Network


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

Hotspot


Dive into the research topics where Michael B. Major is active.

Publication


Featured researches published by Michael B. Major.


Development | 2006

Distinct Wnt signaling pathways have opposing roles in appendage regeneration.

Cristi L. Stoick-Cooper; Gilbert Weidinger; Kimberly J. Riehle; Charlotte Hubbert; Michael B. Major; Nelson Fausto; Randall T. Moon

In contrast to mammals, lower vertebrates have a remarkable capacity to regenerate complex structures damaged by injury or disease. This process, termed epimorphic regeneration, involves progenitor cells created through the reprogramming of differentiated cells or through the activation of resident stem cells. Wnt/β-catenin signaling regulates progenitor cell fate and proliferation during embryonic development and stem cell function in adults, but its functional involvement in epimorphic regeneration has not been addressed. Using transgenic fish lines, we show that Wnt/β-catenin signaling is activated in the regenerating zebrafish tail fin and is required for formation and subsequent proliferation of the progenitor cells of the blastema. Wnt/β-catenin signaling appears to act upstream of FGF signaling, which has recently been found to be essential for fin regeneration. Intriguingly, increased Wnt/β-catenin signaling is sufficient to augment regeneration, as tail fins regenerate faster in fish heterozygous for a loss-of-function mutation in axin1, a negative regulator of the pathway. Likewise, activation of Wnt/β-catenin signaling by overexpression of wnt8 increases proliferation of progenitor cells in the regenerating fin. By contrast, overexpression of wnt5b (pipetail) reduces expression of Wnt/β-catenin target genes, impairs proliferation of progenitors and inhibits fin regeneration. Importantly, fin regeneration is accelerated in wnt5b mutant fish. These data suggest that Wnt/β-catenin signaling promotes regeneration, whereas a distinct pathway activated by wnt5b acts in a negative-feedback loop to limit regeneration.


Science | 2007

Wilms tumor suppressor WTX negatively regulates WNT/β-catenin signaling

Michael B. Major; Nathan D. Camp; Jason D. Berndt; Xianhua Yi; Seth J. Goldenberg; Charlotte Hubbert; Travis L. Biechele; Anne-Claude Gingras; Ning Zheng; Michael J. MacCoss; Stephane Angers; Randall T. Moon

Aberrant WNT signal transduction is involved in many diseases. In colorectal cancer and melanoma, mutational disruption of proteins involved in the degradation of β-catenin, the key effector of the WNT signaling pathway, results in stabilization of β-catenin and, in turn, activation of transcription. We have used tandem-affinity protein purification and mass spectrometry to define the protein interaction network of the β-catenin destruction complex. This assay revealed that WTX, a protein encoded by a gene mutated in Wilms tumors, forms a complex with β-catenin, AXIN1, β-TrCP2 (β-transducin repeat–containing protein 2), and APC (adenomatous polyposis coli). Functional analyses in cultured cells, Xenopus, and zebrafish demonstrate that WTX promotes β-catenin ubiquitination and degradation, which antagonize WNT/β-catenin signaling. These data provide a possible mechanistic explanation for the tumor suppressor activity of WTX.


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

Activated Wnt/ß-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model

Andy J. Chien; Erin C. Moore; Anke S. Lonsdorf; Rima M. Kulikauskas; Bonnie E. Gould Rothberg; Aaron J. Berger; Michael B. Major; Sam T. Hwang; David L. Rimm; Randall T. Moon

This study demonstrates that in malignant melanoma, elevated levels of nuclear ß-catenin in both primary tumors and metastases correlate with reduced expression of a marker of proliferation and with improved survival, in contrast to colorectal cancer. The reduction in proliferation observed in vivo is recapitulated in B16 murine melanoma cells and in human melanoma cell lines cultured in vitro with either WNT3A or small-molecule activators of ß-catenin signaling. Consistent with these results, B16 melanoma cells expressing WNT3A also exhibit decreased tumor size and decreased metastasis when implanted into mice. Genome-wide transcriptional profiling reveals that WNT3A up-regulates genes implicated in melanocyte differentiation, several of which are down-regulated with melanoma progression. These findings suggest that WNT3A can mediate transcriptional changes in melanoma cells in a manner reminiscent of the known role of Wnt/ß-catenin signaling in normal melanocyte development, thereby altering melanoma cell fate to one that may be less proliferative and potentially less aggressive. Our results may explain the observed loss of nuclear ß-catenin with melanoma progression in human tumors, which could reflect a dysregulation of cellular differentiation through a loss of homeostatic Wnt/ß-catenin signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Common genetic variation within the Low-Density Lipoprotein Receptor-Related Protein 6 and late-onset Alzheimer's disease

Giancarlo V. De Ferrari; Andreas Papassotiropoulos; Travis L. Biechele; Fabienne Wavrant DeVrieze; Miguel E. Avila; Michael B. Major; Amanda J. Myers; Katia Sáez; Juan Pablo Henríquez; Alice Zhao; M. Axel Wollmer; Roger M. Nitsch; Christoph Hock; Christopher Morris; John Hardy; Randall T. Moon

Genome-wide linkage studies have defined a broad susceptibility region for late-onset Alzheimers disease on chromosome 12, which contains the Low-Density Lipoprotein Receptor-Related Protein 6 (LRP6) gene, a coreceptor for Wnt signaling. Here, we report the association between common LRP6 variants and late-onset Alzheimers disease in a multicenter case-control series as well as in a large family-based series ascertained by the National Institute of Mental Health–National Institute on Aging Genetics Initiative. As shown in the genome-wide linkage studies, our association depends mainly on apolipoprotein E-ε4 (APOE-ε4) carrier status. Haplotype tagging single-nucleotide polymorphisms (SNPs) with a set of seven allelic variants of LRP6 identified a putative risk haplotype, which includes a highly conserved coding sequence SNP: Ile-1062 → Val. Functional analyses revealed that the associated allele Val-1062, an allele previously linked to low bone mass, has decreased β-catenin signaling in HEK293T cells. Our study unveils a genetic relationship between LRP6 and APOE and supports the hypothesis that altered Wnt/β-catenin signaling may be involved in this neurodegenerative disease.


Science Signaling | 2008

New Regulators of Wnt/β-Catenin Signaling Revealed by Integrative Molecular Screening

Michael B. Major; Brian Roberts; Jason D. Berndt; Shane Marine; Jamie N. Anastas; Namjin Chung; Marc Ferrer; Xian Hua Yi; Cristi L. Stoick-Cooper; Priska D. von Haller; Lorna S. Kategaya; Andy J. Chien; Stephane Angers; Michael J. MacCoss; Michele A. Cleary; William T. Arthur; Randall T. Moon

Integration of protein-protein interaction networks and human genome-wide RNAi screens produces mechanistic insight into Wnt/β-catenin signaling. Finding the Right Candidate A genome-wide RNAi screen in human colon cancer cells, followed by two additional validation steps, reveals new components of the Wnt pathway. Combining RNAi analysis with protein-protein interaction data provides a powerful approach that not only identifies new players in a signaling pathway, but also provides functional insight about the modulators, leading to the generation of testable hypotheses. The identification and characterization of previously unidentified signal transduction molecules has expanded our understanding of biological systems and facilitated the development of mechanism-based therapeutics. We present a highly validated small interfering RNA (siRNA) screen that functionally annotates the human genome for modulation of the Wnt/β-catenin signal transduction pathway. Merging these functional data with an extensive Wnt/β-catenin protein interaction network produces an integrated physical and functional map of the pathway. The power of this approach is illustrated by the positioning of siRNA screen hits into discrete physical complexes of proteins. Similarly, this approach allows one to filter discoveries made through protein-protein interaction screens for functional contribution to the phenotype of interest. Using this methodology, we characterized AGGF1 as a nuclear chromatin-associated protein that participates in β-catenin–mediated transcription in human colon cancer cells.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Small-molecule synergist of the Wnt/β-catenin signaling pathway

Qisheng Zhang; Michael B. Major; Shinichi Takanashi; Nathan D. Camp; Naoyuki Nishiya; Eric C. Peters; Mark H. Ginsberg; Xiaoying Jian; Paul A. Randazzo; Peter G. Schultz; Randall T. Moon; Sheng Ding

The Wnt/β-catenin signaling pathway regulates cell fate and behavior during embryogenesis, adult tissue homeostasis, and regeneration. When inappropriately activated, the pathway has been linked to colorectal cancer and melanoma, and when attenuated it may contribute to Alzheimers disease and osteoporosis. Small molecules that modulate Wnt signaling will likely provide new insights into the regulation of this key developmental pathway and ultimately provide pharmacological agents to control Wnt signaling in vivo. To this end, we screened a library of 100,000 small molecules for activity in a cell-based assay of Wnt/β-catenin signaling and discovered a purine derivative, QS11, that synergizes with Wnt-3a ligand in the activation of Wnt/β-catenin signal transduction. Through affinity chromatography and subsequent functional assays, we showed that QS11 binds and inhibits the GTPase activating protein of ADP-ribosylation factor 1 (ARFGAP1), suggesting that QS11 modulates Wnt/β-catenin signaling through an effect on protein trafficking. Consistent with its function as an ARFGAP inhibitor, QS11 inhibits migration of ARFGAP overexpressing breast cancer cells.


Journal of Biological Chemistry | 2012

Wilms Tumor Gene on X Chromosome (WTX) Inhibits Degradation of NRF2 Protein through Competitive Binding to KEAP1 Protein

Nathan D. Camp; Richard G. James; David W. Dawson; Feng Yan; James M. Davison; Scott A. Houck; Xiaobo Tang; Ning Zheng; Michael B. Major; Randall T. Moon

Background: KEAP1 is a ubiquitin ligase adaptor that promotes the ubiquitination and degradation of NRF2, a transcription factor that drives the antioxidant response. Results: Wilms tumor gene on the X chromosome (WTX) stabilizes NRF2 by competing with NRF2 for binding to KEAP1. Conclusion: WTX regulates the antioxidant response. Significance: This study reveals a novel regulatory mechanism governing the antioxidant response. WTX is a tumor suppressor protein that is lost or mutated in up to 30% of cases of Wilms tumor. Among its known functions, WTX interacts with the β-transducin repeat containing family of ubiquitin ligase adaptors and promotes the ubiquitination and degradation of the transcription factor β-catenin, a key control point in the WNT/β-catenin signaling pathway. Here, we report that WTX interacts with a second ubiquitin ligase adaptor, KEAP1, which functions to regulate the ubiquitination of the transcription factor NRF2, a key control point in the antioxidant response. Surprisingly, we find that unlike its ability to promote the ubiquitination of β-catenin, WTX inhibits the ubiquitination of NRF2. WTX and NRF2 compete for binding to KEAP1, and thus loss of WTX leads to rapid ubiquitination and degradation of NRF2 and a reduced response to cytotoxic insult. These results expand our understanding of the molecular mechanisms of WTX and reveal a novel regulatory mechanism governing the antioxidant response.


Science Signaling | 2009

Bruton's tyrosine kinase revealed as a negative regulator of wnt-b-catenin signaling

Richard G. James; Travis L. Biechele; William H. Conrad; Nathan D. Camp; Daniel M. Fass; Michael B. Major; Karen Sommer; Xianhua Yi; Brian Roberts; Michele A. Cleary; William T. Arthur; Michael J. MacCoss; David J. Rawlings; Stephen J. Haggarty; Randall T. Moon

Combining an siRNA screen with a small-molecule screen reveals BTK as a nuclear inhibitor of the Wnt–β-catenin pathway. BTK Checks Wnt–β-Catenin–Mediated Gene Expression Dysregulated Wnt signaling is associated with several human diseases. James et al. now connect the Wnt–β-catenin pathway to Bruton’s tyrosine kinase, which is encoded by the gene responsible for X-linked agammaglobulinemia, a disease associated with decreased ability to fight infection due to a deficiency in B cells. By combining a small-molecule screen with a targeted siRNA screen, BTK was identified as an inhibitor of β-catenin–mediated gene expression. BTK did not alter the abundance of β-catenin in the presence or absence of Wnt; instead, it appeared to influence the stability of CDC73, a constituent of the PAF elongation complex and known binding partner of β-catenin. In B cells, CDC73 also inhibited β-catenin–mediated gene expression and BTK may act through this nuclear protein to restrain β-catenin’s transcriptional activity. Wnts are secreted ligands that activate several receptor-mediated signal transduction cascades. Homeostatic Wnt signaling through β-catenin is required in adults, because either elevation or attenuation of β-catenin function has been linked to diverse diseases. To contribute to the identification of both protein and pharmacological regulators of this pathway, we describe a combinatorial screen that merged data from a high-throughput screen of known bioactive compounds with an independent focused small interfering RNA screen. Each screen independently revealed Bruton’s tyrosine kinase (BTK) as an inhibitor of Wnt–β-catenin signaling. Loss of BTK function in human colorectal cancer cells, human B cells, zebrafish embryos, and cells derived from X-linked agammaglobulinemia patients with a mutant BTK gene resulted in elevated Wnt–β-catenin signaling, confirming that BTK acts as a negative regulator of this pathway. From affinity purification–mass spectrometry and biochemical binding studies, we found that BTK directly interacts with a nuclear component of Wnt–β-catenin signaling, CDC73. Further, we show that BTK increased the abundance of CDC73 in the absence of stimulation and that CDC73 acted as a repressor of β-catenin–mediated transcription in human colorectal cancer cells and B cells.


PLOS ONE | 2012

WIKI4, a Novel Inhibitor of Tankyrase and Wnt/ß-Catenin Signaling

Richard G. James; Kathryn C. Davidson; Katherine A. Bosch; Travis L. Biechele; Nicholas C. Robin; Russell J. Taylor; Michael B. Major; Nathan D. Camp; Kerry W. Fowler; Timothy J. Martins; Randall T. Moon

The Wnt/ß-catenin signaling pathway controls important cellular events during development and often contributes to disease when dysregulated. Using high throughput screening we have identified a new small molecule inhibitor of Wnt/ß-catenin signaling, WIKI4. WIKI4 inhibits expression of ß-catenin target genes and cellular responses to Wnt/ß-catenin signaling in cancer cell lines as well as in human embryonic stem cells. Furthermore, we demonstrate that WIKI4 mediates its effects on Wnt/ß-catenin signaling by inhibiting the enzymatic activity of TNKS2, a regulator of AXIN ubiquitylation and degradation. While TNKS has previously been shown to be the target of small molecule inhibitors of Wnt/ß-catenin signaling, WIKI4 is structurally distinct from previously identified TNKS inhibitors.


Oncogene | 2014

WNT7B mediates autocrine Wnt/β-catenin signaling and anchorage-independent growth in pancreatic adenocarcinoma.

Michael D. Arensman; Anne N. Kovochich; Rima M. Kulikauskas; Anna R. Lay; Pei-Tzu Yang; Xinmin Li; Timothy R. Donahue; Michael B. Major; Randall T. Moon; Andy J. Chien; David W. Dawson

Developmental and cancer models show Wnt/β-catenin-dependent signaling mediates diverse phenotypic outcomes in the pancreas that are dictated by context, duration and strength of activation. While generally assumed to be pro-tumorigenic, it is unclear to what extent dysregulation of Wnt/β-catenin signaling impacts tumor progression in pancreatic adenocarcinoma (PDAC). In the present study, Wnt/β-catenin activity was characterized across a spectrum of PDAC cell lines and primary tumors. Reporter and gene expression-based assays revealed wide heterogeneity in Wnt/β-catenin transcriptional activity across PDAC cell lines and patient tumors, as well as variable responsiveness to exogenous Wnt ligand stimulation. An experimentally generated, pancreas-specific gene expression signature of Wnt/β-catenin transcriptional activation was used to stratify pathway activation across a cohort of resected, early-stage PDAC tumors (N=41). In this cohort, higher Wnt/β-catenin activation was found to significantly correlate with lymphvascular invasion and worse disease-specific survival (median survival time 20.3 versus 43.9 months, log-rank P=0.03). Supporting the importance of Wnt ligand in mediating autocrine Wnt signaling, Wnt/β-catenin activity was significantly inhibited in PDAC cell lines by WLS gene silencing and the small-molecule inhibitor IWP-2, both of which functionally block Wnt ligand processing and secretion. Transcriptional profiling revealed elevated expression of WNT7B occurred in PDAC cell lines with high levels of cell autonomous Wnt/β-catenin activity. Gene-knockdown studies in AsPC-1 and HPAF-2 cell lines confirmed WNT7B-mediated cell autonomous Wnt/β-catenin activation, as well as an anchorage-independent growth phenotype. Our findings indicate WNT7B can serve as a primary determinant of differential Wnt/β-catenin activation in PDAC. Disrupting the interaction between Wnt ligands and their receptors may be a particularly suitable approach for therapeutic modulation of Wnt/β-catenin signaling in PDAC and other cancer contexts where Wnt activation is mediated by ligand expression rather than mutations in canonical pathway members.

Collaboration


Dive into the Michael B. Major's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dennis Goldfarb

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Feng Yan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Nathan D. Camp

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Matthew P. Walker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Priscila F. Siesser

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andy J. Chien

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge