Network


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

Hotspot


Dive into the research topics where Elsa R. Flores is active.

Publication


Featured researches published by Elsa R. Flores.


Nature | 2002

p63 and p73 are required for p53-dependent apoptosis in response to DNA damage

Elsa R. Flores; Kenneth Y. Tsai; Denise Crowley; Shomit Sengupta; Annie Yang; Frank McKeon; Tyler Jacks

The tumour-suppressor gene p53 is frequently mutated in human cancers and is important in the cellular response to DNA damage. Although the p53 family members p63 and p73 are structurally related to p53, they have not been directly linked to tumour suppression, although they have been implicated in apoptosis. Given the similarity between this family of genes and the ability of p63 and p73 to transactivate p53 target genes, we explore here their role in DNA damage-induced apoptosis. Mouse embryo fibroblasts deficient for one or a combination of p53 family members were sensitized to undergo apoptosis through the expression of the adenovirus E1A oncogene. While using the E1A system facilitated our ability to perform biochemical analyses, we also examined the functions of p63 and p73 using an in vivo system in which apoptosis has been shown to be dependent on p53. Using both systems, we show here that the combined loss of p63 and p73 results in the failure of cells containing functional p53 to undergo apoptosis in response to DNA damage.


Nature | 2000

Role for the p53 homologue p73 in E2F-1-induced apoptosis

Meredith S. Irwin; Maria Carmen Marin; Andrew C. Phillips; Ratnam S. Seelan; David I. Smith; Wanguo Liu; Elsa R. Flores; Kenneth Y. Tsai; Tyler Jacks; Karen H. Vousden; William G. Kaelin

The transcription factor E2F-1 induces both cell-cycle progression and, in certain settings, apoptosis. E2F-1 uses both p53-dependent and p53-independent pathways to kill cells. The p53-dependent pathway involves the induction by E2F-1 of the human tumour-suppressor protein p14ARF, which neutralizes HDM2 (human homologue of MDM2) and thereby stabilizes the p53 protein. Here we show that E2F-1 induces the transcription of the p53 homologue p73. Disruption of p73 function inhibited E2F-1-induced apoptosis in p53-defective tumour cells and in p53-/- mouse embryo fibroblasts. We conclude that activation of p73 provides a means for E2F-1 to induce death in the absence of p53.


Cell Stem Cell | 2009

TAp63 Prevents Premature Aging by Promoting Adult Stem Cell Maintenance

Xiaohua Su; Maryline Paris; Young Jin Gi; Kenneth Y. Tsai; Min Soon Cho; Yu Li Lin; Jeffrey Biernaskie; Satrajit Sinha; Carol Prives; Larysa Pevny; Freda D. Miller; Elsa R. Flores

The cellular mechanisms that regulate the maintenance of adult tissue stem cells are still largely unknown. We show here that the p53 family member, TAp63, is essential for maintenance of epidermal and dermal precursors and that, in its absence, these precursors senesce and skin ages prematurely. Specifically, we have developed a TAp63 conditional knockout mouse and used it to ablate TAp63 in the germline (TAp63(-/-)) or in K14-expressing cells in the basal layer of the epidermis (TAp63(fl/fl);K14cre+). TAp63(-/-) mice age prematurely and develop blisters, skin ulcerations, senescence of hair follicle-associated dermal and epidermal cells, and decreased hair morphogenesis. These phenotypes are likely due to loss of TAp63 in dermal and epidermal precursors since both cell types show defective proliferation, early senescence, and genomic instability. These data indicate that TAp63 serves to maintain adult skin stem cells by regulating cellular senescence and genomic stability, thereby preventing premature tissue aging.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Targeted point mutations of p53 lead to dominant-negative inhibition of wild-type p53 function

Annemieke de Vries; Elsa R. Flores; Barbara Miranda; Harn Mei Hsieh; Conny T. M. van Oostrom; Julien Sage; Tyler Jacks

The p53 tumor suppressor gene is the most frequently mutated gene in human cancers, and germ-line p53 mutations cause a familial predisposition for cancer. Germ-line or sporadic p53 mutations are usually missense and typically affect the central DNA-binding domain of the protein. Because p53 functions as a tetrameric transcription factor, mutant p53 is thought to inhibit the function of wild-type p53 protein. Here, we studied the possible dominant-negative inhibition of wild-type p53 protein by two different, frequently occurring point mutations. The R270H and P275S mutations were targeted into the genome of mouse embryonic stem cells to allow the analysis of the effects of the mutant proteins expressed in normal cells at single-copy levels. In embryonic stem cells, the presence of a heterozygous point-mutated allele resulted in delayed transcriptional activation of several p53 downstream target genes on exposure to γ irradiation. Doxorubicin-induced apoptosis was severely affected in the mutant embryonic stem cells compared with wild-type cells. Heterozygous mutant thymocytes had a severe defect in p53-dependent apoptotic pathways after treatment with γ irradiation or doxorubicin, whereas p53-independent apoptotic pathways were intact. Together these data demonstrate that physiological expression of point-mutated p53 can strongly limit overall cellular p53 function, supporting the dominant-negative action of such mutants. Also, cells heterozygous for such mutations may be compromised in terms of tumor suppression and response to chemotherapeutic agents.


Journal of Clinical Investigation | 2012

ZEB1 drives prometastatic actin cytoskeletal remodeling by downregulating miR-34a expression

Young Ho Ahn; Don L. Gibbons; Deepavali Chakravarti; Chad J. Creighton; Zain H. Rizvi; Henry P. Adams; Alexander Pertsemlidis; Philip A. Gregory; Josephine A. Wright; Gregory J. Goodall; Elsa R. Flores; Jonathan M. Kurie

Metastatic cancer is extremely difficult to treat, and the presence of metastases greatly reduces a cancer patients likelihood of long-term survival. The ZEB1 transcriptional repressor promotes metastasis through downregulation of microRNAs (miRs) that are strong inducers of epithelial differentiation and inhibitors of stem cell factors. Given that each miR can target multiple genes with diverse functions, we posited that the prometastatic network controlled by ZEB1 extends beyond these processes. We tested this hypothesis using a mouse model of human lung adenocarcinoma metastasis driven by ZEB1, human lung carcinoma cells, and human breast carcinoma cells. Transcriptional profiling studies revealed that ZEB1 controls the expression of numerous oncogenic and tumor-suppressive miRs, including miR-34a. Ectopic expression of miR-34a decreased tumor cell invasion and metastasis, inhibited the formation of promigratory cytoskeletal structures, suppressed activation of the RHO GTPase family, and regulated a gene expression signature enriched in cytoskeletal functions and predictive of outcome in human lung adenocarcinomas. We identified several miR-34a target genes, including Arhgap1, which encodes a RHO GTPase activating protein that was required for tumor cell invasion. These findings demonstrate that ZEB1 drives prometastatic actin cytoskeletal remodeling by downregulating miR-34a expression and provide a compelling rationale to develop miR-34a as a therapeutic agent in lung cancer patients.


Nature Reviews Cancer | 2013

P63 steps into the limelight: Crucial roles in the suppression of tumorigenesis and metastasis

Xiaohua Su; Deepavali Chakravarti; Elsa R. Flores

The role of p63 in cancer has been an area of intense debate and controversy. Is TP63 (which encodes p63) a tumour suppressor gene or an oncogene? This debate is partly due to the complexity of the gene. There are several p63 isoforms — some with tumour suppressive functions and others with oncogenic functions. In this Opinion article, we focus on the recent advances in understanding p63 biology and its roles in cancer. In this regard, we discuss the role of p63 in multiple stem cell compartments, ageing, in the response to DNA damage and in DNA repair. Finally, we highlight the importance of understanding the interactions between all three p53 family members and the potential impact of this knowledge on cancer therapy and regenerative medicine.


PLOS Genetics | 2009

p63 and p73 Transcriptionally Regulate Genes Involved in DNA Repair

Yu Li Lin; Shomit Sengupta; Katherine Gurdziel; George W. Bell; Tyler Jacks; Elsa R. Flores

The p53 family activates many of the same genes in response to DNA damage. Because p63 and p73 have structural differences from p53 and play distinct biological functions in development and metastasis, it is likely that they activate a unique transcriptional network. Therefore, we performed a genome-wide analysis using cells lacking the p53 family members after treatment with DNA damage. We identified over 100 genes involved in multiple pathways that were uniquely regulated by p63 or p73, and not p53. Further validation indicated that BRCA2, Rad51, and mre11 are direct transcriptional targets of p63 and p73. Additionally, cells deficient for p63 and p73 are impaired in DNA repair and p63+/−;p73+/− mice develop mammary tumors suggesting a novel mechanism whereby p63 and p73 suppress tumorigenesis.


Cell Metabolism | 2012

TAp63 is a master transcriptional regulator of lipid and glucose metabolism.

Xiaohua Su; Young Jin Gi; Deepavali Chakravarti; Io Long Chan; Aijun Zhang; Xuefeng Xia; Kenneth Y. Tsai; Elsa R. Flores

TAp63 prevents premature aging, suggesting a link to genes that regulate longevity. Further characterization of TAp63-/- mice revealed that these mice develop obesity, insulin resistance, and glucose intolerance similar to those seen in mice lacking two key metabolic regulators, Silent information regulator T1 (Sirt1) and AMPK. While the roles of Sirt1 and AMPK in metabolism have been well studied, their upstream regulators are not well understood. We found that TAp63 is important in regulating energy metabolism by accumulating in response to metabolic stress and transcriptionally activating Sirt1, AMPKα2, and LKB1, resulting in increased fatty acid synthesis and decreased fatty acid oxidation. Moreover, we found that TAp63 lowers blood glucose levels in response to metformin. Restoration of Sirt1, AMPKα2, and LKB1 in TAp63-/- mice rescued some of the metabolic defects of the TAp63-/- mice. Our study defines a role for TAp63 in metabolism and weight control.


The EMBO Journal | 2009

Rescue of key features of the p63-null epithelial phenotype by inactivation of Ink4a and Arf

Xiaohua Su; Min Soon Cho; Young Jin Gi; Bernard A. Ayanga; Charles J. Sherr; Elsa R. Flores

Mice lacking p63 cannot form skin, exhibit craniofacial and skeletal defects, and die soon after birth. The p63 gene regulates a complex network of target genes, and disruption of p63 has been shown to affect the maintenance of epithelial stem cells, the differentiation of keratinocytes, and the preservation of the adhesive properties of stratified epithelium. Here, we show that inactivation of p63 in mice is accompanied by aberrantly increased expression of the Ink4a and Arf tumour suppressor genes. In turn, anomalies of the p63‐null mouse affecting the skin and skeleton are partially ameliorated in mice lacking either Ink4a or Arf. Rescue of epithelialization is accompanied by restoration of keratinocyte proliferative capacity both in vivo and in vitro and by expression of markers of squamous differentiation. Thus, in the absence of p63, abnormal upregulation of Ink4a and Arf is incompatible with skin development.


Nature | 2015

IAPP-driven metabolic reprogramming induces regression of p53-deficient tumours in vivo.

Avinashnarayan Venkatanarayan; Payal Raulji; William T. Norton; Deepavali Chakravarti; Cristian Coarfa; Xiaohua Su; Santosh K. Sandur; Marc S. Ramirez; Jaehuk Lee; Charles Kingsley; Eliot Fletcher Sananikone; Kimal Rajapakshe; Jan Parker-Thornburg; James A. Bankson; Kenneth Y. Tsai; Preethi H. Gunaratne; Elsa R. Flores

TP53 is commonly altered in human cancer, and Tp53 reactivation suppresses tumours in vivo in mice (TP53 and Tp53 are also known as p53). This strategy has proven difficult to implement therapeutically, and here we examine an alternative strategy by manipulating the p53 family members, Tp63 and Tp73 (also known as p63 and p73, respectively). The acidic transactivation-domain-bearing (TA) isoforms of p63 and p73 structurally and functionally resemble p53, whereas the ΔN isoforms (lacking the acidic transactivation domain) of p63 and p73 are frequently overexpressed in cancer and act primarily in a dominant-negative fashion against p53, TAp63 and TAp73 to inhibit their tumour-suppressive functions. The p53 family interacts extensively in cellular processes that promote tumour suppression, such as apoptosis and autophagy, thus a clear understanding of this interplay in cancer is needed to treat tumours with alterations in the p53 pathway. Here we show that deletion of the ΔN isoforms of p63 or p73 leads to metabolic reprogramming and regression of p53-deficient tumours through upregulation of IAPP, the gene that encodes amylin, a 37-amino-acid peptide co-secreted with insulin by the β cells of the pancreas. We found that IAPP is causally involved in this tumour regression and that amylin functions through the calcitonin receptor (CalcR) and receptor activity modifying protein 3 (RAMP3) to inhibit glycolysis and induce reactive oxygen species and apoptosis. Pramlintide, a synthetic analogue of amylin that is currently used to treat type 1 and type 2 diabetes, caused rapid tumour regression in p53-deficient thymic lymphomas, representing a novel strategy to target p53-deficient cancers.

Collaboration


Dive into the Elsa R. Flores's collaboration.

Top Co-Authors

Avatar

Kenneth Y. Tsai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristian Coarfa

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xiaohua Su

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Napoli

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Deepavali Chakravarti

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Avinashnarayan Venkatanarayan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tyler Jacks

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Min Soon Cho

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Payal Raulji

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge