Network


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

Hotspot


Dive into the research topics where Chris Neff is active.

Publication


Featured researches published by Chris Neff.


American Journal of Human Genetics | 2003

Predisposition locus for major depression at chromosome 12q22-12q23.2

Victor Abkevich; Nicola J. Camp; Charles H. Hensel; Chris Neff; Deanna L. Russell; Dana C. Hughes; Agnes M. Plenk; Michael R. Lowry; R. Lynn Richards; Catherine Carter; Georges C. Frech; Steven Stone; Kerry Rowe; Chi Ai Chau; Kathleen Cortado; Angelene Hunt; Karanina Luce; Gayanne O’Neil; Jeff Poarch; Jennifer Potter; Gregg H. Poulsen; Heidi Saxton; Michelle Bernat-Sestak; Victor Thompson; Alexander Gutin; Mark H. Skolnick; Donna Shattuck; Lisa Cannon-Albright

Major depression disorder is a common psychiatric disease with a major economic impact on society. In many cases, no effective treatment is available. The etiology of major depression is complex, but it is clear that the disease is, to a large extent, determined genetically, especially among individuals with a familial history of major depression, presumably through the involvement of multiple predisposition genes in addition to an environmental component. As a first step toward identification of chromosomal loci contributing to genetic predisposition to major depression, we have conducted a genomewide scan by using 628 microsatellite markers on 1,890 individuals from 110 Utah pedigrees with a strong family history of major depression. We identified significant linkage to major depression in males at marker D12S1300 (multipoint heterogeneity LOD score 4.6; P=.00003 after adjustment for multiple testing). With additional markers, the linkage evidence became highly significant, with the multipoint heterogeneity LOD score at marker D12S1706 increasing to 6.1 (P=.0000007 after adjustment for multiple testing). This study confirms the presence of one or more genes involved in psychiatric diseases on the q arm of chromosome 12 and provides strong evidence for the existence of a sex-specific predisposition gene to major depression at 12q22-q23.2.


American Journal of Human Genetics | 2002

A major predisposition locus for severe obesity, at 4p15-p14.

Steven Stone; Victor Abkevich; Steven C. Hunt; Alexander Gutin; Deanna L. Russell; Chris Neff; Robyn Riley; Georges C. Frech; Charles H. Hensel; Srikanth Jammulapati; Jennifer Potter; David Sexton; Thanh Tran; Drew Gibbs; Diana Iliev; Richard E. Gress; Brian T. Bloomquist; John Amatruda; M.M. Peter Rae; D. Ted Adams; H. Mark Skolnick; Donna Shattuck

Although the predisposition to morbid obesity is heritable, the identities of the disease-causing genes are largely unknown. Therefore, we have conducted a genomewide search with 628 markers, using multigenerational Utah pedigrees to identify genes involved in predisposition to obesity. In the genomewide search, we identified a highly significant linkage to high body-mass index in female patients, at D4S2632, with a multipoint heterogeneity LOD (HLOD) score of 6.1 and a nonparametric linkage (NPL) score of 5.3. To further delineate the linkage, we increased both the marker density around D4S2632 and the size of our pedigree data set. As a result, the linkage evidence increased to a multipoint HLOD score of 9.2 (at D4S3350) and an NPL score of 11.3. Evidence from almost half of the families in this analysis support this linkage, and therefore the gene in this region might account for a significant percentage of the genetic predisposition to severe obesity in females. However, further studies are necessary to clarify the effect that this gene has in males and in the general population.


Clinical Cancer Research | 2016

Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer

Melinda L. Telli; Kirsten Timms; Julia Reid; Bryan T. Hennessy; Gordon B. Mills; Kristin C. Jensen; Zoltan Szallasi; William T. Barry; Nadine Tung; Steven J. Isakoff; Paula D. Ryan; April Greene-Colozzi; Alexander Gutin; Zaina Sangale; Diana Iliev; Chris Neff; Victor Abkevich; Joshua Jones; Jerry S. Lanchbury; Anne-Renee Hartman; Judy Garber; James M. Ford; Daniel P. Silver; Andrea L. Richardson

Purpose: BRCA1/2-mutated and some sporadic triple-negative breast cancers (TNBC) have DNA repair defects and are sensitive to DNA-damaging therapeutics. Recently, three independent DNA-based measures of genomic instability were developed on the basis of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST). Experimental Design: We assessed a combined homologous recombination deficiency (HRD) score, an unweighted sum of LOH, TAI, and LST scores, in three neoadjuvant TNBC trials of platinum-containing therapy. We then tested the association of HR deficiency, defined as HRD score ≥42 or BRCA1/2 mutation, with response to platinum-based therapy. Results: In a trial of neoadjuvant platinum, gemcitabine, and iniparib, HR deficiency predicted residual cancer burden score of 0 or I (RCB 0/I) and pathologic complete response (pCR; OR = 4.96, P = 0.0036; OR = 6.52, P = 0.0058). HR deficiency remained a significant predictor of RCB 0/I when adjusted for clinical variables (OR = 5.86, P = 0.012). In two other trials of neoadjuvant cisplatin therapy, HR deficiency predicted RCB 0/I and pCR (OR = 10.18, P = 0.0011; OR = 17.00, P = 0.0066). In a multivariable model of RCB 0/I, HR deficiency retained significance when clinical variables were included (OR = 12.08, P = 0.0017). When restricted to BRCA1/2 nonmutated tumors, response was higher in patients with high HRD scores: RCB 0/I P = 0.062, pCR P = 0.063 in the neoadjuvant platinum, gemcitabine, and iniparib trial; RCB 0/I P = 0.0039, pCR P = 0.018 in the neoadjuvant cisplatin trials. Conclusions: HR deficiency identifies TNBC tumors, including BRCA1/2 nonmutated tumors more likely to respond to platinum-containing therapy. Clin Cancer Res; 22(15); 3764–73. ©2016 AACR.


PLOS ONE | 2010

The Human-Bacterial Pathogen Protein Interaction Networks of Bacillus anthracis, Francisella tularensis, and Yersinia pestis

Matthew D. Dyer; Chris Neff; Max Dufford; Corban G. Rivera; Donna Shattuck; Josep Bassaganya-Riera; T. M. Murali; Bruno W. S. Sobral

Background Bacillus anthracis, Francisella tularensis, and Yersinia pestis are bacterial pathogens that can cause anthrax, lethal acute pneumonic disease, and bubonic plague, respectively, and are listed as NIAID Category A priority pathogens for possible use as biological weapons. However, the interactions between human proteins and proteins in these bacteria remain poorly characterized leading to an incomplete understanding of their pathogenesis and mechanisms of immune evasion. Methodology In this study, we used a high-throughput yeast two-hybrid assay to identify physical interactions between human proteins and proteins from each of these three pathogens. From more than 250,000 screens performed, we identified 3,073 human-B. anthracis, 1,383 human-F. tularensis, and 4,059 human-Y. pestis protein-protein interactions including interactions involving 304 B. anthracis, 52 F. tularensis, and 330 Y. pestis proteins that are uncharacterized. Computational analysis revealed that pathogen proteins preferentially interact with human proteins that are hubs and bottlenecks in the human PPI network. In addition, we computed modules of human-pathogen PPIs that are conserved amongst the three networks. Functionally, such conserved modules reveal commonalities between how the different pathogens interact with crucial host pathways involved in inflammation and immunity. Significance These data constitute the first extensive protein interaction networks constructed for bacterial pathogens and their human hosts. This study provides novel insights into host-pathogen interactions.


Breast Cancer Research | 2014

Association of BRCA1/2 defects with genomic scores predictive of DNA damage repair deficiency among breast cancer subtypes

Kirsten Timms; Victor Abkevich; Elisha Hughes; Chris Neff; Julia Reid; Brian Morris; Saritha Kalva; Jennifer Potter; Thanh Tran; Jian Chen; Diana Iliev; Zaina Sangale; Eliso Tikishvili; Michael Perry; Andrey Zharkikh; Alexander Gutin; Jerry S. Lanchbury

IntroductionHomologous recombination (HR) DNA repair is of clinical relevance in breast cancer. Three DNA-based homologous recombination deficiency (HRD) scores (HRD-loss of heterozygosity score (LOH), HRD-telomeric allelic imbalance score (TAI), and HRD-large-scale state transition score (LST)) have been developed that are highly correlated with defects in BRCA1/2, and are associated with response to platinum therapy in triple negative breast and ovarian cancer. This study examines the frequency of BRCA1/2 defects among different breast cancer subtypes, and the ability of the HRD scores to identify breast tumors with defects in the homologous recombination DNA repair pathway.Methods215 breast tumors representing all ER/HER2 subtypes were obtained from commercial vendors. Next-generation sequencing based assays were used to generate genome wide SNP profiles, BRCA1/2 mutation screening, and BRCA1 promoter methylation data.ResultsBRCA1/2 deleterious mutations were observed in all breast cancer subtypes. BRCA1 promoter methylation was observed almost exclusively in triple negative breast cancer. BRCA1/2 deficient tumors were identified with BRCA1/2 mutations, or BRCA1 promoter methylation, and loss of the second allele of the affected gene. All three HRD scores were highly associated with BRCA1/2 deficiency (HRD-LOH: P = 1.3 × 10-17; HRD-TAI: P = 1.5 × 10-19; HRD-LST: P = 3.5 × 10-18). A combined score (HRD-mean) was calculated using the arithmetic mean of the three scores. In multivariable analyses the HRD-mean score captured significant BRCA1/2 deficiency information not captured by the three individual scores, or by clinical variables (P values for HRD-Mean adjusted for HRD-LOH: P = 1.4 × 10-8; HRD-TAI: P = 2.9 × 10-7; HRD-LST: P = 2.8 × 10-8; clinical variables: P = 1.2 × 10-16).ConclusionsThe HRD scores showed strong correlation with BRCA1/2 deficiency regardless of breast cancer subtype. The frequency of elevated scores suggests that a significant proportion of all breast tumor subtypes may carry defects in the homologous recombination DNA repair pathway. The HRD scores can be combined to produce a more robust predictor of HRD. The combination of a robust score, and the FFPE compatible assay described in this study, may facilitate use of agents targeting homologous recombination DNA repair in the clinical setting.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Proteomic screening of variola virus reveals a unique NF-κB inhibitor that is highly conserved among pathogenic orthopoxviruses

Mohamed R. Mohamed; Masmudur M. Rahman; Jerry S. Lanchbury; Donna Shattuck; Chris Neff; Max Dufford; Nicholas van Buuren; Katharine Fagan; Michele Barry; Scott A. Smith; Inger K. Damon; Grant McFadden

Identification of the binary interactions between viral and host proteins has become a valuable tool for investigating viral tropism and pathogenesis. Here, we present the first systematic protein interaction screening of the unique variola virus proteome by using yeast 2-hybrid screening against a variety of human cDNA libraries. Several protein–protein interactions were identified, including an interaction between variola G1R, an ankryin/F-box containing protein, and human nuclear factor kappa-B1 (NF-κB1)/p105. This represents the first direct interaction between a pathogen-encoded protein and NF-κB1/p105. Orthologs of G1R are present in a variety of pathogenic orthopoxviruses, but not in vaccinia virus, and expression of any one of these viral proteins blocks NF-κB signaling in human cells. Thus, proteomic screening of variola virus has the potential to uncover modulators of the human innate antiviral responses.


Journal of Proteome Research | 2009

Analysis of Vaccinia Virus−Host Protein−Protein Interactions: Validations of Yeast Two-Hybrid Screenings

Leiliang Zhang; Nancy Y. Villa; Masmudur M. Rahman; Sherin Smallwood; Donna Shattuck; Chris Neff; Max Dufford; Jerry S. Lanchbury; Joshua LaBaer; Grant McFadden

Vaccinia virus, a large double-stranded DNA virus, is the prototype of the Orthopoxvirus genus, which includes several pathogenic poxviruses of humans, such as monkeypox virus and variola virus. Here, we report a comprehensive yeast two-hybrid (Y2H) screening for the protein−protein interactions between vaccinia and human proteins. A total of 109 novel vaccinia−human protein interactions were detected among 33 viral proteins. To validate subsets of those interactions, we constructed an ORFeome library of vaccinia virus strain WR using the Gateway plasmid cloning system. By co-expressing selected vaccinia and host proteins in a variety of expression systems, we found that at least 17 of the Y2H hits identified between vaccinia and human proteins can be verified by independent methods using GST pull-down assays, representing a 63% validation rate for the Y2H hits examined (17/27). Because the cloned ORFs are conveniently transferable from the entry vectors to various destination expression vectors, the vaccinia ORFeome library will be a useful resource for future high-throughput functional proteomic experiments.


Journal of Virology | 2009

The Myxoma Virus M-T5 Ankyrin Repeat Host Range Protein Is a Novel Adaptor That Coordinately Links the Cellular Signaling Pathways Mediated by Akt and Skp1 in Virus-Infected Cells

Steven J. Werden; Jerry S. Lanchbury; Donna Shattuck; Chris Neff; Max Dufford; Grant McFadden

ABSTRACT Most poxviruses express multiple proteins containing ankyrin (ANK) repeats accounting for a large superfamily of related but unique determinants of poxviral tropism. Recently, select members of this novel family of poxvirus proteins have drawn considerable attention for their potential roles in modulating intracellular signaling networks during viral infection. The rabbit-specific poxvirus, myxoma virus (MYXV), encodes four unique ANK repeat proteins, termed M-T5, M148, M149, and M150, all of which include a carboxy-terminal PRANC domain which closely resembles a cellular protein motif called the F-box domain. Here, we show that each MYXV-encoded ANK repeat protein, including M-T5, interacts directly with the Skp1 component of the host SCF ubiquitin ligase complex, and that the binding of M-T5 to cullin 1 is indirect via binding to Skp1 in the host SCF complex. To understand the significance of these virus-host protein interactions, the various binding domains of M-T5 were mapped. The N-terminal ANK repeats I and II were identified as being important for interaction with Akt, whereas the C-terminal PRANC/F-box-like domain was essential for binding to Skp1. We also report that M-T5 can bind Akt and the host SCF complex (via Skp1) simultaneously in MYXV-infected cells. Finally, we report that M-T5 specifically mediates the relocalization of Akt from the nucleus to the cytoplasm during infection with the wild-type MYXV, but not the M-T5 knockout version of the virus. These results indicate that ANK/PRANC proteins play a critical role in reprogramming disparate cellular signaling cascades to establish a new cellular environment more favorable for virus replication.


American Journal of Medical Genetics | 2009

Evidence for epistasis between SLC6A4 and a chromosome 4 gene as risk factors in major depression

Chris Neff; Victor Abkevich; Jennifer Potter; Robin Riley; Donna Shattuck; David A. Katz

Linkage analysis on Utah pedigrees with strong family histories of major depression including only cases with the SLC6A4 HTTLPR short allele revealed a linkage peak on chromosome 4 (maximum HLOD = 3.5). This evidence suggests epistasis between SLC6A4 and an unknown gene as risk factors for major depression.


Annals of Oncology | 2014

884PDNEXT GENERATION SEQUENCING OF BRCA1/2 IN HIGH GRADE OVARIAN TUMORS EXPANDS BRCA DEFECTS BEYOND GERMLINE MUTATIONS

M. Yates; Kirsten Timms; M. Daniels; B. Batte; K. Ring; Chris Neff; Jennifer Potter; S. Chau; J. Chen; D. Williams; M. Perry; B. Morris; Alexander Gutin; Y. Amin; Mark F. Munsell; Kathleen M. Schmeler; Jerry S. Lanchbury; Karen H. Lu

ABSTRACT Aim: Germline BRCA1/2 mutation screening is critical in ovarian cancer patients to identify a subset of women who might benefit from PARP inhibitors. A recent clinical study suggests that women with either germline or somatic BRCA1/2 mutations may benefit from PARP inhibitors1. Tumor mutation screening could expand the number of ovarian cancer patients that might consider PARP inhibitor treatment2. Methods: Previously untreated high grade epithelial ovarian cancer patients were prospectively enrolled between January 2011 and December 2013 in accordance with IRB protocol. Patients were offered germline and tumor BRCA1/2 mutation screening. Germline mutation screening was performed on DNA from blood via custom amplicon assay and next generation sequencing. DNA from FFPE tumor samples was sequenced using custom hybridization enrichment and next generation sequencing. Variants and large rearrangements were identified and classified according to established criteria. Results: 263 patients were enrolled and germline testing was completed. Germline mutation analysis revealed 39 deleterious mutations (15% mutation rate, 25 BRCA1 and 14 BRCA2 mutations). Somatic tumor analysis has been completed for 88 patients receiving upfront surgical treatment (out of 123 total upfront surgery patients). 14 (16%) had germline deleterious mutations and all 14 mutations were independently identified in the tumor, giving 100% concordance between the assays. An additional 8 (9%) deleterious somatic mutations were identified, representing an overall BRCA1/2 mutation rate of 25%. One somatic BRCA2 mutation was observed in a patient with a previously identified germline BRCA2 mutation. Conclusions: Data from this prospective cohort support previously reported frequencies of BRCA1/2 deleterious mutations in high grade epithelial ovarian tumors2. Next generation sequencing assays on blood and tumor DNA were 100% concordant. Tumor sequencing identified additional BRCA1/2 defects. Further molecular profiling and clinical follow-up is ongoing to evaluate outcomes in these subgroups of ovarian cancer patients with BRCA1/2 defects. References 1. Ledermann et al, 2013 ASCO Annual Meeting 2. Hennessy et al, 2010 JCO 28:3570-6. Disclosure: K. Timms, C. Neff, J. Potter, S. Chau, J. Chen, D. Williams, M. Perry, B. Morris, A. Gutin and J. Lanchbury: is an employee and stockholder of Myriad Genetics, Inc.; K. Lu: Participating MD Anderson researchers do not receive funds from Myriad Genetics, Inc.All other authors have declared no conflicts of interest.

Collaboration


Dive into the Chris Neff's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge