Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(9): e2320129121, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38377195

ABSTRACT

Despite numerous female contraceptive options, nearly half of all pregnancies are unintended. Family planning choices for men are currently limited to unreliable condoms and invasive vasectomies with questionable reversibility. Here, we report the development of an oral contraceptive approach based on transcriptional disruption of cyclical gene expression patterns during spermatogenesis. Spermatogenesis involves a continuous series of self-renewal and differentiation programs of spermatogonial stem cells (SSCs) that is regulated by retinoic acid (RA)-dependent activation of receptors (RARs), which control target gene expression through association with corepressor proteins. We have found that the interaction between RAR and the corepressor silencing mediator of retinoid and thyroid hormone receptors (SMRT) is essential for spermatogenesis. In a genetically engineered mouse model that negates SMRT-RAR binding (SMRTmRID mice), the synchronized, cyclic expression of RAR-dependent genes along the seminiferous tubules is disrupted. Notably, the presence of an RA-resistant SSC population that survives RAR de-repression suggests that the infertility attributed to the loss of SMRT-mediated repression is reversible. Supporting this notion, we show that inhibiting the action of the SMRT complex with chronic, low-dose oral administration of a histone deacetylase inhibitor reversibly blocks spermatogenesis and fertility without affecting libido. This demonstration validates pharmacologic targeting of the SMRT repressor complex for non-hormonal male contraception.


Subject(s)
DNA-Binding Proteins , Repressor Proteins , Humans , Female , Male , Animals , Mice , DNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Co-Repressor Proteins/genetics , Nuclear Receptor Co-Repressor 2/genetics , Tretinoin/pharmacology , Contraception , Nuclear Receptor Co-Repressor 1
2.
Cell Chem Biol ; 24(7): 858-869.e5, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28669525

ABSTRACT

Cutaneous reactions represent one of the most common adverse drug effects observed in clinical trials leading to substantial compound attrition. Three negative allosteric modulators (NAMs) of metabotropic glutamate receptors (mGluRs), which represent an important target for neurological diseases, developed by Pfizer, were recently failed in preclinical development due to delayed type IV skin hypersensitivity observed in non-human primates (NHPs). Here we employed large-scale phenotypic profiling in standardized panels of human primary cell/co-culture systems to characterize the skin toxicity mechanism(s) of mGluR5 NAMs from two different series. Investigation of a database of chemicals tested in these systems and transcriptional profiling suggested that the mechanism of toxicity may involve modulation of nuclear receptor targets RAR/RXR, and/or VDR with AhR antagonism. The studies reported here demonstrate how phenotypic profiling of preclinical drug candidates using human primary cells can provide insights into the mechanisms of toxicity and inform early drug discovery and development campaigns.


Subject(s)
Fibroblasts/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Skin Diseases/chemically induced , Allosteric Regulation , Cells, Cultured , Databases, Chemical , Dinoprostone/metabolism , Down-Regulation/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-2/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Protein Binding , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/chemistry , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/metabolism , Skin Diseases/metabolism , Skin Diseases/pathology , Small Molecule Libraries/chemistry , Small Molecule Libraries/toxicity , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
3.
Clin Diabetes ; 35(1): 27-34, 2017 01.
Article in English | MEDLINE | ID: mdl-28144043

ABSTRACT

IN BRIEF This study quantitatively measures diabetes stigma and its associated psychosocial impact in a large population of U.S. patients with type 1 or type 2 diabetes using an online survey sent to 12,000 people with diabetes. A majority of respondents with type 1 (76%) or type 2 (52%) diabetes reported that diabetes comes with stigma. Perceptions of stigma were significantly higher among respondents with type 1 diabetes than among those with type 2 diabetes, with the highest rate in parents of children with type 1 diabetes (83%) and the lowest rate in people with type 2 diabetes who did not use insulin (49%). Our results suggest that a disturbingly high percentage of people with diabetes experience stigma, particularly those with type 1 or type 2 diabetes who are on intensive insulin therapy. The experience of stigma disproportionately affects those with a higher BMI, higher A1C, and poorer self-reported blood glucose control, suggesting that those who need the most help are also the most affected by stigma.

4.
Cell Stem Cell ; 16(1): 18-31, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25465113

ABSTRACT

Cellular identity is established by genetic, epigenetic, and environmental factors that regulate organogenesis and tissue homeostasis. Although some flexibility in fate potential is beneficial to overall organ health, dramatic changes in cellular identity can have disastrous consequences. Emerging data within the field of pancreas biology are revising current beliefs about how cellular identity is shaped by developmental and environmental cues under homeostasis and stress conditions. Here, we discuss the changes occurring in cellular states upon fate modulation and address how our understanding of the nature of this fluidity is shaping therapeutic approaches to pancreatic disorders such as diabetes and cancer.


Subject(s)
Cell Dedifferentiation , Homeostasis , Organogenesis , Pancreas/cytology , Pancreas/growth & development , Pancreatic Diseases/pathology , Cell Lineage , Epigenesis, Genetic , Humans , Models, Biological
5.
PLoS One ; 9(7): e102125, 2014.
Article in English | MEDLINE | ID: mdl-25010227

ABSTRACT

Normal tissue architecture is disrupted following injury, as resident tissue cells become damaged and immune cells are recruited to the site of injury. While injury and inflammation are critical to tissue remodeling, the inability to resolve this response can lead to the destructive complications of chronic inflammation. In the pancreas, acinar cells of the exocrine compartment respond to injury by transiently adopting characteristics of progenitor cells present during embryonic development. This process of de-differentiation creates a window where a mature and stable cell gains flexibility and is potentially permissive to changes in cellular fate. How de-differentiation can turn an acinar cell into another cell type (such as a pancreatic ß-cell), or a cell with cancerous potential (as in cases of deregulated Kras activity) is of interest to both the regenerative medicine and cancer communities. While it is known that inflammation and acinar de-differentiation increase following pancreatic injury, it remains unclear which immune cells are involved in this process. We used a combination of genetically modified mice, immunological blockade and cellular characterization to identify the immune cells that impact pancreatic regeneration in an in vivo model of pancreatitis. We identified the innate inflammatory response of macrophages and neutrophils as regulators of pancreatic regeneration. Under normal conditions, mild innate inflammation prompts a transient de-differentiation of acinar cells that readily dissipates to allow normal regeneration. However, non-resolving inflammation developed when elevated pancreatic levels of neutrophils producing interferon-γ increased iNOS levels and the pro-inflammatory response of macrophages. Pancreatic injury improved following in vivo macrophage depletion, iNOS inhibition as well as suppression of iNOS levels in macrophages via interferon-γ blockade, supporting the impairment in regeneration and the development of chronic inflammation arises from aberrant activation of the innate inflammatory response. Collectively these studies identify targetable inflammatory factors that can be used to influence the development of non-resolving inflammation and pancreatic regeneration following injury.


Subject(s)
Immunity, Innate , Pancreas/immunology , Pancreas/pathology , Regeneration , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Dedifferentiation/drug effects , Ceruletide , Homeodomain Proteins/metabolism , Immunity, Innate/drug effects , Inflammation/pathology , Interferon-gamma/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Pancreas/physiopathology , Regeneration/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
Nat Rev Endocrinol ; 10(5): 253-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24566816

ABSTRACT

A genetic analysis of patients with permanent neonatal diabetes mellitus has revealed functional conservation of transcription factors critical for ß-cell development in both mouse and man. This finding supports the use of mice for modelling human disease but also highlights the need for additional human-specific studies of ß-cell function.


Subject(s)
Diabetes Mellitus/genetics , Homeodomain Proteins/genetics , Mutation/genetics , Pancreas/growth & development , Pancreas/metabolism , Transcription Factors/genetics , Animals , Female , Humans , Male
7.
Genesis ; 50(5): 415-23, 2012 May.
Article in English | MEDLINE | ID: mdl-22162152

ABSTRACT

Retinoic acid (RA) signaling is necessary for proper patterning and morphogenesis during embryonic development. Tissue-specific RA signaling requires precise spatial and temporal synthesis of RA from retinal by retinaldehyde dehydrogenases (Raldh) and the conversion of retinol to retinal by retinol dehydrogenases (Rdh) of the short-chain dehydrogenase/reducatase gene family (SDR). The SDR, retinol dehydrogenase 10 (RDH10), is a major contributor to retinal biosynthesis during mid-gestation. We have identified a missense mutation in the Rdh10 gene (Rdh10(m366Asp) ) using an N-ethyl-N-nitrosourea-induced forward genetic screen that result in reduced RA levels and signaling during embryonic development. Rdh10(m366Asp) mutant embryos have unique phenotypes, such as edema, a massive midline facial cleft, and neurogenesis defects in the forebrain, that will allow the identification of novel RA functions.


Subject(s)
Alcohol Oxidoreductases/genetics , Mutation, Missense , Signal Transduction , Tretinoin/metabolism , Animals , Cloning, Molecular , Mice
8.
Mol Cell Biol ; 31(16): 3277-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21670153

ABSTRACT

Cellular retinol-binding protein type I (CrbpI), encoded by Rpb1, serves as a chaperone of retinol homeostasis, but its physiological effects remain incompletely understood. We show here that the Rbp1(-/-) mouse has disrupted retinoid homeostasis in multiple tissues, with abnormally high 9-cis-retinoic acid (9cRA), a pancreas autacoid that attenuates glucose-stimulated insulin secretion. The Rbp1(-/-) pancreas has increased retinol and intense ectopic expression of Rpb2 mRNA, which encodes CrbpII: both would contribute to increased ß-cell 9cRA biosynthesis. 9cRA in Rbp1(-/-) pancreas resists postprandial and glucose-induced decreases. Rbp1(-/-) mice have defective islet expression of genes involved in glucose sensing and insulin secretion, as well as islet α-cell infiltration, which contribute to reduced glucose-stimulated insulin secretion, high glucagon secretion, an abnormally high rate of gluconeogenesis, and hyperglycemia. A diet rich in vitamin A (as in a standard chow diet) increases pancreas 9cRA and impairs glucose tolerance. Crbp1 attenuates the negative impact of vitamin A (retinol) on glucose tolerance, regardless of the dietary retinol content. Rbp1(-/-) mice have an increased rate of fatty acid oxidation and resist obesity when fed a high-fat diet. Thus, glucose homeostasis and energy metabolism rely on Rbp1 expression and its moderation of pancreas retinol and of the autacoid 9cRA.


Subject(s)
Glucose/metabolism , Homeostasis , Pancreas/metabolism , Retinol-Binding Proteins, Cellular/physiology , Tretinoin/metabolism , Alitretinoin , Animals , Antineoplastic Agents , Energy Metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Retinol-Binding Proteins, Cellular/deficiency , Vitamin A/metabolism
9.
Cancer Cell ; 19(4): 441-55, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21481787

ABSTRACT

Chronic pancreatitis is a well-known risk factor for pancreatic ductal adenocarcinoma (PDA) development in humans, and inflammation promotes PDA initiation and progression in mouse models of the disease. However, the mechanistic link between inflammatory damage and PDA initiation is unclear. Using a Kras-driven mouse model of PDA, we establish that the inflammatory mediator Stat3 is a critical component of spontaneous and pancreatitis-accelerated PDA precursor formation and supports cell proliferation, metaplasia-associated inflammation, and MMP7 expression during neoplastic development. Furthermore, we show that Stat3 signaling enforces MMP7 expression in PDA cells and that MMP7 deletion limits tumor size and metastasis in mice. Finally, we demonstrate that serum MMP7 level in human patients with PDA correlated with metastatic disease and survival.


Subject(s)
Adenocarcinoma/etiology , Carcinoma, Pancreatic Ductal/etiology , Matrix Metalloproteinase 7/physiology , Pancreatic Neoplasms/etiology , STAT3 Transcription Factor/physiology , Acute Disease , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Female , Genes, ras , Humans , Interleukin-6/physiology , Male , Matrix Metalloproteinase 7/blood , Mice , Middle Aged , Pancreatic Neoplasms/pathology , Pancreatitis/complications , Pancreatitis/pathology , Proto-Oncogene Proteins p21(ras)/genetics
10.
Proc Natl Acad Sci U S A ; 107(50): 21884-9, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21115832

ABSTRACT

The all-trans-retinoic acid (atRA) isomer, 9-cis-retinoic acid (9cRA), activates retinoic acid receptors (RARs) and retinoid X receptors (RXRs) in vitro. RARs control multiple genes, whereas RXRs serve as partners for RARs and other nuclear receptors that regulate metabolism. Physiological function has not been determined for 9cRA, because it has not been detected in serum or multiple tissues with analytically validated assays. Here, we identify 9cRA in mouse pancreas by liquid chromatography/tandem mass spectrometry (LC/MS/MS), and show that 9cRA decreases with feeding and after glucose dosing and varies inversely with serum insulin. 9cRA reduces glucose-stimulated insulin secretion (GSIS) in mouse islets and in the rat ß-cell line 832/13 within 15 min by reducing glucose transporter type 2 (Glut2) and glucokinase (GK) activities. 9cRA also reduces Pdx-1 and HNF4α mRNA expression, ∼8- and 80-fold, respectively: defects in Pdx-1 or HNF4α cause maturity onset diabetes of the young (MODY4 and 1, respectively), as does a defective GK gene (MODY2). Pancreas ß-cells generate 9cRA, and mouse models of reduced ß-cell number, heterozygous Akita mice, and streptozotocin-treated mice have reduced 9cRA. 9cRA is abnormally high in glucose-intolerant mice, which have ß-cell hypertropy, including mice with diet-induced obesity (DIO) and ob/ob and db/db mice. These data establish 9cRA as a pancreas-specific autacoid with multiple mechanisms of action and provide unique insight into GSIS.


Subject(s)
Autacoids/metabolism , Glucose/pharmacology , Insulin/metabolism , Pancreas/drug effects , Pancreas/metabolism , Tretinoin/metabolism , Alitretinoin , Animals , Antineoplastic Agents/metabolism , Cell Line , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Pancreas/cytology , Rats
11.
FASEB J ; 24(3): 823-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19890016

ABSTRACT

All-trans-retinoic acid (atRA) supports embryonic development, central nervous system function, and the immune response. atRA initiates neurogenesis and dendritic growth in the hippocampus and is required for spatial memory; superphysiological atRA inhibits neurogenesis, causes teratology and/or embryo toxicity, and alters cognitive function and behavior. Because abnormal atRA shares pathological conditions with alcoholism, inhibition of retinol (vitamin A) activation into atRA has been credited widely as a mechanism of ethanol toxicity. Here, we analyze the effects of ethanol on retinoid concentrations in vivo during normal vitamin A nutriture, using sensitive and analytically robust assays. Ethanol either increased or had no effect on atRA, regardless of changes in retinol and retinyl esters. Acute ethanol (3.5 g/kg) increased atRA in adult hippocampus (1.6-fold), liver (2.4-fold), and testis (1.5-fold). Feeding dams a liquid diet with 6.5% ethanol from embryonic day 13 (e13) to e19 increased atRA in fetal hippocampus (up to 20-fold) and cortex (up to 50-fold), depending on blood alcohol content. One-month feeding of the 6.5% ethanol diet increased atRA in adult hippocampus (20-fold), cortex (2-fold), testis (2-fold), and serum (10-fold). Tissue-specific increases in retinoid dehydrogenase mRNAs and activities, extrahepatic retinol concentrations, and atRA catabolism combined to produce site-specific effects. Because a sustained increase in atRA has deleterious effects on the central nervous system and embryo development, these data suggest that superphysiological atRA contributes to ethanol pathological conditions, including cognitive dysfunction and fetal alcohol syndrome.-Kane, M. A., Folias, A. E., Wang, C., Napoli, J. L. Ethanol elevates physiological all-trans-retinoic acid levels in select loci through altering retinoid metabolism in multiple loci: a potential mechanism of ethanol toxicity.


Subject(s)
Ethanol/toxicity , Retinoids/metabolism , Tretinoin/metabolism , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chromatography, Liquid , Female , Fetal Alcohol Spectrum Disorders/etiology , Fetal Alcohol Spectrum Disorders/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Liver/drug effects , Liver/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Pregnancy , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Testis/drug effects , Testis/metabolism , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism
12.
Cell ; 139(3): 597-609, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19879845

ABSTRACT

Extrinsic signals controlling generation of neocortical neurons during embryonic life have been difficult to identify. In this study we demonstrate that the dorsal forebrain meninges communicate with the adjacent radial glial endfeet and influence cortical development. We took advantage of Foxc1 mutant mice with defects in forebrain meningeal formation. Foxc1 dosage and loss of meninges correlated with a dramatic reduction in both neuron and intermediate progenitor production and elongation of the neuroepithelium. Several types of experiments demonstrate that retinoic acid (RA) is the key component of this secreted activity. In addition, Rdh10- and Raldh2-expressing cells in the dorsal meninges were either reduced or absent in the Foxc1 mutants, and Rdh10 mutants had a cortical phenotype similar to the Foxc1 null mutants. Lastly, in utero RA treatment rescued the cortical phenotype in Foxc1 mutants. These results establish RA as a potent, meningeal-derived cue required for successful corticogenesis.


Subject(s)
Meninges/metabolism , Neurogenesis , Neurons/cytology , Tretinoin/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , In Vitro Techniques , Mice , Prosencephalon/cytology , Prosencephalon/metabolism
13.
Anal Biochem ; 378(1): 71-9, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18410739

ABSTRACT

We report robust HPLC/UV methods for quantifying retinyl esters (RE), retinol (ROL), and retinal (RAL) applicable to diverse biological samples with lower limits of detection of 0.7, 0.2, and 0.2 pmol, respectively, and linear ranges greater than 3 orders of magnitude. These assays function well with small, complex biological samples (10-20mg tissue). Coefficients of variation range from 5.9 to 10.0% (intraday) and from 5.9 to 11.0% (interday). Quantification of endogenous RE, ROL, and RAL in mouse serum and tissues (liver, kidney, adipose, muscle, spleen, testis, skin, brain, and brain regions) reveals utility. Ability to discriminate spatial concentrations of ROL and RE is illustrated with C57BL/6 mouse brain loci (hippocampus, cortex, olfactory bulb, thalamus, cerebellum, and striatum). We also developed a method to distinguish isomeric forms of ROL to investigate precursors of retinoic acid. The ROL isomer assay has limits of detection between 3.5 and 4.5 pmol and has a linear range and coefficient of variation similar to those of the ROL/RE and RAL assays. The assays described here provide for sensitive and rigorous quantification of endogenous RE, ROL, and RAL to elucidate retinoid homeostasis in disease states such as Alzheimer's disease, type 2 diabetes, obesity, and cancer.


Subject(s)
Adipose Tissue , Chromatography, High Pressure Liquid/methods , Esters/analysis , Liver , Retinaldehyde/analysis , Spectrophotometry/methods , Vitamin A/analysis , Adipose Tissue/metabolism , Animals , Esters/chemistry , Esters/metabolism , Isomerism , Liver/metabolism , Male , Mice , Molecular Structure , Oximes/metabolism , Reproducibility of Results , Retinaldehyde/metabolism , Ultraviolet Rays , Vitamin A/metabolism
14.
Anal Chem ; 80(5): 1702-8, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18251521

ABSTRACT

We report an improved tandem mass spectrometric assay for retinoic acid (RA) applicable to in vitro and in vivo biological samples. This liquid chromatography tandem mass spectrometric (LC/MS/MS) assay for direct RA quantification is the most sensitive to date, with a 62.5 attomol lower limit of detection and a linear range spanning greater than 4 orders of magnitude (from 250 attomol to 10 pmol). This assay resolves all-trans-RA (atRA) from its endogenous geometric isomers, is applicable to samples of limited size (10-20 mg of tissue), and functions with complex biological matrixes. Coefficients of variation are as follows: instrumental, < or =2.6%; intraday, 5.2% +/- 0.7%; interday, 6.7% +/- 0.9%. In vitro capabilities are demonstrated by quantification of endogenous RA and RA production (from retinol) in primary cultured astrocytes. Quantification of endogenous atRA and its geometric isomers in 129SV mouse serum and tissues (liver, kidney, adipose, muscle, spleen, testis, and brain) reveals in vivo utility of the assay. The ability to discriminate spatial concentrations of RA in vivo is illustrated with C57BL/6 mouse brain loci (hippocampus, cortex, olfactory bulb, thalamus, cerebellum, and striatum), as well as with Lewis rat proximal/distal mammary gland regions during various morphological stages: virgin, early pregnancy (e7), late pregnancy (e20), lactating (day 4), involuting day 1, and involuting day 11. This assay provides the sensitivity necessary for direct, endogenous RA quantification necessary to elucidate RA function, e.g., in neurogenesis, morphogenesis, and the contribution of altered RA homeostasis to diseases, such as Alzheimer's disease, type 2 diabetes, obesity, and cancer.


Subject(s)
Tandem Mass Spectrometry/methods , Tretinoin/chemistry , Tretinoin/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Sprague-Dawley , Tretinoin/analogs & derivatives , Vitamin A/administration & dosage , Vitamin A/metabolism
15.
DNA Repair (Amst) ; 2(9): 1007-13, 2003 Sep 18.
Article in English | MEDLINE | ID: mdl-12967657

ABSTRACT

BRCA1 and BRCA2 proteins act in repair of interstrand crosslinks (ICLs) and maintenance of genome stability and are known to be part of the Fanconi anemia (FA) pathway. We have investigated the role of the BRCA1 and BRCA2 genes in genome stability following ICL damage in normal and FA cells. To circumvent cell lethality of complete disruptions in BRCA1 or BRCA2, small inhibitory RNA (siRNA) was used to transiently deplete the expression of the proteins. Using chromosomal stability after ICL damage as the end point, we find that BRCA1 functions in more than just the FA pathway for genome maintenance, whereas BRCA2 appears to act predominantly in the FA pathway. Depletion of BRCA1 causes a marked decrease, although not a complete absence of, ubiquitination of FANCD2. In contrast to BRCA1, BRCA2 is not needed for normal ubiquitination of FANCD2 after DNA damage, a requirement for the FA pathway to function. Thus, BRCA2 is epistatic to FA genes for ICL repair, but not for damage-induced modification of FANCD2 and may act downstream form FANCD2.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , DNA Damage , Fanconi Anemia/genetics , RNA, Small Interfering/pharmacology , DNA Repair , DNA-Binding Proteins , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein , Humans , Karyotyping , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins
16.
Hum Mol Genet ; 11(21): 2591-7, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12354784

ABSTRACT

Fanconi anemia (FA) is a rare autosomal recessive disease characterized by skeletal defects, anemia, chromosomal instability and increased risk of leukemia. At the cellular level FA is characterized by increased sensitivity to agents forming interstrand crosslinks (ICL) in DNA. Six FA genes have been cloned and interactions among individual FANC proteins have been found. The FANCD2 protein co-localizes in nuclear foci with the BRCA1 protein following DNA damage and during S-phase, requiring the FANCA, C, E and G proteins to do so. This finding may reflect a direct role for the BRCA1 protein in double strand break (DSB) repair and interaction with the FANC proteins. Therefore interactions between BRCA1 and the FANC proteins were investigated. Among the known FANC proteins, we find evidence for direct interaction only between the FANCA protein and BRCA1. The evidence rests on three different tests: yeast two-hybrid analysis, coimmunoprecipitation from in vitro synthesis, and coimmunoprecipitation from cell extracts. The amino terminal portion of FANCA and the central part (aa 740-1083) of BRCA1 contain the sites of interaction. The interaction does not depend on DNA damage, thus FANCA and BRCA1 are constitutively interacting. The demonstrated interaction directly connects BRCA1 to the FA pathway of DNA repair.


Subject(s)
BRCA1 Protein/metabolism , DNA-Binding Proteins , Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group D2 Protein , Humans , In Vitro Techniques , Nuclear Proteins/metabolism , Precipitin Tests , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...