Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 41(5): 111581, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36323264

ABSTRACT

The dynamic regulation of ß-cell abundance is poorly understood. Since chromatin remodeling plays critical roles in liver regeneration, these mechanisms could be generally important for regeneration in other tissues. Here, we show that the ARID1A mammalian SWI/SNF complex subunit is a critical regulator of ß-cell regeneration. Arid1a is highly expressed in quiescent ß-cells but is physiologically suppressed when ß-cells proliferate during pregnancy or after pancreas resection. Whole-body Arid1a knockout mice are protected against streptozotocin-induced diabetes. Cell-type and temporally specific genetic dissection show that ß-cell-specific Arid1a deletion can potentiate ß-cell regeneration in multiple contexts. Transcriptomic and epigenomic profiling of mutant islets reveal increased neuregulin-ERBB-NR4A signaling. Chemical inhibition of ERBB or NR4A1 blocks increased regeneration associated with Arid1a loss. Mammalian SWI/SNF (mSWI/SNF) complex activity is a barrier to ß-cell regeneration in physiologic and disease states.


Subject(s)
Epidermal Growth Factor , Nuclear Proteins , Mice , Animals , Pregnancy , Female , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Chromatin Assembly and Disassembly , Signal Transduction , Liver Regeneration , Mammals/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/genetics
2.
Nat Cancer ; 1(9): 909-922, 2020 09.
Article in English | MEDLINE | ID: mdl-34386776

ABSTRACT

SWI/SNF chromatin remodelers play critical roles in development and cancer. The causal links between SWI/SNF complex disassembly and carcinogenesis are obscured by redundancy between paralogous components. Canonical cBAF-specific paralogs ARID1A and ARID1B are synthetic lethal in some contexts, but simultaneous mutations in both ARID1s are prevalent in cancer. To understand if and how cBAF abrogation causes cancer, we examined the physiologic and biochemical consequences of ARID1A/ARID1B loss. In double knockout liver and skin, aggressive carcinogenesis followed de-differentiation and hyperproliferation. In double mutant endometrial cancer, add-back of either induced senescence. Biochemically, residual cBAF subcomplexes resulting from loss of ARID1 scaffolding were unexpectedly found to disrupt polybromo containing pBAF function. 37 of 69 mutations in the conserved scaffolding domains of ARID1 proteins observed in human cancer caused complex disassembly, partially explaining their mutation spectra. ARID1-less, cBAF-less states promote carcinogenesis across tissues, and suggest caution against paralog-directed therapies for ARID1-mutant cancer.


Subject(s)
Carcinogenesis , DNA-Binding Proteins , Neoplasms , Transcription Factors , Carcinogenesis/genetics , Chromatin , DNA-Binding Proteins/genetics , Humans , Mutation , Neoplasms/genetics , Transcription Factors/genetics
3.
Hepatology ; 69(5): 1931-1945, 2019 05.
Article in English | MEDLINE | ID: mdl-30584660

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a rapidly growing cause of chronic liver damage, cirrhosis, and hepatocellular carcinoma. How fatty liver pathogenesis is subject to epigenetic regulation is unknown. We hypothesized that chromatin remodeling is important for the pathogenesis of fatty liver disease. AT-rich interactive domain-containing protein 1A (ARID1A), a DNA-binding component of the SWItch/sucrose nonfermentable adenosine triphosphate-dependent chromatin-remodeling complex, contributes to nucleosome repositioning and access by transcriptional regulators. Liver-specific deletion of Arid1a (Arid1a liver knockout [LKO]) caused the development of age-dependent fatty liver disease in mice. Transcriptome analysis revealed up-regulation of lipogenesis and down-regulation of fatty acid oxidation genes. As evidence of direct regulation, ARID1A demonstrated direct binding to the promoters of many of these differentially regulated genes. Additionally, Arid1a LKO mice were more susceptible to high-fat diet-induced liver steatosis and fibrosis. We deleted Pten in combination with Arid1a to synergistically drive fatty liver progression. Inhibition of lipogenesis using CAT-2003, a potent sterol regulatory element-binding protein inhibitor, mediated improvements in markers of fatty liver disease progression in this Arid1a/Pten double knockout model. Conclusion: ARID1A plays a role in the epigenetic regulation of hepatic lipid homeostasis, and its suppression contributes to fatty liver pathogenesis. Combined Arid1a and Pten deletion shows accelerated fatty liver disease progression and is a useful mouse model for studying therapeutic strategies for NASH.


Subject(s)
DNA-Binding Proteins/deficiency , Lipogenesis , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Transcription Factors/deficiency , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Fatty Acids/metabolism , Mice , Mice, Knockout , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
4.
Elife ; 72018 03 20.
Article in English | MEDLINE | ID: mdl-29555020

ABSTRACT

Recent advances in single-cell transcriptomics techniques have opened the door to the study of gene regulatory networks (GRNs) at the single-cell level. Here, we studied the GRNs controlling the emergence of hematopoietic stem and progenitor cells from mouse embryonic endothelium using a combination of single-cell transcriptome assays. We found that a heptad of transcription factors (Runx1, Gata2, Tal1, Fli1, Lyl1, Erg and Lmo2) is specifically co-expressed in an intermediate population expressing both endothelial and hematopoietic markers. Within the heptad, we identified two sets of factors of opposing functions: one (Erg/Fli1) promoting the endothelial cell fate, the other (Runx1/Gata2) promoting the hematopoietic fate. Surprisingly, our data suggest that even though Fli1 initially supports the endothelial cell fate, it acquires a pro-hematopoietic role when co-expressed with Runx1. This work demonstrates the power of single-cell RNA-sequencing for characterizing complex transcription factor dynamics.


Subject(s)
Gene Expression Profiling/methods , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Single-Cell Analysis/methods , Transcription Factors/genetics , Animals , Cluster Analysis , Core Binding Factor alpha Subunits/genetics , Endothelium/cytology , Endothelium/embryology , Endothelium/metabolism , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Protein c-fli-1/genetics
5.
Mol Genet Genomic Med ; 6(2): 230-248, 2018 03.
Article in English | MEDLINE | ID: mdl-29397575

ABSTRACT

BACKGROUND: The camptodactyly-arthropathy-coxa vara-pericarditis syndrome (CACP) is a rare autosomal recessive condition characterized by camptodactyly, noninflammatory arthropathy, coxa vara, and pericarditis. CACP is caused by mutations in the proteoglycan 4 (PRG4) gene, which encodes a lubricating glycoprotein present in the synovial fluid and at the surface of articular cartilage. METHODS: In the present study, we compared the clinical and molecular findings of CACP syndrome in 35 patients from 11 unrelated families. In 28 patients, whole exome sequencing was used to investigate genomic variations. RESULTS: We found that camptodactyly of hands was the first symptom presented by most patients. Swelling of wrists, knees, and elbows began before 4 years of age, while the age of joint involvement was variable. Patients reported an increased pain level after the age of 10, and severe hip involvement developed after 20 years old. All patients presented developmental coxa vara and seven patients (~22%) had pleural effusion, pericarditis, and/or ascites. We identified nine novel genomic alterations, including the first case of homozygous complete deletion of exon 1 in the PRG4 gene. CONCLUSION: With this study, we contribute to the catalog of CACP causing variants. We confirm that the skeletal component of this disease worsens with age, and presents the potential mechanisms for interfamily variability, by discussing the influence of a modifier gene and escape from nonsense-mediated mRNA decay. We believe that this report will increase awareness of this familial arthropathic condition and the characteristic clinical and radiological findings will facilitate the differentiation from the common childhood rheumatic diseases such as juvenile idiopathic arthritis.


Subject(s)
Arthropathy, Neurogenic/diagnosis , Arthropathy, Neurogenic/genetics , Coxa Vara/diagnosis , Coxa Vara/genetics , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Proteoglycans/genetics , Synovitis/diagnosis , Synovitis/genetics , Adolescent , Adult , Child , Child, Preschool , Exons/genetics , Female , Genetic Association Studies/methods , Humans , Male , Middle Aged , Mutation , Proteoglycans/metabolism , Retrospective Studies , Sequence Deletion , Exome Sequencing/methods
7.
Cancer Cell ; 32(5): 574-589.e6, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29136504

ABSTRACT

ARID1A, an SWI/SNF chromatin-remodeling gene, is commonly mutated in cancer and hypothesized to be tumor suppressive. In some hepatocellular carcinoma patients, ARID1A was highly expressed in primary tumors but not in metastatic lesions, suggesting that ARID1A can be lost after initiation. Mice with liver-specific homozygous or heterozygous Arid1a loss were resistant to tumor initiation while ARID1A overexpression accelerated initiation. In contrast, homozygous or heterozygous Arid1a loss in established tumors accelerated progression and metastasis. Mechanistically, gain of Arid1a function promoted initiation by increasing CYP450-mediated oxidative stress, while loss of Arid1a within tumors decreased chromatin accessibility and reduced transcription of genes associated with migration, invasion, and metastasis. In summary, ARID1A has context-dependent tumor-suppressive and oncogenic roles in cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , Nuclear Proteins/genetics , Oncogenes/genetics , Animals , Blotting, Western , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Nuclear Proteins/metabolism , RNA Interference , Transcription Factors
8.
Elife ; 62017 07 11.
Article in English | MEDLINE | ID: mdl-28695822

ABSTRACT

Sequencing studies have implicated haploinsufficiency of ARID1B, a SWI/SNF chromatin-remodeling subunit, in short stature (Yu et al., 2015), autism spectrum disorder (O'Roak et al., 2012), intellectual disability (Deciphering Developmental Disorders Study, 2015), and corpus callosum agenesis (Halgren et al., 2012). In addition, ARID1B is the most common cause of Coffin-Siris syndrome, a developmental delay syndrome characterized by some of the above abnormalities (Santen et al., 2012; Tsurusaki et al., 2012; Wieczorek et al., 2013). We generated Arid1b heterozygous mice, which showed social behavior impairment, altered vocalization, anxiety-like behavior, neuroanatomical abnormalities, and growth impairment. In the brain, Arid1b haploinsufficiency resulted in changes in the expression of SWI/SNF-regulated genes implicated in neuropsychiatric disorders. A focus on reversible mechanisms identified Insulin-like growth factor (IGF1) deficiency with inadequate compensation by Growth hormone-releasing hormone (GHRH) and Growth hormone (GH), underappreciated findings in ARID1B patients. Therapeutically, GH supplementation was able to correct growth retardation and muscle weakness. This model functionally validates the involvement of ARID1B in human disorders, and allows mechanistic dissection of neurodevelopmental diseases linked to chromatin-remodeling.


Subject(s)
Developmental Disabilities/genetics , Haploinsufficiency , Mental Disorders/genetics , Transcription Factors/deficiency , Animals , Behavior, Animal , Brain/pathology , Developmental Disabilities/physiopathology , Disease Models, Animal , Gene Expression Regulation , Growth Hormone-Releasing Hormone/metabolism , Heterozygote , Insulin-Like Growth Factor I/metabolism , Mental Disorders/physiopathology , Mice , Protein Serine-Threonine Kinases/metabolism
9.
Cell Stem Cell ; 18(4): 456-66, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27044474

ABSTRACT

Mammals have partially lost the extensive regenerative capabilities of some vertebrates, possibly as a result of chromatin-remodeling mechanisms that enforce terminal differentiation. Here, we show that deleting the SWI/SNF component Arid1a substantially improves mammalian regeneration. Arid1a expression is suppressed in regenerating tissues, and genetic deletion of Arid1a increases tissue repair following an array of injuries. Arid1a deficiency in the liver increases proliferation, reduces tissue damage and fibrosis, and improves organ function following surgical resection and chemical injuries. Hepatocyte-specific deletion is also sufficient to increase proliferation and regeneration without excessive overgrowth, and global Arid1a disruption potentiates soft tissue healing in the ear. We show that Arid1a loss reprograms chromatin to restrict promoter access by transcription factors such as C/ebpα, which enforces differentiation, and E2F4, which suppresses cell-cycle re-entry. Thus, epigenetic reprogramming mediated by deletion of a single gene improves mammalian regeneration and suggests strategies to promote tissue repair after injury.


Subject(s)
DNA-Binding Proteins/metabolism , Liver Regeneration , Liver/metabolism , Nuclear Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...