Network


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

Hotspot


Dive into the research topics where Utpal P. Davé is active.

Publication


Featured researches published by Utpal P. Davé.


Molecular Cell | 2000

ER Stress Induces Cleavage of Membrane-Bound ATF6 by the Same Proteases that Process SREBPs

Jin Ye; Robert B. Rawson; Ryutaro Komuro; Xi Chen; Utpal P. Davé; Ron Prywes; Michael S. Brown; Joseph L. Goldstein

ATF6 is a membrane-bound transcription factor that activates genes in the endoplasmic reticulum (ER) stress response. When unfolded proteins accumulate in the ER, ATF6 is cleaved to release its cytoplasmic domain, which enters the nucleus. Here, we show that ATF6 is processed by Site-1 protease (S1P) and Site-2 protease (S2P), the enzymes that process SREBPs in response to cholesterol deprivation. ATF6 processing was blocked completely in cells lacking S2P and partially in cells lacking S1P. ATF6 processing required the RxxL and asparagine/proline motifs, known requirements for S1P and S2P processing, respectively. Cells lacking S2P failed to induce GRP78, an ATF6 target, in response to ER stress. ATF6 processing did not require SCAP, which is essential for SREBP processing. We conclude that S1P and S2P are required for the ER stress response as well as for lipid synthesis.


Journal of Biological Chemistry | 1998

Second-site Cleavage in Sterol Regulatory Element-binding Protein Occurs at Transmembrane Junction as Determined by Cysteine Panning

Elizabeth A. Duncan; Utpal P. Davé; Juro Sakai; Joseph L. Goldstein; Michael S. Brown

In response to sterol deprivation, two sequential proteolytic cleavages release the NH2-terminal fragments of sterol regulatory element-binding proteins (SREBPs) from cell membranes. The fragments translocate to the nucleus where they activate genes involved in cholesterol and fatty acid metabolism. The SREBPs are bound to membranes in a hairpin fashion. The NH2-terminal and COOH-terminal domains face the cytoplasm, separated by two membrane spanning segments and a short lumenal loop. The first cleavage occurs at Site-1 in the lumenal loop. The NH2-terminal fragment is then released by cleavage at Site-2, which is believed to lie within the first transmembrane segment. Here, we use a novel cysteine panning method to identify the second cleavage site (Site-2) in human SREBP-2 as the Leu484-Cys485 bond that lies at the junction between the cytoplasmic NH2-terminal fragment and the first transmembrane segment. We transfected cells with cDNAs encoding fusion proteins with single cysteine residues at positions to the NH2-terminal and COOH-terminal sides of cysteine 485. The NH2-terminal fragments were tested for susceptibility to modification withN α-(3-maleimidylpropionyl)biocytin, which attaches a biotin group to cysteine sulfhydryls. Cysteines to the NH2-terminal side of cysteine 485 were retained on the NH2-terminal fragment, but cysteines to the COOH-terminal side of leucine 484 were lost. Leucine 484 is three residues to the COOH-terminal side of the tetrapeptide Asp-Arg-Ser-Arg, which immediately precedes the first transmembrane segment and is required for Site-2 cleavage.


Blood | 2015

Whole-genome sequencing reveals oncogenic mutations in mycosis fungoides

Laura Y. McGirt; Peilin Jia; Devin A. Baerenwald; Robert J. Duszynski; Kimberly B. Dahlman; John A. Zic; Jeffrey P. Zwerner; Donald Hucks; Utpal P. Davé; Zhongming Zhao; Christine M. Eischen

The pathogenesis of mycosis fungoides (MF), the most common cutaneous T-cell lymphoma (CTCL), is unknown. Although genetic alterations have been identified, none are considered consistently causative in MF. To identify potential drivers of MF, we performed whole-genome sequencing of MF tumors and matched normal skin. Targeted ultra-deep sequencing of MF samples and exome sequencing of CTCL cell lines were also performed. Multiple mutations were identified that affected the same pathways, including epigenetic, cell-fate regulation, and cytokine signaling, in MF tumors and CTCL cell lines. Specifically, interleukin-2 signaling pathway mutations, including activating Janus kinase 3 (JAK3) mutations, were detected. Treatment with a JAK3 inhibitor significantly reduced CTCL cell survival. Additionally, the mutation data identified 2 other potential contributing factors to MF, ultraviolet light, and a polymorphism in the tumor suppressor p53 (TP53). Therefore, genetic alterations in specific pathways in MF were identified that may be viable, effective new targets for treatment.


PLOS Genetics | 2009

Murine Leukemias with Retroviral Insertions at Lmo2 Are Predictive of the Leukemias Induced in SCID-X1 Patients Following Retroviral Gene Therapy

Utpal P. Davé; Keiko Akagi; Rati Tripathi; Susan M. Cleveland; Mary Ann Thompson; Ming Yi; Robert M. Stephens; James R. Downing; Nancy A. Jenkins; Neal G. Copeland

Five X-linked severe combined immunodeficiency patients (SCID-X1) successfully treated with autologous bone marrow stem cells infected ex vivo with an IL2RG-containing retrovirus subsequently developed T-cell leukemia and four contained insertional mutations at LMO2. Genetic evidence also suggests a role for IL2RG in tumor formation, although this remains controversial. Here, we show that the genes and signaling pathways deregulated in murine leukemias with retroviral insertions at Lmo2 are similar to those deregulated in human leukemias with high LMO2 expression and are highly predictive of the leukemias induced in SCID-X1 patients. We also provide additional evidence supporting the notion that IL2RG and LMO2 cooperate in leukemia induction but are not sufficient and require additional cooperating mutations. The highly concordant nature of the genetic events giving rise to mouse and human leukemias with mutations at Lmo2 are an encouraging sign to those wanting to use mice to model human cancer and may help in designing safer methods for retroviral gene therapy.


Blood | 2011

FERM domain mutations induce gain of function in JAK3 in adult T-cell leukemia/lymphoma

Natalina Elliott; Susan M. Cleveland; Victor R. Grann; John E. Janik; Thomas A. Waldmann; Utpal P. Davé

Adult T-cell leukemia/lymphoma (ATLL) is an incurable disease where most patients succumb within the first year of diagnosis. Both standard chemotherapy regimens and mAbs directed against ATLL tumor markers do not alter this aggressive clinical course. Therapeutic development would be facilitated by the discovery of genes and pathways that drive or initiate ATLL, but so far amenable drug targets have not been forthcoming. Because the IL-2 signaling pathway plays a prominent role in ATLL pathogenesis, mutational analysis of pathway components should yield interesting results. In this study, we focused on JAK3, the nonreceptor tyrosine kinase that signals from the IL-2R, where activating mutations have been found in diverse neoplasms. We screened 36 ATLL patients and 24 ethnically matched controls and found 4 patients with mutations in JAK3. These somatic, missense mutations occurred in the N-terminal FERM (founding members: band 4.1, ezrin, radixin, and moesin) domain and induced gain of function in JAK3. Importantly, we show that these mutant JAK3s are inhibited with a specific kinase inhibitor already in human clinical testing. Our findings underscore the importance of this pathway in ATLL development and offer a therapeutic handle for this incurable cancer.


Stem Cells | 2013

Lmo2 Induces Hematopoietic Stem Cell-Like Features in T-Cell Progenitor Cells Prior to Leukemia†‡§

Susan M. Cleveland; Stephen B. Smith; Rati Tripathi; Elizabeth Mathias; Charnise Goodings; Natalina Elliott; DunFa Peng; Wael El-Rifai; Dajun Yi; Xi Chen; LiQi Li; Charles G. Mullighan; James R. Downing; Paul E. Love; Utpal P. Davé

LIM domain only 2 (Lmo2) is frequently deregulated in sporadic and gene therapy‐induced acute T‐cell lymphoblastic leukemia (T‐ALL) where its overexpression is an important initiating mutational event. In transgenic and retroviral mouse models, Lmo2 expression can be enforced in multiple hematopoietic lineages but leukemia only arises from T cells. These data suggest that Lmo2 confers clonal growth advantage in T‐cell progenitors. We analyzed proliferation, differentiation, and cell death in CD2‐Lmo2 transgenic thymic progenitor cells to understand the cellular effects of enforced Lmo2 expression. Most impressively, Lmo2 transgenic T‐cell progenitor cells were blocked in differentiation, quiescent, and immortalized in vitro on OP9‐DL1 stromal cells. These cellular effects were concordant with a transcriptional signature in Lmo2 transgenic T‐cell progenitor cells that is also present in hematopoietic stem cells (HSCs) and early T‐cell precursor ALL. These results are significant in light of the crucial role of Lmo2 in the maintenance of the HSC. The cellular effects and transcriptional effects have implications for LMO2‐dependent leukemogenesis and the treatment of LMO2‐induced T‐ALL. STEM CELLS 2013;31:882–894


PLOS ONE | 2014

LIM Domain Only-2 (LMO2) Induces T-Cell Leukemia by Two Distinct Pathways

Stephen B. Smith; Rati Tripathi; Charnise Goodings; Susan M. Cleveland; Elizabeth Mathias; J. Andrew Hardaway; Natalina Elliott; Yajun Yi; Xi Chen; James R. Downing; Charles G. Mullighan; Deborah A. Swing; Lino Tessarollo; LiQi Li; Paul E. Love; Nancy A. Jenkins; Neal G. Copeland; Mary Ann Thompson; Yang Du; Utpal P. Davé

The LMO2 oncogene is deregulated in the majority of human T-cell leukemia cases and in most gene therapy-induced T-cell leukemias. We made transgenic mice with enforced expression of Lmo2 in T-cells by the CD2 promoter/enhancer. These transgenic mice developed highly penetrant T-ALL by two distinct patterns of gene expression: one in which there was concordant activation of Lyl1, Hhex, and Mycn or alternatively, with Notch1 target gene activation. Most strikingly, this gene expression clustering was conserved in human Early T-cell Precursor ALL (ETP-ALL), where LMO2, HHEX, LYL1, and MYCN were most highly expressed. We discovered that HHEX is a direct transcriptional target of LMO2 consistent with its concordant gene expression. Furthermore, conditional inactivation of Hhex in CD2-Lmo2 transgenic mice markedly attenuated T-ALL development, demonstrating that Hhex is a crucial mediator of Lmo2s oncogenic function. The CD2-Lmo2 transgenic mice offer mechanistic insight into concordant oncogene expression and provide a model for the highly treatment-resistant ETP-ALL subtype.


Genes, Chromosomes and Cancer | 2012

Systematic screen for tyrosine kinase rearrangements identifies a novel C6orf204‐PDGFRB fusion in a patient with recurrent T‐ALL and an associated myeloproliferative neoplasm

Juliann Chmielecki; Martin Peifer; Agnes Viale; Katherine E. Hutchinson; Jennifer M. Giltnane; Nicholas D. Socci; Clayton J. Hollis; Rebecca S. Dean; Ashwini Yenamandra; Madan Jagasia; Annette S. Kim; Utpal P. Davé; Roman K. Thomas; William Pao

Gene fusions involving the catalytic domain of tyrosine kinases (TKs) are found in a variety of hematological and solid tumor malignancies. Clinically, TK fusions have emerged as prime targets for therapy with small molecule kinase inhibitors. Unfortunately, identification of TK fusions has been hampered by experimental limitations. Here, we developed version 2.0 of a genomically based systematic kinase fusion screen and used it to detect a novel imatinib‐sensitive C6orf204‐PDGFRB fusion in a patient with precursor T lymphoblastic lymphoma (T‐ALL) and an associated myeloproliferative neoplasm with eosinophilia. These data validate the ability of this targeted capture‐sequencing approach to detect TK fusion events in small amounts of DNA extracted directly from patient samples.


Blood | 2011

Sox4 cooperates with PU.1 haploinsufficiency in murine myeloid leukemia

Georg Aue; Yang Du; Susan M. Cleveland; Stephen B. Smith; Utpal P. Davé; Delong Liu; Marc A. Weniger; Jean Yves Metais; Nancy A. Jenkins; Neal G. Copeland; Cynthia E. Dunbar

Cooperation of multiple mutations is thought to be required for cancer development. In previous studies, murine myeloid leukemias induced by transducing wild-type bone marrow progenitors with a SRY sex determining region Y-box 4 (Sox4)-expressing retrovirus frequently carried proviral insertions at Sfpi1, decreasing its mRNA levels, suggesting that reduced Sfpi1 expression cooperates with Sox4 in myeloid leukemia induction. In support of this hypothesis, we show here that mice receiving Sox4 virus-infected Sfpi1(ko/+) bone marrow progenitors developed myeloid leukemia with increased penetrance and shortened latency. Interestingly, Sox4 expression further decreased Sfpi1 transcription. Ectopic SOX4 expression reduced endogenous PU.1 mRNA levels in HL60 promyelocytes, and decreased Sfpi1 mRNA levels were also observed in the spleens of leukemic and preleukemic mice receiving Sox4 virus-infected wild-type bone marrow cells. In addition, Sox4 protein bound to a critical upstream regulatory element of Sfpi1 in ChIP assays. Such cooperation probably occurs in de novo human acute myeloid leukemias, as an analysis of 285 acute myeloid leukemia patient samples found a significant negative correlation between SOX4 and PU.1 expression. Our results establish a novel cooperation between Sox4 and reduced Sfpi1 expression in myeloid leukemia development and suggest that SOX4 could be an important new therapeutic target in human acute myeloid leukemia.


Molecular and Cellular Biology | 2013

Tgif1 regulates quiescence and self-renewal of hematopoietic stem cells

Ling Yan; Bethany Womack; David Wotton; Yan Guo; Yu Shyr; Utpal P. Davé; Chun Li; Scott W. Hiebert; Stephen J. Brandt; Rizwan Hamid

ABSTRACT TG-interacting factor 1 (TGIF1) is a transcriptional repressor that can modulate retinoic acid and transforming growth factor β signaling pathways. It is required for myeloid progenitor cell differentiation and survival, and mutations in the TGIF1 gene cause holoprosencephaly. Furthermore, we have previously observed that acute myelogenous leukemia (AML) patients with low TGIF1 levels had worse prognoses. Here, we explored the role of Tgif1 in murine hematopoietic stem cell (HSC) function. CFU assays showed that Tgif1−/− bone marrow cells produced more total colonies and had higher serial CFU potential. These effects were also observed in vivo, where Tgif1−/− bone marrow cells had higher repopulation potential in short- and long-term competitive repopulation assays than wild-type cells. Serial transplantation and replating studies showed that Tgif1−/− HSCs exhibited greater self-renewal and were less proliferative and more quiescent than wild-type cells, suggesting that Tgif1 is required for stem cells to enter the cell cycle. Furthermore, HSCs from Tgif1+/− mice had a phenotype similar to that of HSCs from Tgif1−/− mice, while bone marrow cells with overexpressing Tgif1 showed increased proliferation and lower survival in long-term transplant studies. Taken together, our data suggest that Tgif1 suppresses stem cell self-renewal and provide clues as to how reduced expression of TGIF1 may contribute to poor long-term survival in patients with AML.

Collaboration


Dive into the Utpal P. Davé's collaboration.

Top Co-Authors

Avatar

Susan M. Cleveland

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rati Tripathi

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Natalina Elliott

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nancy A. Jenkins

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Neal G. Copeland

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Charnise Goodings

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Mathias

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mary Ann Thompson

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen B. Smith

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xi Chen

Vanderbilt University

View shared research outputs
Researchain Logo
Decentralizing Knowledge