients more than several years of age, the entire complement of germ cells in streak gonads disappeared. It is suggested that patients with MGD be raised as females. Early removal of gonads will prevent the development of gonadoblastoma and dysgerminoma. If the uterus is retained and the patient is subsequently given exogenous estrogen, care should be taken to detect early any signs of the development of endometrial carcinoma or its precursor, to which these patients may be prone.


American Journal of Human Genetics | 2005

Congenital Diaphragmatic Hernia and Chromosome 15q26: Determination of a Candidate Region by Use of Fluorescent In Situ Hybridization and Array-Based Comparative Genomic Hybridization

M. Klaassens; M.F. van Dooren; H. J. Eussen; Hannie Douben; A.T. den Dekker; Charles Lee; Patricia K. Donahoe; Robert Jan H. Galjaard; Natascha Goemaere; R.R. de Krijger; Cokkie H. Wouters; J. Wauters; Ben A. Oostra; Dick Tibboel; A. de Klein

Congenital diaphragmatic hernia (CDH) has an incidence of 1 in 3,000 births and a high mortality rate (33%-58%). Multifactorial inheritance, teratogenic agents, and genetic abnormalities have all been suggested as possible etiologic factors. To define candidate regions for CDH, we analyzed cytogenetic data collected on 200 CDH cases, of which 7% and 5% showed numerical and structural abnormalities, respectively. This study focused on the most frequent structural anomaly found: a deletion on chromosome 15q. We analyzed material from three of our patients and from four previously published patients with CDH and a 15q deletion. By using array-based comparative genomic hybridization and fluorescent in situ hybridization to determine the boundaries of the deletions and by including data from two individuals with terminal 15q deletions but without CDH, we were able to exclude a substantial portion of the telomeric region from the genetic etiology of this disorder. Moreover, one patient with CDH harbored a small interstitial deletion. Together, these findings allowed us to define a minimal deletion region of approximately 5 Mb at chromosome 15q26.1-26.2. The region contains four known genes, of which two--NR2F2 and CHD2--are particularly intriguing gene candidates for CDH.

Collaboration


Dive into the Patricia K. Donahoe's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary M. Lee

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

W. Hardy Hendren

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

John M. Hutson

Royal Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge