36 results on '"Hector L. Franco"'
Search Results
2. CRISPR‐based knock‐in mutagenesis of the pioneer transcription factor FOXA1: optimization of strategies for multi‐allelic proteins in cancer cells
- Author
-
Shen Li, Joseph P. Garay, Colby A. Tubbs, and Hector L. Franco
- Subjects
breast cancer ,Cas9 ,CRISPR ,FOXA1 ,knock‐in ,MCF7 ,Biology (General) ,QH301-705.5 - Abstract
Precise genome engineering of living cells has been revolutionized by the introduction of the highly specific and easily programmable properties of the clustered regularly interspaced short palindromic repeats (CRISPR) technology. This has greatly accelerated research into human health and has facilitated the discovery of novel therapeutics. CRISPR‐Cas9 is most widely employed for its ability to inactivate or knockout specific genes, but can be also used to introduce subtle site‐specific substitutions of DNA sequences that can lead to changes in the amino acid composition of proteins. Despite the proven success of CRISPR‐based knock‐in strategies of genes in typical diploid cells (i.e., cells containing two sets of chromosomes), precise editing of cancer cells, that typically have unstable genomes and multiple copies of chromosomes, is more challenging and not adequately addressed in the literature. Herein, we detail our methodology for replacing endogenous proteins with intended knock‐in mutants in polyploid cancer cells and discuss our experimental design, screening strategy, and facile allele frequency estimation methodology. As proof of principle, we performed genome editing of specific amino acids within the pioneer transcription factor FOXA1, a critical component of estrogen and androgen receptor signaling, in MCF‐7 breast cancer cells. We confirm mutant FOXA1 protein expression and intended amino acid substitutions via western blotting and mass spectrometry. In addition, we show that mutant allele frequency estimation is easily achieved by topoisomerase‐based cloning combined with allele‐specific PCR, which we later confirmed by next‐generation RNA‐sequencing. Typically, there are 4 ‐ 5 copies (alleles) of FOXA1 in breast cancer cells, making the editing of this protein inherently challenging. As a result, most studies that focus on FOXA1 mutants rely on ectopic overexpression of FOXA1 from a plasmid. Therefore, we provide an optimized methodology for replacing endogenous wild‐type FOXA1 with precise knock‐in mutants to enable the systematic analysis of its molecular mechanisms within the appropriate physiological context.
- Published
- 2021
- Full Text
- View/download PDF
3. Histone modification profiling in breast cancer cell lines highlights commonalities and differences among subtypes
- Author
-
Yuanxin Xi, Jiejun Shi, Wenqian Li, Kaori Tanaka, Kendra L. Allton, Dana Richardson, Jing Li, Hector L. Franco, Anusha Nagari, Venkat S. Malladi, Luis Della Coletta, Melissa S. Simper, Khandan Keyomarsi, Jianjun Shen, Mark T. Bedford, Xiaobing Shi, Michelle C. Barton, W. Lee Kraus, Wei Li, and Sharon Y. R. Dent
- Subjects
Breast cancer subtypes ,Epigenetics ,Histone modifications ,Chromatin states ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Epigenetic regulators are frequently mutated or aberrantly expressed in a variety of cancers, leading to altered transcription states that result in changes in cell identity, behavior, and response to therapy. Results To define alterations in epigenetic landscapes in breast cancers, we profiled the distributions of 8 key histone modifications by ChIP-Seq, as well as primary (GRO-seq) and steady state (RNA-Seq) transcriptomes, across 13 distinct cell lines that represent 5 molecular subtypes of breast cancer and immortalized human mammary epithelial cells. Discussion Using combinatorial patterns of distinct histone modification signals, we defined subtype-specific chromatin signatures to nominate potential biomarkers. This approach identified AFAP1-AS1 as a triple negative breast cancer-specific gene associated with cell proliferation and epithelial-mesenchymal-transition. In addition, our chromatin mapping data in basal TNBC cell lines are consistent with gene expression patterns in TCGA that indicate decreased activity of the androgen receptor pathway but increased activity of the vitamin D biosynthesis pathway. Conclusions Together, these datasets provide a comprehensive resource for histone modification profiles that define epigenetic landscapes and reveal key chromatin signatures in breast cancer cell line subtypes with potential to identify novel and actionable targets for treatment.
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract PD4-08: PD4-08 A Novel Single Cell Model of Tamoxifen Response in Primary Human Breast Tumors
- Author
-
Hyunsoo Kim, Austin Whitman, Kamila Wisniewska, Susana Garcia-Recio, Rasha Kakati, Hector L. Franco, Charles M. Perou, and Philip Spanheimer
- Subjects
Cancer Research ,Oncology - Abstract
A Novel Single Cell Model of Tamoxifen Response in Primary Human Breast Tumors Austin Whitman, Hyunsoo Kim, Kamila Wisniewska, Rasha Kakati, Susana Garcia Recio, Hector Franco, Charles Perou, Philip Spanheimer Background: Resistance to endocrine therapy is a primary cause of treatment failure and death in patients with estrogen receptor (ER)-positive breast cancer. Intratumor heterogeneity is associated with resistance to therapy across tumors, and specifically in ER+/HER2- breast cancer, heterogeneity in ER and PR expression is associated with a worse response to endocrine therapy. We hypothesize that subpopulations within and across ER+/HER2- human breast tumors have distinct responses to tamoxifen and that discerning heterogeneity in response will improve understanding of inherent and emerging resistance to endocrine therapy. Methods: We developed an operating room-to-laboratory pipeline immediately after surgical resection for studies using alive tissue. Tissue samples were obtained and single cell suspensions created using physical and enzymatic dissociation. Cells were treated with tamoxifen (10 M) or control media for 12 hours in suspension and single cell RNA libraries generated using the 10X Genomics droplet-based kit and sequenced using the Illumina NextSeq2000. Results: We obtained normal breast tissue from 2 women undergoing reduction mammoplasty and tumor tissue from 10 women with ER+/HER2- invasive breast carcinoma. In tamoxifen treated and control matched pairs, a total of 22,195 cells from normal breast and 94,558 cells from tumor samples were sequenced. Computational analysis using consensus clustering was performed and cell types assigned using canonical correlation. Both tumor and normal samples identified clustering by cell type and not by patient revealing significant variability in cell type abundance between samples. In the normal breast samples, we performed differentially expressed genes (DEG) analysis comparing tamoxifen treatment to control for each cell type (Immune cells, fibroblasts, basal epithelial cells, luminal progenitor cells, and mature luminal cells) and enrichment analysis of up- and down-regulated genes performed. Strong depletion of estrogen induced genes was observed in tamoxifen-treated normal luminal progenitor and mature luminal cells, but not in basal epithelial cells or fibroblasts, demonstrating distinct, subpopulation-specific response to tamoxifen. In the 10 tumor matched pairs, 4 had a high epithelial proportion and tumor cells identified using inferred copy number variation. Tumor cells in 3 of these 4 samples showed significant down regulation of estrogen response genes with tamoxifen treatment. Using scBCSubtype to assign PAM50 subtype to individual tumor cells, the 3 responsive tumors were comprised primarily of LumA cells while the unresponsive tumor was predominantly LumB. Finally, we developed a novel score to quantify responsiveness at the single cell level based on downregulation of estrogen response genes with tamoxifen treatment relative to matched cluster-specific untreated expression. This analysis demonstrated heterogeneity in response to tamoxifen in tumor cells and identified distinct subpopulations of responsive and unresponsive tumor cells to tamoxifen treatment. Conclusion: We developed a novel ex vivo model to determine heterogeneity in therapeutic response to tamoxifen in normal human breast tissue and primary human breast tumors. We demonstrate differences in tamoxifen response by cell type and identify distinctly responsive and resistant subpopulations within human tumors. This provides a foundation to define features of responsive and resistant populations on the individual cell and specimen basis, and should allow us to develop precise, single cell-based predictors of response to endocrine therapy, and to identify genes and pathways driving resistance to therapy. Citation Format: Hyunsoo Kim, Austin Whitman, Kamila Wisniewska, Susana Garcia-Recio, Rasha Kakati, Hector L. Franco, Charles M. Perou, Philip Spanheimer. PD4-08 A Novel Single Cell Model of Tamoxifen Response in Primary Human Breast Tumors [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD4-08.
- Published
- 2023
- Full Text
- View/download PDF
5. Abstract P6-12-08: Identifying FOXA1 Binding Partners using Proximity Labeling
- Author
-
Rosemary N. Plagens, Shen Li, Christine A. Mills, Laura Herring, and Hector L. Franco
- Subjects
Cancer Research ,Oncology - Abstract
Approximately 75% of breast cancers are driven by the estrogen receptor alpha (ER), and despite the advent of endocrine therapy to block ER signaling pathways, a significant portion of women develop resistance to these drugs. The pioneer factor FOXA1 has been shown to facilitate nearly all DNA-binding events of ER in response to estrogen in ER+ breast cancer (ER+BC). Notably, up-regulation of FOXA1 is a hallmark of endocrine-resistant phenotypes and has been shown to reprogram enhancer elements, leading to an altered transcriptome. However, FOXA1 is a critical pioneer factor for multiple nuclear hormone receptors, aside from ER, and is implicated in regulation of important factors such as HER2 and the androgen receptor (AR). With the diverse array of breast cancer molecular subtypes displaying complex interplay between ER, HER2, AR, PR, and other hormone receptors, describing the complete ensemble of FOXA1 binding partners in various contexts, such as endocrine-resistant tumors, is of increasing importance. To define a comprehensive catalog of FOXA1 binding partners under basal conditions, we generated MCF-7 cell lines stably expressing constructs of FOXA1 fused at its N- or C-terminus to the biotin ligase miniTurbo. Using proximity labeling coupled with mass-spectrometry, we have comprehensively cataloged binding partners of FOXA1, including many expected proteins such as ER, AR, MLL3, YAP1, and GATA-3. Moreover, we have discovered more than 150 previously unidentified binding partners of FOXA1, which may exert profound effects on FOXA1 function. Importantly, high hazard ratios and significant dependencies are associated with several of these new binding partners, such as subunits of a previously described histone deacetylase (HDAC) complex containing genetic suppressor element 1 (GSE1) and lysine-specific histone demethylase 1A (KDM1A). Genomic approaches are currently underway to characterize where in the genome FOXA1 is interacting with these novel proteins and to guide future exploration into the physiological significance of these interactions. Integrating biochemical, molecular, and genomic approaches, we have potentially highlighted new mechanisms of FOXA1, which could have significant clinical impact in the future. Citation Format: Rosemary N. Plagens, Shen Li, Christine A. Mills, Laura Herring, Hector L. Franco. Identifying FOXA1 Binding Partners using Proximity Labeling [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P6-12-08.
- Published
- 2023
- Full Text
- View/download PDF
6. Abstract P5-14-02: Identifying oncogenic enhancer elements in TNBC of the Basal-like subtype using single-cell ATAC-seq and RNA-seq
- Author
-
Matthew J. Regner, Aatish Thennavan, Kamila Wisniewska, Susana Garcia-Recio, Raul Mendez-Giraldez, Philip Spanheimer, Charles M. Perou, and Hector L. Franco
- Subjects
Cancer Research ,Oncology - Abstract
Identification of the cis-regulatory elements controlling oncogenic transcriptional programs is critical to understanding tumor biology. To find cis-regulatory elements (i.e. gene enhancers) of oncogenic dependencies in Triple-Negative Breast Cancers (TNBC) of the Basal-like gene expression subtype, we generated matched single-cell transcriptome (scRNA-seq) and chromatin accessibility (scATAC-seq) profiles for two human Basal-like tumors and four normal mammary reduction samples. This unique dataset enabled us to correlate variations in chromatin structure with variations in gene expression revealing putative enhancers that are specifically active within cancer cells, but not within normal mammary ductal epithelial cells. We then leveraged the Cancer Dependency Map (DepMap) portal at the BROAD Institute to infer gene expression dependencies in breast cancer cell lines of the Basal-like molecular subtype. Putative cancer-specific enhancers were prioritized based on the transcriptional dependency of their target gene(s) in Basal-like cell lines as reported by the DepMap portal. Based on our preliminary analyses, we report several cancer-specific enhancers that drive the expression of important transcription factors such as EN1 and SOX4. These transcription factors are known to have profound effects on tumor biology, especially considering that high expression of EN1 is associated with brain metastasis and SOX4 is known to regulate immune evasion and PI3K/Akt signaling. Moreover, both of these transcription factors portend a worse outcome in TNBC patients. Thus, our analysis suggests that high levels of expression of these transcription factors is sustained specifically within the malignant cell types of these tumors, by the activity of these cancer-specific enhancers that are not typically active in normal epithelial cells. We are now performing CRISPR dCas9-KRAB experiments to epigenetically silence these cancer-specific enhancers and measure the consequences on expression of their predicted target genes. Additionally, we are investigating the trans-acting transcription factors that may physically bind to these enhancers to further regulate oncogenic transcription. By defining the regulatory logic of cancer cells at single-cell resolution, our work highlights the importance of cancer-specific and clinically relevant oncogenic regulatory elements in TNBC of the Basal-like subtype. Citation Format: Matthew J. Regner, Aatish Thennavan, Kamila Wisniewska, Susana Garcia-Recio, Raul Mendez-Giraldez, Philip Spanheimer, Charles M. Perou, Hector L. Franco. Identifying oncogenic enhancer elements in TNBC of the Basal-like subtype using single-cell ATAC-seq and RNA-seq [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P5-14-02.
- Published
- 2023
- Full Text
- View/download PDF
7. Abstract P6-12-03: Proinflammatory and estrogen signaling modulates the chemoresistance and metastasis of breast cancer cells through post-translational modifications of pioneering factor FOXA1
- Author
-
Shen Li, Hector L. Franco, Hyunsoo Kim, Rosemary N. Plagens, Raul Mendez-Giraldez, Colby Tubbs, Venkat Malladi, and Joseph Garay
- Subjects
Cancer Research ,Oncology - Abstract
ER-positive breast cancers compose most breast cancers at the time of diagnosis and are primarily driven by mitogenic estrogen signaling. In ER-positive breast cancers, the pioneer transcription factor FOXA1 plays a critical role in the estrogen receptor (ER) function. It binds to condensed chromatin and promotes chromatin accessibility for subsequent ER binding upon estrogen stimulation. We have reported that TNFa-stimulated proinflammatory signaling relocates FOXA1 to a new set of latent enhancers, which initiates the binding of estrogen liganded ER and subsequent expression of a unique transcriptome with clinical significance. The redistribution of FOXA1 occurs within 40 mins of the TNFa treatment, which implies a rapid signaling cascade that arises from changes to either FOXA1’s post-translational modifications (PTMs) or its binding partners. To understand this genomic redistribution of FOXA1, we compared the post-translational modifications (PTMs) of FOXA1 from Vehicle, E2, TNFa, and E2+TNFa treated MCF-7 breast cancer cells. More than five acetylation and phosphorylation events have been identified around the DNA binding domain of FOXA1 by semi-quantitative and quantitative mass spectrometry approaches, and their abundance varies across treatments. To study these PTMs of FOXA1, we used CRISPR/Cas9 to create specific knock-in mutations to mimic or prevent acetylation events in MCF-7 cells. Specifically, we engineered MCF-7 cell lines where K270 was mutated to glutamine (K270Q) to mimic acetylation. And for comparison, we also created cell lines where K270 was mutated to arginine (K270R) to prevent acetylation of FOXA1. Our data, including FOXA1 ChIP-seq and RNA-seq, revealed the genomic redistribution of FOXA1 with these PTMs, which subsequently alters gene expression programs and promotes cell growth, migration, or chemoresistance. These results were confirmed in other ER+ cell lines (such as T47D cells) providing evidence for the generalizability of our findings. Taken together, our data suggest that inflammatory signaling signaling can reshape the enhancer landscape of FOXA1 through post-translational modifications, resulting in changes to estrogen signaling that have profound effects on breast cancer biology. Citation Format: Shen Li, Hector L. Franco, Hyunsoo Kim, Rosemary N. Plagens, Raul Mendez-Giraldez, Colby Tubbs, Venkat Malladi, Joseph Garay. Proinflammatory and estrogen signaling modulates the chemoresistance and metastasis of breast cancer cells through post-translational modifications of pioneering factor FOXA1 [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P6-12-03.
- Published
- 2023
- Full Text
- View/download PDF
8. Tamoxifen Response at Single Cell Resolution in Estrogen Receptor-Positive Primary Human Breast Tumors
- Author
-
Hyunsoo Kim, Austin A. Whitman, Kamila Wisniewska, Rasha T. Kakati, Susana Garcia-Recio, Benjamin C. Calhoun, Hector L. Franco, Charles M. Perou, and Philip M. Spanheimer
- Abstract
In ER+/HER2- breast cancer, multiple measures of intra-tumor heterogeneity are associated with worse response to endocrine therapy. To investigate heterogeneity in response to treatment, we developed an operating room-to-laboratory pipeline for the collection of live human tumors and normal breast specimens immediately after surgical resection for processing into single-cell workflows for experimentation and genomic analyses. We demonstrate differences in tamoxifen response by cell type and identify distinctly responsive and resistant subpopulations within the malignant cell compartment of human tumors. Tamoxifen resistance signatures from 3 distinct resistant subpopulations are prognostic in large cohorts of ER+ breast cancer patients and enriched in endocrine therapy resistant tumors. This novel ex vivo model system now provides a foundation to define responsive and resistant sub-populations within heterogeneous tumors, to develop precise single cell-based predictors of response to therapy, and to identify genes and pathways driving resistance to therapy.
- Published
- 2023
- Full Text
- View/download PDF
9. Single-Cell Transcriptional and Epigenetic Profiles of Male Breast Cancer Nominate Salient Cancer-Specific Enhancers
- Author
-
Hyunsoo Kim, Kamila Wisniewska, Matthew J. Regner, Philip M. Spanheimer, and Hector L. Franco
- Abstract
Male breast cancer represents about 1% of all breast cancer diagnoses and, although there are some similarities between male and female breast cancer, the paucity of data available on male breast cancer makes it difficult to establish targeted therapies. To date, most male breast cancers (MBC) are treated according to protocols established for female breast cancer (FBC). Thus, defining the transcriptional and epigenetic landscape of MBC with improved resolution is critical for developing better avenues for therapeutic intervention. In this study, we present matched transcriptional (scRNA-seq) and epigenetic (scATAC-seq) profiles at single-cell resolution of two treatment naïve MBC tumors processed immediately after surgical resection. These data enable the detection of differentially expressed genes between male and female breast tumors across immune, stromal, and malignant cell types, to highlight several genes that may have therapeutic implications. Notably,MYCtarget genes andmTORC1signaling genes were significantly upregulated in the malignant cells of MBC compared to the female counterparts. To understand how the regulatory landscape of MBC give rise to these male-specific gene expression patterns, we leveraged the scATAC-seq data to systematically link changes in chromatin accessibility to changes in gene expression within each cell type. We observed cancer-specific rewiring of several salient enhancers and posit that these enhancers have a higher regulatory load than lineage specific enhancers. We highlight two examples of previously unannotated cancer-cell specific enhancers ofANXA2andPRDX4gene expression and show evidence for super-enhancer regulation ofLAMB3andCD47in male breast cancer cells. Overall, this dataset annotates clinically relevant regulatory networks in male breast tumors, providing a useful resource that expands our current understanding of the gene expression programs that underlie the biology of MBC.
- Published
- 2022
- Full Text
- View/download PDF
10. Abstract 5754: Determining the regulatory logic of breast cancer cells using single-cell multi-omics
- Author
-
Matthew J. Regner, Aatish Thennavan, Susana Garcia-Recio, Kamila Wisniewska, Philip M. Spanheimer, Joel S. Parker, Charles M. Perou, and Hector L. Franco
- Subjects
Cancer Research ,Oncology - Abstract
Cancer cells rewire regulatory elements scattered throughout the genome (such as enhancers) to drive aberrant gene expression. Thus, deconvoluting the regulatory mechanisms that contribute to oncogenic gene expression in cancer cells is key to understanding tumor biology. To this end, we have charted the transcriptional and epigenetic landscape of breast cancer at single-cell resolution to quantitatively link variation in chromatin accessibility to gene expression across malignant and non-malignant cell types. Our comprehensive dataset profiles the chromatin landscape (scATAC-seq) in concert with the transcriptional profiles (scRNA-seq) of 4 breast cancer cell lines, 12 primary breast tumors, and 4 normal mammary reduction tissue specimens collected and processed immediately after surgical resection. This dataset, encompassing over 250,000 individual cells, allowed us to define the regulatory logic of cancer cells by 1) revealing how the epigenome underlies cellular heterogeneity of these tumors in comparison to normal mammary tissue, 2) defining how malignant cells hijack enhancer elements to drive key transcriptional programs in a subtype-specific manner, and 3) annotating which cancer-specific enhancer-to-gene connections portend a worse outcome in patients. Notably, we discovered that cancer cells acquire de novo non-coding enhancer elements to modulate hallmark cancer pathways that were previously hidden using bulk genomics approaches. This highlights the potential for cancer-specific enhancers to serve as markers with diagnostic and prognostic potential, or even serve as tractable targets for therapeutic intervention. Together these data enable the annotation of the cellular composition, transcriptional, and epigenetic landscape of breast tumors to help pinpoint clinically relevant mechanisms of tumorigenesis. Citation Format: Matthew J. Regner, Aatish Thennavan, Susana Garcia-Recio, Kamila Wisniewska, Philip M. Spanheimer, Joel S. Parker, Charles M. Perou, Hector L. Franco. Determining the regulatory logic of breast cancer cells using single-cell multi-omics. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5754.
- Published
- 2023
- Full Text
- View/download PDF
11. Enhancer RNA Transcription Is Essential for a Novel CSF1 Enhancer in Triple-Negative Breast Cancer
- Author
-
Michael W. Lewis, Kamila Wisniewska, Caitlin M. King, Shen Li, Alisha Coffey, Michael R. Kelly, Matthew J. Regner, and Hector L. Franco
- Subjects
Cancer Research ,breast cancer ,ovarian cancer ,enhancer ,eRNA ,CRISPR-Cas9 ,dCas9-KRAB ,Cas13 ,gene expression ,Oncology - Abstract
Enhancers are critical regulatory elements in the genome that help orchestrate spatiotemporal patterns of gene expression during development and normal physiology. In cancer, enhancers are often rewired by various genetic and epigenetic mechanisms for the activation of oncogenes that lead to initiation and progression. A key feature of active enhancers is the production of non-coding RNA molecules called enhancer RNAs, whose functions remain unknown but can be used to specify active enhancers de novo. Using a combination of eRNA transcription and chromatin modifications, we have identified a novel enhancer located 30 kb upstream of Colony Stimulating Factor 1 (CSF1). Notably, CSF1 is implicated in the progression of breast cancer, is overexpressed in triple-negative breast cancer (TNBC) cell lines, and its enhancer is primarily active in TNBC patient tumors. Genomic deletion of the enhancer (via CRISPR/Cas9) enabled us to validate this regulatory element as a bona fide enhancer of CSF1 and subsequent cell-based assays revealed profound effects on cancer cell proliferation, colony formation, and migration. Epigenetic silencing of the enhancer via CRISPR-interference assays (dCas9-KRAB) coupled to RNA-sequencing, enabled unbiased identification of additional target genes, such as RSAD2, that are predictive of clinical outcome. Additionally, we repurposed the RNA-guided RNA-targeting CRISPR-Cas13 machinery to specifically degrade the eRNAs transcripts produced at this enhancer to determine the consequences on CSF1 mRNA expression, suggesting a post-transcriptional role for these non-coding transcripts. Finally, we test our eRNA-dependent model of CSF1 enhancer function and demonstrate that our results are extensible to other forms of cancer. Collectively, this work describes a novel enhancer that is active in the TNBC subtype, which is associated with cellular growth, and requires eRNA transcripts for proper enhancer function. These results demonstrate the significant impact of enhancers in cancer biology and highlight their potential as tractable targets for therapeutic intervention.
- Published
- 2022
- Full Text
- View/download PDF
12. Transcriptional control by enhancers and enhancer RNAs
- Author
-
Shen Li, Michael W. Lewis, and Hector L. Franco
- Subjects
Regulation of gene expression ,Super Enhancer ,0303 health sciences ,Enhancer RNA ,eRNA ,Enhancer RNAs ,Computational biology ,Review ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Super-enhancer ,Spatiotemporal gene expression ,Genetics ,Transcriptional regulation ,Gene Regulation ,Enhancer ,Gene ,Transcription factor ,030217 neurology & neurosurgery ,030304 developmental biology ,Biotechnology - Abstract
The regulation of gene expression is a fundamental cellular process and its misregulation is a key component of disease. Enhancers are one of the most salient regulatory elements in the genome and help orchestrate proper spatiotemporal gene expression during development, in homeostasis, and in response to signaling. Notably, molecular aberrations at enhancers, such as translocations and single nucleotide polymorphisms, are emerging as an important source of human variation and susceptibility to disease. Herein we discuss emerging paradigms addressing how genes are regulated by enhancers, common features of active enhancers, and how non-coding enhancer RNAs (eRNAs) can direct gene expression programs that underlie cellular phenotypes. We survey the current evidence, which suggests that eRNAs can bind to transcription factors, mediate enhancer-promoter interactions, influence RNA Pol II elongation, and act as decoys for repressive cofactors. Furthermore, we discuss current methodologies for the identification of eRNAs and novel approaches to elucidate their functions.
- Published
- 2019
13. A multi-omic dissection of super-enhancer driven oncogenic gene expression programs in ovarian cancer
- Author
-
Michael R. Kelly, Kamila Wisniewska, Matthew J. Regner, Michael W. Lewis, Andrea A. Perreault, Eric S. Davis, Douglas H. Phanstiel, Joel S. Parker, and Hector L. Franco
- Subjects
Ovarian Neoplasms ,Multidisciplinary ,Enhancer Elements, Genetic ,Carcinogenesis ,General Physics and Astronomy ,Gene Expression ,Humans ,Female ,General Chemistry ,Carcinoma, Ovarian Epithelial ,General Biochemistry, Genetics and Molecular Biology ,Chromatin - Abstract
The human genome contains regulatory elements, such as enhancers, that are often rewired by cancer cells for the activation of genes that promote tumorigenesis and resistance to therapy. This is especially true for cancers that have little or no known driver mutations within protein coding genes, such as ovarian cancer. Herein, we have utilized an integrated set of genomic and epigenomic datasets to identify clinically relevant super-enhancers that are preferentially amplified in ovarian cancer patients. We have systematically probed the top 86 super-enhancers, using CRISPR-interference and CRISPR-deletion assays coupled to RNA-sequencing, to nominate two salient super-enhancers that drive proliferation and migration of cancer cells. Utilizing Hi-C, we constructed chromatin interaction maps that enabled the annotation of direct target genes for these super-enhancers and later confirmed their activity specifically within the cancer cell compartment of human tumors using single-cell genomics data. Together, our multi-omic approach has examined a number of fundamental questions about how regulatory information encoded into super-enhancers drives gene expression networks that underlie the biology of ovarian cancer.
- Published
- 2021
14. CRISPR-based knockin mutagenesis of the pioneer transcription factor FOXA1; optimization of strategies for multi-allelic proteins in cancer cells
- Author
-
Shen Li, Colby A. Tubbs, Hector L. Franco, and Joseph P. Garay
- Subjects
Genome editing ,Cancer cell ,Mutagenesis (molecular biology technique) ,CRISPR ,Computational biology ,FOXA1 ,Biology ,Gene ,Genome ,Genome engineering - Abstract
Precise genome engineering of living cells has been revolutionized by the introduction of the highly specific and easily programmable properties of CRISPR-Cas9 technology. This has greatly accelerated research into human health and has facilitated the discovery of novel therapeutics. CRISPR-Cas9 is most widely employed for its ability to inactivate, or knockout, specific genes, but can be also used to introduce subtle site-specific substitutions of DNA sequences that can lead to changes in the amino acid composition of proteins. Despite the proven success of CRISPR-based knockin strategies of genes in typical diploid cells (i.e. cells containing two sets of chromosomes), precise editing of cancer cells, that typically have unstable genomes and multiple copies of chromosomes, is more challenging and not adequately addressed in the literature. Herein we detail our methodology for replacing endogenous proteins with intended knockin mutants in polyploid cancer cells and discuss our experimental design, screening strategy, and facile allele-frequency estimation methodology. As proof of principle, we performed genome editing of specific amino acids within the pioneer transcription factor FOXA1, a critical component of estrogen and androgen receptor signaling, in MCF-7 breast cancer cells. We confirm proper levels of mutant FOXA1 protein expression and intended amino acids substitutions via western blotting and mass spectrometry. In addition, we show that mutant allele-frequency estimation is easily achieved by TOPO cloning combined with allele-specific PCR, which we later confirmed by next-generation RNA-sequencing. Typically, there are 4 - 5 copies (alleles) of FOXA1 in breast cancer cells making the editing of this protein inherently challenging. As a result, most studies that focus on FOXA1 mutants rely on ectopic overexpression of FOXA1 from a plasmid. Therefore, we provide an optimized methodology for replacing endogenous wildtype FOXA1 with precise knockin mutants to enable the systematic analysis of its molecular mechanisms within the appropriate physiological context.
- Published
- 2020
- Full Text
- View/download PDF
15. Total Functional Score of Enhancer Elements Identifies Lineage-Specific Enhancers That Drive Differentiation of Pancreatic Cells
- Author
-
W. Lee Kraus, Hector L. Franco, Anusha Nagari, and Venkat S. Malladi
- Subjects
Regulation of gene expression ,Applied Mathematics ,Methods and Protocols ,tissue-specific transcription ,epigenome ,Computational biology ,Biology ,Biochemistry ,Embryonic stem cell ,Computer Science Applications ,Chromatin ,Computational Mathematics ,Histone ,lcsh:Biology (General) ,biology.protein ,pancreas ,Stem cell ,Enhancer ,gene regulation ,Molecular Biology ,Transcription factor ,lcsh:QH301-705.5 ,transcription factor ,Epigenomics - Abstract
The differentiation of embryonic stem cells into various lineages is highly dependent on the chromatin state of the genome and patterns of gene expression. To identify lineage-specific enhancers driving the differentiation of progenitors into pancreatic cells, we used a previously described computational framework called Total Functional Score of Enhancer Elements (TFSEE), which integrates multiple genomic assays that probe both transcriptional and epigenomic states. First, we evaluated and compared TFSEE as an enhancer-calling algorithm with enhancers called using GRO-seq-defined enhancer transcripts (method 1) versus enhancers called using histone modification ChIP-seq data (method 2). Second, we used TFSEE to define the enhancer landscape and identify transcription factors (TFs) that maintain the multipotency of a subpopulation of endodermal stem cells during differentiation into pancreatic lineages. Collectively, our results demonstrate that TFSEE is a robust enhancer-calling algorithm that can be used to perform multilayer genomic data integration to uncover cell type-specific TFs that control lineage-specific enhancers.
- Published
- 2020
16. A multi-omic single-cell landscape of human gynecologic malignancies
- Author
-
Susana García-Recio, Charles M. Perou, Kamila Wisniewska, Victoria L. Bae-Jump, Aatish Thennavan, Venkat S. Malladi, Gabrielle Hawkins, Matthew J. Regner, Joel S. Parker, Raul Mendez-Giraldez, and Hector L. Franco
- Subjects
Proteomics ,Epithelial-Mesenchymal Transition ,Carcinogenesis ,Gastrointestinal Stromal Tumors ,Kaplan-Meier Estimate ,Computational biology ,Biology ,medicine.disease_cause ,Article ,Transcriptome ,RNA, Small Cytoplasmic ,medicine ,Humans ,RNA-Seq ,Regulatory Elements, Transcriptional ,Molecular Biology ,Transcription factor ,Aged ,Gene Library ,Ovarian Neoplasms ,Regulation of gene expression ,Ovary ,Cancer ,Genomics ,Oncogenes ,Cell Biology ,Middle Aged ,medicine.disease ,Chromatin ,Enhancer Elements, Genetic ,Genetic Techniques ,Cancer cell ,Female ,Ovarian cancer ,Transcription Factors - Abstract
Deconvolution of regulatory mechanisms that drive transcriptional programs in cancer cells is key to understanding tumor biology. Herein, we present matched transcriptome (scRNA-seq) and chromatin accessibility (scATAC-seq) profiles at single-cell resolution from human ovarian and endometrial tumors processed immediately following surgical resection. This dataset reveals the complex cellular heterogeneity of these tumors and enabled us to quantitatively link variation in chromatin accessibility to gene expression. We show that malignant cells acquire previously unannotated regulatory elements to drive hallmark cancer pathways. Moreover, malignant cells from within the same patients show substantial variation in chromatin accessibility linked to transcriptional output, highlighting the importance of intratumoral heterogeneity. Finally, we infer the malignant cell type-specific activity of transcription factors. By defining the regulatory logic of cancer cells, this work reveals an important reliance on oncogenic regulatory elements and highlights the ability of matched scRNA-seq/scATAC-seq to uncover clinically relevant mechanisms of tumorigenesis in gynecologic cancers.
- Published
- 2021
- Full Text
- View/download PDF
17. Reciprocal Feedback Between miR-181a and E2/ERα in Myometrium Enhances Inflammation Leading to Labor
- Author
-
Wei Na Liu, Gang Wang, Carole R. Mendelson, Holly E Kinser, Lu Gao, and Hector L. Franco
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Myocytes, Smooth Muscle ,Clinical Biochemistry ,Cell Culture Techniques ,Estrogen receptor ,Inflammation ,Context (language use) ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,Endocrinology ,Downregulation and upregulation ,Pregnancy ,Internal medicine ,microRNA ,medicine ,Animals ,Humans ,reproductive and urinary physiology ,Mice, Inbred ICR ,Labor, Obstetric ,Estradiol ,Biochemistry (medical) ,Estrogen Receptor alpha ,Myometrium ,Original Articles ,Up-Regulation ,MicroRNAs ,Interleukin 10 ,030104 developmental biology ,Cytokine ,Cytokines ,Female ,medicine.symptom ,Signal Transduction - Abstract
Context: The initiation of term and preterm labor is associated with an up-regulated inflammatory response in myometrium; however, the underlying signaling pathways remain incompletely defined. Objective: To define the regulatory mechanisms that mediate the increased myometrial inflammatory response leading to labor, we investigated the roles of microRNAs (miRNA/miR). Design and Setting: Human myometrial tissues, isolated smooth muscle cells, and animal models were used to study miR-181a regulation of uterine inflammatory pathways and contractility. Patients: Myometrial tissues from 15 term pregnant women undergoing elective cesarean section (not in labor) and 10 term pregnant women undergoing emergency cesarean section (in labor) were used. Results: Expression of the highly conserved microRNA, miR-181a, was significantly decreased in mouse and human myometrium during late gestation. By contrast, the putative miR-181a targets, TNF-α, and estrogen receptor (ER)-α, and the validated target, c-Fos, key factors in the inflammatory response leading to parturition, were coordinately up-regulated. In studies using human myometrial cells, overexpression of miR-181a mimics repressed basal as well as IL-1β-induced TNF-α, C-C motif chemokine ligand 2 and 8 expression, whereas the expression of the antiinflammatory cytokine, IL-10, was increased. Overexpression of miR-181a dramatically inhibited both spontaneous and IL-1β-induced contraction of human myometrial cells. Notably, miR-181a directly targeted ERα and decreased its expression, whereas estradiol-17β reciprocally inhibited expression of mature miR-181a in myometrial cells. Conclusions: Thus, increased estradiol-17β/ERα signaling in myometrium near term inhibits miR-181a, resulting in a further increase in ERα and proinflammatory signaling. This escalating feedback loop provides novel targets and therapeutic strategies for the prevention of preterm labor and its consequences.
- Published
- 2016
- Full Text
- View/download PDF
18. TNFα Signaling Exposes Latent Estrogen Receptor Binding Sites to Alter the Breast Cancer Cell Transcriptome
- Author
-
W. Lee Kraus, Hector L. Franco, and Anusha Nagari
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Breast Neoplasms ,Biology ,Article ,Transcriptome ,Humans ,RNA, Small Interfering ,Enhancer ,Molecular Biology ,Cell Proliferation ,Regulation of gene expression ,Binding Sites ,Estradiol ,Estrogen receptor binding ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Estrogen Receptor alpha ,NF-kappa B ,Cell Biology ,Molecular biology ,Survival Analysis ,Cell biology ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,MCF-7 Cells ,Female ,FOXA1 ,Signal transduction ,Estrogen receptor alpha ,Protein Binding ,Signal Transduction - Abstract
The interplay between mitogenic and proinflammatory signaling pathways plays key roles in determining the phenotypes and clinical outcomes of breast cancers. Using GRO-seq in MCF-7 cells, we defined the immediate transcriptional effects of crosstalk between estradiol (E2) and TNFα, identifying a large set of target genes whose expression is rapidly altered with combined E2 + TNFα treatment, but not with either agent alone. The pleiotropic effects on gene transcription in response to E2 + TNFα are orchestrated by extensive remodeling of the ERα enhancer landscape in an NF-κB- and FoxA1-dependent manner. In addition, expression of the de novo and synergistically regulated genes is strongly associated with clinical outcomes in breast cancers. Together, our genomic and molecular analyses indicate that TNFα signaling, acting in pathways culminating in the redistribution of NF-κB and FoxA1 binding sites across the genome, creates latent ERα binding sites that underlie altered patterns of gene expression and clinically relevant cellular responses.
- Published
- 2015
- Full Text
- View/download PDF
19. Nuclear receptors, transcriptional enhancers, and gene regulation
- Author
-
W. Lee Kraus, Hector L. Franco, Venkat S. Malladi, Anusha Nagari, Shino Murakami, Tulip Nandu, Ziying Liu, Tim Y. Hou, and Yasmin M. Vasquez
- Subjects
Regulation of gene expression ,Nuclear receptor ,Biology ,Enhancer ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
20. Enhancer transcription reveals subtype-specific gene expression programs controlling breast cancer pathogenesis
- Author
-
Kaori Tanaka, Venkat S. Malladi, Xiaobing Shi, Hector L. Franco, Kendra Allton, Sharon Y.R. Dent, Yuanxin Xi, W. Lee Kraus, Jing Li, Anusha Nagari, Shino Murakami, Mark T. Bedford, Dana Richardson, Wenqian Li, Michelle Craig Barton, Khandan Keyomarsi, and Wei Li
- Subjects
0301 basic medicine ,Adult ,Carcinogenesis ,Cell Survival ,Triple Negative Breast Neoplasms ,Biology ,Histones ,03 medical and health sciences ,Breast cancer ,Transcription (biology) ,Cell Line, Tumor ,Gene expression ,Genetics ,medicine ,Humans ,Gene Regulatory Networks ,RNA, Messenger ,Enhancer ,Transcription factor ,Genetics (clinical) ,Triple-negative breast cancer ,Cell Proliferation ,Regulation of gene expression ,Research ,FOSL1 ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Enhancer Elements, Genetic ,Cancer research ,Female ,Transcriptome ,Transcription Factors - Abstract
Noncoding transcription is a defining feature of active enhancers, linking transcription factor (TF) binding to the molecular mechanisms controlling gene expression. To determine the relationship between enhancer activity and biological outcomes in breast cancers, we profiled the transcriptomes (using GRO-seq and RNA-seq) and epigenomes (using ChIP-seq) of 11 different human breast cancer cell lines representing five major molecular subtypes of breast cancer, as well as two immortalized (“normal”) human breast cell lines. In addition, we developed a robust and unbiased computational pipeline that simultaneously identifies putative subtype-specific enhancers and their cognate TFs by integrating the magnitude of enhancer transcription, TF mRNA expression levels, TF motif P-values, and enrichment of H3K4me1 and H3K27ac. When applied across the 13 different cell lines noted above, the Total Functional Score of Enhancer Elements (TFSEE) identified key breast cancer subtype-specific TFs that act at transcribed enhancers to dictate gene expression patterns determining growth outcomes, including Forkhead TFs, FOSL1, and PLAG1. FOSL1, a Fos family TF, (1) is highly enriched at the enhancers of triple negative breast cancer (TNBC) cells, (2) acts as a key regulator of the proliferation and viability of TNBC cells, but not Luminal A cells, and (3) is associated with a poor prognosis in TNBC breast cancer patients. Taken together, our results validate our enhancer identification pipeline and reveal that enhancers transcribed in breast cancer cells direct critical gene regulatory networks that promote pathogenesis.
- Published
- 2017
21. Abstract 4496: Cytokine-induced post-translational modifications of FOXA1 affect enhancer selection and estrogen signaling in breast cancer cells
- Author
-
Shen Li, Raul Mendez-Giraldez, Joseph P. Garay, Kamila Wisniewska, Colby A. Tubbs, Charles M. Perou, and Hector L. Franco
- Subjects
Cancer Research ,Oncology - Abstract
The pioneer transcription factor FOXA1 is a critical determinant for estrogen receptor (ER) function in hormone-dependent breast cancers. Upon estrogen stimulation, liganded ER binds to poised enhancer regions across the genome that are demarcated by FOXA1 and histone modifications such as H3K4me1 and H3K27ac. In a recent publication, we show that proinflammatory signaling, caused by the cytokine TNFa, drives FOXA1 to latent enhancer binding sites to promote chromatin accessibility for subsequent ER binding upon estrogen ligation. These latent enhancers, activated by the combined treatment of estrogen and TNFa, induced the expression of a unique transcriptome with clinical significance. The effects of TNFa treatment on FOXA1 chromatin redistribution and subsequent gene expression occur within 40 minutes, which points to a rapid signaling cascade that culminates in either changes in FOXA1's posttranslational modifications (PTMs) or its binding partners. To understand how proinflammatory TNFa signaling can redirect FOXA1 to new sites across the genome, we started by characterizing the posttranslational modifications (PTMs) of FOXA1. We immunoprecipitated FOXA1 from MCF-7 breast cancer cells that were treated by E2, TNFa or E2+ TNFa, and then examined their posttranslational status using semi-quantitative and quantitative mass spectrometry approaches. Several phosphorylation sites and acetylation sites have been identified near the DNA binding domain of FOXA1, and acetylation of lysine 295 (K295) was found specifically enriched in TNFa treatment. To test if acetylation of FOXA1 at K295 changes its binding preference and genomic distribution, we used the programmable properties of CRISPR/Cas9 to create specific knockin mutations to mimic or prevent acetylation of K295 in MCF-7 cells. More specifically, we mutated K295 to glutamine (K295Q) to mimic acetylation and essentially “lock” FOXA1 into a permanently acetylated state and, for comparison, we created another cell line where K295 was mutated to arginine (K295R) to prevent acetylation of FOXA1. Our preliminary data shows changes in the genomic redistribution of FOXA1 in the knock-in cell lines resulting in altered gene expression programs. These data suggest that inflammation-based acetylation of FOXA1 can affect estrogen signaling pathways in breast cancer cells by altering the enhancer landscape of FOXA1 and consequently the estrogen receptor. Supported by a grant from the NIH/NCI (R00 CA204628) to H.L.F Citation Format: Shen Li, Raul Mendez-Giraldez, Joseph P. Garay, Kamila Wisniewska, Colby A. Tubbs, Charles M. Perou, Hector L. Franco. Cytokine-induced post-translational modifications of FOXA1 affect enhancer selection and estrogen signaling in breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4496.
- Published
- 2019
- Full Text
- View/download PDF
22. Interplay between inflammatory and estrogen signaling in breast cancer
- Author
-
Hector L. Franco and Anusha Nagari
- Subjects
medicine.drug_class ,Immunology ,Estrogen receptor ,Breast Neoplasms ,Context (language use) ,Biology ,medicine.disease_cause ,Biochemistry ,Proinflammatory cytokine ,Pathogenesis ,Breast cancer ,medicine ,Humans ,Immunology and Allergy ,Molecular Biology ,Inflammation ,Estrogens ,Hematology ,medicine.disease ,Estrogen ,Cancer research ,Female ,FOXA1 ,Carcinogenesis ,Signal Transduction - Abstract
Inflammation is known to have a paradoxical effect in cancers, in some cases promoting pathogenesis while in others inhibiting pathogenesis, depending on the cellular context. In an effort to answer a number of fundamental questions about two of the major signaling cascades that affect breast tumorigenesis and impact clinical outcome, we examined the genome-wide consequences of treating ERα-positive breast cancer cells with both estrogen and TNFα. Below, we highlight our observations, their biological significance, and how they provide a framework for understanding the molecular basis for integration of proinflammatory and estrogen signaling in breast cancer.
- Published
- 2015
- Full Text
- View/download PDF
23. No Driver behind the Wheel? Targeting Transcription in Cancer
- Author
-
W. Lee Kraus and Hector L. Franco
- Subjects
Regulation of gene expression ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Kinase ,Triple Negative Breast Neoplasms ,General Biochemistry, Genetics and Molecular Biology ,Cyclin-dependent kinase ,Behind the wheel ,Transcription (biology) ,Immunology ,Cancer cell ,biology.protein ,Cancer research ,Cyclin-dependent kinase 7 - Abstract
Exploiting the dependence of cancer cells on transcription can be used as an effective strategy for targeting aggressive and therapeutically recalcitrant tumors. Wang et al. show that inhibiting transcription using THZ1, a small-molecule inhibitor of cyclin-dependent kinase CDK7, induces apoptotic cell death in triple-negative breast cancers.
- Published
- 2015
- Full Text
- View/download PDF
24. Nonsense mutations of the bHLH transcription factor TWIST2 found in Setleis Syndrome patients cause dysregulation of periostin
- Author
-
Yongchao Ge, Ruth G. Leon, Robert Friesel, Hector L. Franco, Carmen L. Cadilla, Jose J Casasnovas, and Robert J. Desnick
- Subjects
Transcriptional Activation ,Mutant ,Nonsense mutation ,Biology ,Periostin ,Skin Diseases ,Biochemistry ,Article ,Twist transcription factor ,Transactivation ,Ectodermal Dysplasia ,medicine ,Humans ,Transcription factor ,Cells, Cultured ,Setleis syndrome ,Helix-Loop-Helix Motifs ,Twist-Related Protein 1 ,Focal Facial Dermal Dysplasias ,Cell Biology ,Fibroblasts ,medicine.disease ,Molecular biology ,Focal Dermal Hypoplasia ,Repressor Proteins ,Codon, Nonsense ,Protein Multimerization ,Cell Adhesion Molecules ,Chromatin immunoprecipitation - Abstract
Setleis Syndrome (OMIM ID: 227260) is a rare autosomal recessive disease characterized by abnormal facial development. Recently, we have reported that two nonsense mutations (c.486C > T [Q119X] and c.324C > T [Q65X]) of the basic helix-loop-helix (bHLH) transcription factor TWIST2 cause Setleis Syndrome. Here we show that periostin, a cell adhesion protein involved in connective tissue development and maintenance, is down-regulated in Setleis Syndrome patient fibroblast cells and that periostin positively responds to manipulations in TWIST2 levels, suggesting that TWIST2 is a transactivator of periostin. Functional analysis of the TWIST2 mutant form (Q119X) revealed that it maintains the ability to localize to the nucleus, forms homo and heterodimers with the ubiquitous bHLH protein E12, and binds to dsDNA. Reporter gene assays using deletion constructs of the human periostin promoter also reveal that TWIST2 can activate this gene more specifically than Twist1, while the Q119X mutant results in no significant transactivation. Chromatin immunoprecipitation assays show that both wild-type TWIST2 and the Q119X mutant bind the periostin promoter, however only wild-type TWIST2 is associated with higher levels of histone acetylation across the 5′-regulatory region of periostin. Taken together, these data suggest that the C-terminal domain of TWIST2, which is missing in the Q119X mutant form of TWIST2, is responsible for proper transactivation of the periostin gene. Improper regulation of periostin by the mutant form of TWIST2 could help explain some of the soft tissue abnormalities seen in these patients therefore providing a genotype–phenotype relationship for Setleis Syndrome.
- Published
- 2011
- Full Text
- View/download PDF
25. Redundant or separate entities?--roles of Twist1 and Twist2 as molecular switches during gene transcription
- Author
-
Carmen L. Cadilla, Jose Casasnovas, Hector L. Franco, and José R. Rodríguez-Medina
- Subjects
Transcriptional Activation ,animal structures ,Transcription, Genetic ,Repressor ,Biology ,Muscle Development ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,Genetics ,Transcriptional regulation ,Animals ,Survey and Summary ,Gene ,Transcription factor ,Cellular localization ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Activator (genetics) ,Twist-Related Protein 1 ,Nuclear Proteins ,Cell biology ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,E-Box Elements ,Gene Expression Regulation ,Immune System ,030220 oncology & carcinogenesis - Abstract
Twist1 and Twist2 are highly conserved members of the Twist subfamily of bHLH proteins responsible for the transcriptional regulation of the developmental programs in mesenchymal cell lineages. The regulation of such processes requires that Twist1 and Twist2 function as molecular switches to activate and repress target genes by employing several direct and indirect mechanisms. Modes of action by these proteins include direct DNA binding to conserved E-box sequences and recruitment of coactivators or repressors, sequestration of E-protein modulators, and interruption of proper activator/repressor function through protein–protein interactions. Regulatory outcomes of Twist1 and Twist2 are themselves controlled by spatial-temporal expression, phosphoregulation, dimer choice and cellular localization. Although these two proteins are highly conserved and exhibit similar functions in vitro, emerging literature have demonstrated different roles in vivo. The involvement of Twist1 and Twist2 in a broad spectrum of regulatory pathways highlights the importance of understanding their roles in normal development, homeostasis and disease. Here we focus on the mechanistic models of transcriptional regulation and summarize the similarities and differences between Twist1 and Twist2 in the context of myogenesis, osteogenesis, immune system development and cancer.
- Published
- 2010
- Full Text
- View/download PDF
26. Erythema, Atrophy, and Scarring on the Face and Arm of a Young Girl
- Author
-
D.A.B.D. Hector L. Franco M.D. and B S Hongfei Fang
- Subjects
medicine.medical_specialty ,Erythema ,business.industry ,media_common.quotation_subject ,Dermatology ,medicine.disease ,Surgery ,Diagnosis, Differential ,Cicatrix ,Atrophy ,Panniculitis, Lupus Erythematosus ,Pediatrics, Perinatology and Child Health ,medicine ,Humans ,Female ,Girl ,Enzyme Inhibitors ,medicine.symptom ,Child ,business ,Hydroxychloroquine ,media_common - Published
- 2015
- Full Text
- View/download PDF
27. Mechanism of gene regulation employed by the bHLH transcription factor TWIST2: Repression of the CHRDL1 gene
- Author
-
Jose Casasnovas, Carmen L. Cadilla, and Hector L. Franco
- Subjects
Regulation of gene expression ,Mechanism (biology) ,Genetics ,Biology ,Molecular Biology ,Biochemistry ,Gene ,Transcription factor ,Psychological repression ,Biotechnology ,Cell biology - Published
- 2013
- Full Text
- View/download PDF
28. Role of the TWIST proteins and ADD1 in the regulation of the CHRDL1 gene
- Author
-
Jose Casasnovas, Carmen L. Cadilla, and Hector L. Franco
- Subjects
ADD1 ,Genetics ,Twist ,Biology ,Molecular Biology ,Biochemistry ,Gene ,Biotechnology ,Cell biology - Published
- 2012
- Full Text
- View/download PDF
29. Homozygous Nonsense Mutations in TWIST2 Cause Setleis Syndrome
- Author
-
Carmen L. Cadilla, Robert J. Desnick, Lihadh Al-Gazali, Turgut Tukel, Dražen Šošić, James A. Richardson, Monica Erazo, Hector L. Franco, Jose J Casasnovas, and Eric N. Olson
- Subjects
Male ,Candidate gene ,Nonsense mutation ,Molecular Sequence Data ,United Arab Emirates ,Locus (genetics) ,Biology ,03 medical and health sciences ,Twist transcription factor ,Mice ,Report ,Genetics ,medicine ,Animals ,Humans ,Genetics(clinical) ,Abnormalities, Multiple ,Amino Acid Sequence ,Gene ,Genetics (clinical) ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Setleis syndrome ,Base Sequence ,030305 genetics & heredity ,Haplotype ,Homozygote ,Puerto Rico ,Twist-Related Protein 1 ,Chromosome Mapping ,Facies ,Nuclear Proteins ,Syndrome ,medicine.disease ,Disease gene identification ,Molecular biology ,Pedigree ,Repressor Proteins ,Phenotype ,Codon, Nonsense ,Child, Preschool ,Female ,Chromosomes, Human, Pair 3 ,Sequence Alignment - Abstract
The focal facial dermal dysplasias (FFDDs) are a group of inherited developmental disorders in which the characteristic diagnostic feature is bitemporal scar-like lesions that resemble forceps marks. To date, the genetic defects underlying these ectodermal dysplasias have not been determined. To identify the gene defect causing autosomal-recessive Setleis syndrome (type III FFDD), homozygosity mapping was performed with genomic DNAs from five affected individuals and 26 members of the consanguineous Puerto Rican (PR) family originally described by Setleis and colleagues. Microsatellites D2S1397 and D2S2968 were homozygous in all affected individuals, mapping the disease locus to 2q37.3. Haplotype analyses of additional markers in the PR family and a consanguineous Arab family further limited the disease locus to approximately 3 Mb between D2S2949 and D2S2253. Of the 29 candidate genes in this region, the bHLH transcription factor, TWIST2, was initially sequenced on the basis of its known involvement in murine facial development. Homozygous TWIST2 nonsense mutations, c.324CT and c.486CT, were identified in the affected members of the Arab and PR families, respectively. Characterization of the expressed mutant proteins, p.Q65X and p.Q119X, by electrophoretic mobility shift assays and immunoblot analyses indicated that they were truncated and unstable. Notably, Setleis syndrome patients and Twist2 knockout mice have similar facial features, indicating the gene's conserved role in mammalian development. Although human TWIST2 and TWIST1 encode highly homologous bHLH transcription factors, the finding that TWIST2 recessive mutations cause an FFDD and dominant TWIST1 mutations cause Saethre-Chotzen craniocynostosis suggests that they function independently in skin and bone development.
- Published
- 2010
30. Periostin gene expression is regulated by the bHLH transcription factor TWIST2 in human skin fibroblasts
- Author
-
Hector L. Franco, Jose J Casasnovas, and Carmen L. Cadilla
- Subjects
Periostin Gene ,Genetics ,Human skin ,Biology ,Molecular Biology ,Biochemistry ,Transcription factor ,Biotechnology ,Cell biology - Published
- 2009
- Full Text
- View/download PDF
31. The bHLH protein Twist2 regulates expression of the CHRDL1 (Chordin‐like 1) gene in human skin fibroblasts
- Author
-
Carmen L. Cadilla, Jose J Casasnovas, and Hector L. Franco
- Subjects
Genetics ,Human skin ,Biology ,Chordin ,Molecular Biology ,Biochemistry ,Gene ,Biotechnology ,Cell biology - Published
- 2009
- Full Text
- View/download PDF
32. Abstract PR06: Analysis of enhancer transcription reveals novel gene regulatory networks in breast cancer
- Author
-
Khandan Keyomarsi, Sharon Y.R. Dent, W. Lee Kraus, Dana Richardson, Yuanxin Xi, Wenqian Li, Anusha Nagari, Kaori Tanaka, Michelle Craig Barton, Hector L. Franco, Wei Li, Mark T. Bedford, Xiaobing Shi, and Jing Li
- Subjects
Genetics ,Cancer Research ,Oncology ,Sp3 transcription factor ,Transcription (biology) ,Response element ,E-box ,Enhancer RNAs ,Promoter ,Biology ,Enhancer ,Transcription factor - Abstract
Enhancer transcription is a defining feature of active enhancers. We and others have shown that enhancers that produce transcripts (so called “eRNAs”) are more likely to (1) be associated with active chromatin marks, such as H3K4me1 and K3K27ac, (2) loop to target gene promoters, and (3) be associated with target gene activation. Thus, enhancer transcription is a good predictor of active enhancers. In this regard, we have shown that enhancer transcription can be used in the absence of any other genomic information to predict enhancers. We have used Global Run-On coupled deep sequencing (GRO-seq) in 14 different breast cancer cell lines representing the five distinct molecular subtypes of breast cancer, coupled with a computational pipeline that we have developed, to predict enhancers based solely on enhancer transcription. We found both common and unique sites of enhancer transcription across the cell lines. In addition, we observed that enhancer transcription correlates with nearest gene transcription. Unsupervised hierarchical clustering of enhancer transcription was sufficient to segregate the breast cancer cell lines into their specific molecular subtypes. Transcription factor motif analysis performed at the sites of enhancer transcription identified transcription factors that may be important for the transcriptional programs of each cell type. Transcription factors whose motifs were uniquely enriched in a specific cell type were observed to be bound at the enhancers using locus-specific ChIP-qPCR assay. siRNA-mediated knockdown of the cognate transcription factors reduced enhancer transcription and cell proliferation in those cell types. The GRO-seq data were integrated with ChIP-seq data for several histone modifications typically enriched at promoters, gene bodies, enhancers, and repressive regions of the genome. The results from these analyses provide additional support for our enhancer identification pipeline. Taken together, our analyses have revealed novel gene regulatory networks that underlie breast cancer subtype-specific biology. This work was supported by a postdoctoral fellowship from the American Cancer Society - Lee National Denim Day Fellowship to H.L.F., grants from the NIH/NIDDK and CPRIT to W.L.K. and a grant from CPRIT to the LONESTAR Consortium. Citation Format: Hector L. Franco, Anusha Nagari, Yuanxin Xi, Wenqian Li, Dana Richardson, Kaori Tanaka, Jing Li, The CPRIT LONESTAR Consortium, Michelle C. Barton, Xiaobing Shi, Khandan Keyomarsi, Mark T. Bedford, Wei Li, Sharon Y.R. Dent, W. Lee Kraus. Analysis of enhancer transcription reveals novel gene regulatory networks in breast cancer. [abstract]. In: Proceedings of the AACR Special Conference on Chromatin and Epigenetics in Cancer; Sep 24-27, 2015; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2016;76(2 Suppl):Abstract nr PR06.
- Published
- 2016
- Full Text
- View/download PDF
33. Abstract 1815: Genome-wide analysis of the molecular crosstalk between mitogenic and proinflammatory signaling in breast cancer
- Author
-
W. Lee Kraus, Anusha Nagari, and Hector L. Franco
- Subjects
Genetics ,Transcriptome ,Cancer Research ,Crosstalk (biology) ,Oncology ,Signal transduction ,Biology ,Gene ,Chromatin immunoprecipitation ,Estrogen receptor alpha ,Transcription factor ,Proinflammatory cytokine ,Cell biology - Abstract
The interplay between mitogenic (e.g., estradiol) and proinflammatory (e.g., TNFα) signaling pathways play key roles in determining the phenotypes and clinical outcomes of breast cancers. Both pathways promote dramatic changes in the transcriptomes of breast cancer cells, as well as downstream cellular effects, but the interplay between these two pathways at the transcriptional level has not yet been analyzed in a comprehensive way across the genome. We have used global nuclear run-on coupled with deep sequencing (GRO-seq), a genomic method that maps the location and orientation of all actively engaged RNA polymerases, to characterize the immediate transcriptional responses of MCF-7 breast cancer cells treated for short durations (40 min.) with either estradiol (E2), TNFα, or both. In addition, we have integrated these data with chromatin immunoprecipitation coupled with deep sequencing (ChIP-seq) for both estrogen receptor alpha (ERα), which mediates E2 signaling, and the p65 subunit of the NF-κB transcription factor, which mediates TNFα signaling. Our results indicate extensive transcriptional interplay between these two signaling pathways, which is observed for a number of classical mitogenic and proinflammatory protein-coding genes. In addition, GRO-seq has allowed us to capture the transcriptional crosstalk at genomic locations encoding for long non-coding RNAs (lncRNAs), a poorly characterized class of RNAs which have been shown to play important roles in cancer outcomes. The synergistic and antagonistic interplay between E2 and TNFα also manifests itself at the level of ERα and p65 binding, which relocalize to new binding sites that are not occupied by either treatment alone. Taken together, our results identify rapid, signal-dependent transcriptional changes that occur during proliferative signaling versus inflammatory signaling and show that these effects are more extensive than previously anticipated. Uncovering the transcriptional landscape of these two important pathways outside of the protein coding regions of the genome may lead to new ways of diagnosing and treating breast cancer. Citation Format: Hector L. Franco, Anusha Nagari, W. Lee Kraus. Genome-wide analysis of the molecular crosstalk between mitogenic and proinflammatory signaling in breast cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1815. doi:10.1158/1538-7445.AM2013-1815
- Published
- 2013
- Full Text
- View/download PDF
34. Steroid Rosacea in Children
- Author
-
Hector L. Franco and William L. Weston
- Subjects
medicine.medical_specialty ,business.industry ,Rosacea ,Pediatrics, Perinatology and Child Health ,Adult population ,Medicine ,business ,medicine.disease ,Dermatology ,Steroid rosacea ,Acne - Abstract
Steroid rosacea is a facial dermatitis clinically resembling acne rosacea. Fluorinated topical steroids have been implicated as the cause or precipitating factor in previous case reports mainly involving an adult population. Four cases of pediatric acne rosacea associated with the use of topical fluorinated glucocorticosteroids are described. The process worsened during the two weeks following steroid cessation. We recommend that fluorinated glucocorticosteroids should not be used on the face of infants and children.
- Published
- 1979
- Full Text
- View/download PDF
35. Autoantibodies directed against sicca syndrome antigens in the neonatal lupus syndrome
- Author
-
Carol L. Peebles, S. Lance Forstot, Hector L. Franco, Prapan Phanuphak, and William L. Weston
- Subjects
musculoskeletal diseases ,Adult ,Male ,Anti-nuclear antibody ,Dermatology ,Asymptomatic ,Infant, Newborn, Diseases ,Serology ,Lupus Erythematosus, Discoid ,Pregnancy ,Sicca syndrome ,medicine ,Rheumatoid factor ,Humans ,Neonatal lupus erythematosus ,Maternal-Fetal Exchange ,Autoantibodies ,business.industry ,Autoantibody ,Infant, Newborn ,Infant ,medicine.disease ,eye diseases ,stomatognathic diseases ,Sjogren's Syndrome ,Immunology ,Female ,medicine.symptom ,business ,Anti-SSA/Ro autoantibodies ,Follow-Up Studies - Abstract
Clinical and serologic studies on three infants who had the neonatal lupus syndrome and studies on their mothers revealed an association with antibodies to sicca syndrome antigens. From initial studies and a 2-year follow-up, there is evidence that indicates transplacental passage of autoantibodies directed against Sjogren's (sicca) syndrome-associated nuclear antigens from asymptomatic mothers to newborns who subsequently developed neonatal lupus. Besides the presence of antinuclear antibodies, the mothers of these infants also showed high rheumatoid factor titers, and two had evidence of mild decreasing tearing on ophthalmologic examination. On follow-up examination 2 to 3 years later, both infants and mothers lacked evidence of active disease, and only the mothers continued to demonstrate the serologic abnormalities seen initially. Based on our findings, we postulate newborns of mothers with serologic or clinical evidence of Sjogren's (sicca) syndrome may be at greater risk for developing neonatal lupus.
- Published
- 1981
36. Total Functional Score of Enhancer Elements Identifies Lineage-Specific Enhancers That Drive Differentiation of Pancreatic Cells
- Author
-
Venkat S. Malladi, Anusha Nagari, Hector L Franco, and W Lee Kraus
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The differentiation of embryonic stem cells into various lineages is highly dependent on the chromatin state of the genome and patterns of gene expression. To identify lineage-specific enhancers driving the differentiation of progenitors into pancreatic cells, we used a previously described computational framework called Total Functional Score of Enhancer Elements (TFSEE), which integrates multiple genomic assays that probe both transcriptional and epigenomic states. First, we evaluated and compared TFSEE as an enhancer-calling algorithm with enhancers called using GRO-seq-defined enhancer transcripts (method 1) versus enhancers called using histone modification ChIP-seq data (method 2). Second, we used TFSEE to define the enhancer landscape and identify transcription factors (TFs) that maintain the multipotency of a subpopulation of endodermal stem cells during differentiation into pancreatic lineages. Collectively, our results demonstrate that TFSEE is a robust enhancer-calling algorithm that can be used to perform multilayer genomic data integration to uncover cell type-specific TFs that control lineage-specific enhancers.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.