Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Subcell Biochem ; 81: 21-76, 2016.
Article in English | MEDLINE | ID: mdl-27830500

ABSTRACT

Multiple binding and transport proteins facilitate many aspects of retinoid biology through effects on retinoid transport, cellular uptake, metabolism, and nuclear delivery. These include the serum retinol binding protein sRBP (aka Rbp4), the plasma membrane sRBP receptor Stra6, and the intracellular retinoid binding-proteins such as cellular retinol-binding proteins (CRBP) and cellular retinoic acid binding-proteins (CRABP). sRBP transports the highly lipophilic retinol through an aqueous medium. The major intracellular retinol-binding protein, CRBP1, likely enhances efficient retinoid use by providing a sink to facilitate retinol uptake from sRBP through the plasma membrane or via Stra6, delivering retinol or retinal to select enzymes that generate retinyl esters or retinoic acid, and protecting retinol/retinal from excess catabolism or opportunistic metabolism. Intracellular retinoic acid binding-proteins (CRABP1 and 2, and FABP5) seem to have more diverse functions distinctive to each, such as directing retinoic acid to catabolism, delivering retinoic acid to specific nuclear receptors, and generating non-canonical actions. Gene ablation of intracellular retinoid binding-proteins does not cause embryonic lethality or gross morphological defects. Metabolic and functional defects manifested in knockouts of CRBP1, CRBP2 and CRBP3, however, illustrate their essentiality to health, and in the case of CRBP2, to survival during limited dietary vitamin A. Future studies should continue to address the specific molecular interactions that occur between retinoid binding-proteins and their targets and their precise physiologic contributions to retinoid homeostasis and function.


Subject(s)
Retinoids/physiology , Retinol-Binding Proteins, Cellular/physiology , Alcohol Oxidoreductases/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Biological Transport , Cell Nucleus/metabolism , Eye/metabolism , Gene Knockout Techniques , Homeostasis , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Models, Molecular , Neoplasm Proteins/metabolism , Protein Conformation , Receptors, Cytoplasmic and Nuclear/metabolism , Retinaldehyde/metabolism , Retinol-Binding Proteins, Cellular/chemistry , Retinol-Binding Proteins, Cellular/deficiency , Retinol-Binding Proteins, Cellular/genetics , Signal Transduction/physiology , Tretinoin/metabolism , Vitamin A/metabolism , Vitamin A/toxicity
2.
FASEB J ; 27(5): 1904-16, 2013 May.
Article in English | MEDLINE | ID: mdl-23362116

ABSTRACT

Cellular retinol-binding protein, type I (CrbpI), encoded by retinol-binding protein, type 1 (Rbp1), is a chaperone of vitamin A (retinol) that is epigenetically silenced in ~25% of human breast cancers. CrbpI delivers vitamin A to enzymes for metabolism into an active metabolite, all-trans retinoic acid (atRA), where atRA is essential to cell proliferation, apoptosis, differentiation, and migration. Here, we show the effect of CrbpI loss on mammary atRA homeostasis using the Rbp1(-/-) mouse model. Rbp1(-/-) mouse mammary tissue has disrupted retinoid homeostasis that results in 40% depleted endogenous atRA. CrbpI loss and atRA depletion precede defects in atRA biosynthesis enzyme expression. Compensation by CrbpIII as a retinoid chaperone does not functionally replace CrbpI. Mammary subcellular fractions isolated from Rbp1(-/-) mice have altered retinol dehydrogenase/reductase (Rdh) enzyme activity that results in 24-42% less atRA production. Rbp1(-/-) mammary tissue has epithelial hyperplasia, stromal hypercellularity, increased collagen, and increased oxidative stress characteristic of atRA deficiency and early tissue dysfunction that precedes tumor formation. Consistent with the findings from the Rbp1(-/-) mouse, tumorigenic epithelial cells lacking CrbpI expression produce 51% less atRA. Together, these data show that CrbpI loss disrupts atRA homeostasis in mammary tissue, resulting in microenvironmental defects similar to those observed at the early stages of tumorigenesis.


Subject(s)
Mammary Glands, Animal/metabolism , Retinol-Binding Proteins, Cellular/physiology , Alcohol Oxidoreductases/metabolism , Animals , Female , Homeostasis , Mice , Retinol-Binding Proteins, Cellular/deficiency , Retinol-Binding Proteins, Cellular/metabolism , Tretinoin/metabolism
3.
Med Hypotheses ; 88: 60-2, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880640

ABSTRACT

Vitamin A is a fat-soluble vitamin required for many physiological functions. The intracellular transport of vitamin A is assisted by proteins called cellular retinol-binding proteins (CRBP I/II). The absorption, storage and usage of vitamin A are regulated by a protein called lecithin:retinol acyltransferase (LRAT), a retinol-related enzyme that transfers an acyl group derived from an sn-1 position of phosphatidylcholine to retinol. LRAT is a member of the protein family which includes HRAS-like tumor suppressors (HRASLS). However, the HRASLS proteins never use retinol as an acyl acceptor. The mechanisms underlying the different substrate specificities between LRAT and HRASLS proteins are unknown. We propose in this report that LRAT physically interacts with CRBP and the LRAT-CRBP complex represents the binding pockets for both an acyl group and retinol, thus assuring the substrate specificity of LRAT.


Subject(s)
Acyltransferases/physiology , Retinol-Binding Proteins, Cellular/physiology , Vitamin A/chemistry , Acyltransferases/chemistry , Esterification , Esters/chemistry , Humans , Lecithins/chemistry , Liver/metabolism , Magnetic Resonance Spectroscopy , Models, Theoretical , Retinol-Binding Proteins, Cellular/chemistry , Substrate Specificity , Vitamin A Deficiency/immunology
4.
PLoS One ; 11(3): e0152142, 2016.
Article in English | MEDLINE | ID: mdl-27008505

ABSTRACT

Despite recent increases in the cure rate of acute lymphoblastic leukemia (ALL), adult ALL remains a high-risk disease that exhibits a high relapse rate. In this study, we found that the histone demethylase retinoblastoma binding protein-2 (RBP2) was overexpressed in both on-going and relapse cases of adult ALL, which revealed that RBP2 overexpression was not only involved in the pathogenesis of ALL but that its overexpression might also be related to relapse of the disease. RBP2 knockdown induced apoptosis and attenuated leukemic cell viability. Our results demonstrated that BCL2 is a novel target of RBP2 and supported the notion of RBP2 being a regulator of BCL2 expression via directly binding to its promoter. As the role of RBP2 in regulating apoptosis was confirmed, RBP2 overexpression and activation of BCL2 might play important roles in ALL development and progression.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Retinol-Binding Proteins, Cellular/physiology , Adult , Apoptosis/genetics , Blotting, Western , Chromatin Immunoprecipitation , Disease Progression , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Real-Time Polymerase Chain Reaction , Up-Regulation
5.
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
6.
Mol Cell Biol ; 30(14): 3412-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20498279

ABSTRACT

Adipogenesis is governed by a well-documented cascade of transcription factors. However, less is known about non-transcription factors that govern early stages of adipogenesis. Here we show that cellular retinol-binding protein type I (CRBP-I), a small cytosolic binding protein for retinol and retinaldehyde, is specifically restricted to preadipocytes in white adipose tissue. The absence of CRBP-I in mice (CRBP-I-KO mice) leads to increased adiposity. Despite increased adiposity, CRBP-I-KO mice remain more glucose tolerant and insulin sensitive during high-fat-diet feeding. 3T3-L1 cells deficient in CRBP-I or mouse embryonic fibroblasts derived from CRBP-I-KO mice had increased adipocyte differentiation and triglyceride (TG) accumulation. This was due to increased expression and activity of PPAR gamma, while other transcription factor pathways in early and late differentiation remained unchanged. Conversely, the overexpression of CRBP-I in 3T3-L1 cells results in decreased TG accumulation. In conclusion, CRBP-I is a cytosolic protein specifically expressed in preadipocytes that regulates adipocyte differentiation in part by affecting PPAR gamma activity.


Subject(s)
Adipogenesis/physiology , Retinol-Binding Proteins, Cellular/physiology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adiposity , Animals , Base Sequence , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Glucose Tolerance Test , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/pathology , PPAR gamma/metabolism , Phenotype , RNA, Small Interfering/genetics , Retinol-Binding Proteins, Cellular/antagonists & inhibitors , Retinol-Binding Proteins, Cellular/deficiency , Retinol-Binding Proteins, Cellular/genetics , Triglycerides/metabolism
7.
Int J Oncol ; 37(6): 1379-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21042705

ABSTRACT

We have previously reported a simple technique that combines microarray data from clinical bladder cancer (BC) specimens with those from a BC cell line (BOY) treated with a pharmacological demethylating agent [5-aza-2'-deoxycytidine (5-aza-dC)] to find candidate genes that have tumor suppressive functions. We focused on the cellular retinol-binding protein 1 (CRBP1) gene that was selected by using the microarray data. As CRBP1 regulates intracellular retinoic acid (vitamin A) homeostasis, which is involved in morphogenesis, and cellular proliferation and differentiation, the loss of CRBP1 could cause tumorigenesis in BC. We hypothesized that the inactivation of the CRBP1 gene through CpG methylation contributes to cell viability, including the migration and invasion activity of human BC cells. After the 5-aza-dC treatment, the mRNA and protein expression levels of CRBP1 markedly increased in all BOY and T24 BC cell lines. Combined bisulfite-restriction analysis and bisulfite DNA sequencing revealed that promoter CpG hypermethylation existed in 28 out of the 65 BCs (43%) and in none of the 16 normal bladder epithelia (NBEs). Conversely, CRBP1 mRNA expression in the BCs was significantly lower than that in the NBEs (0.63 ± 0.11 vs. 4.92 ± 0.80, p<0.0001). We found significant inhibition of cell growth (p<0.0001) and migration (p<0.0001) in the CRBP1 stable transfectants compared to the control cell line, in a cell proliferation and wound-healing assay, respectively. In conclusion, the aberrant CpG hypermethylation of the CRBP1 gene promoter could be involved in the development of BC. We demonstrate here for the first time that the CRBP1 gene could have a tumor suppressive function in BC.


Subject(s)
Carcinoma/pathology , Cell Movement/genetics , Cell Proliferation , CpG Islands/genetics , DNA Methylation/physiology , Retinol-Binding Proteins, Cellular/genetics , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Base Sequence , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Neoplasm Invasiveness , Retinol-Binding Proteins, Cellular/metabolism , Retinol-Binding Proteins, Cellular/physiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
8.
J Biol Chem ; 283(20): 13510-9, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18348983

ABSTRACT

The intestine and other tissues are able to synthesize retinyl esters in an acyl-CoA-dependent manner involving an acyl-CoA:retinol acyltransferase (ARAT). However, the molecular identity of this ARAT has not been established. Recent studies of lecithin:retinol acyltransferase (LRAT)-deficient mice indicate that LRAT is responsible for the preponderance of retinyl ester synthesis in the body, aside from in the intestine and adipose tissue. Our present studies, employing a number of mutant mouse models, identify diacylglycerol acyltransferase 1 (DGAT1) as an important intestinal ARAT in vivo. The contribution that DGAT1 makes to intestinal retinyl ester synthesis becomes greater when a large pharmacologic dose of retinol is administered by gavage to mice. Moreover, when large retinol doses are administered another intestinal enzyme(s) with ARAT activity becomes apparent. Surprisingly, although DGAT1 is expressed in adipose tissue, DGAT1 does not catalyze retinyl ester synthesis in adipose tissue in vivo. Our data also establish that cellular retinol-binding protein, type II (CRBPII), which is expressed solely in the adult intestine, in vivo channels retinol to LRAT for retinyl ester synthesis. Contrary to what has been proposed in the literature based on in vitro studies, CRBPII does not directly prevent retinol from being acted upon by DGAT1 or other intestinal ARATs in vivo.


Subject(s)
Diacylglycerol O-Acyltransferase/physiology , Retinoids/metabolism , Retinol-Binding Proteins, Cellular/physiology , Adipose Tissue/metabolism , Animals , Chromatography, High Pressure Liquid , Diacylglycerol O-Acyltransferase/genetics , Esters , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Models, Genetic , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Retinol-Binding Proteins, Cellular/genetics
9.
Am J Physiol Endocrinol Metab ; 295(6): E1358-68, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18840764

ABSTRACT

Cellular retinol-binding protein (CRBP) type III (CRBP-III) belongs to the family of intracellular lipid-binding proteins, which includes the adipocyte-binding protein aP2. In the cytosol, CRBP-III binds retinol, the precursor of retinyl ester and the active metabolite retinoic acid. The goal of the present work is to understand the regulation of CRBP-III expression and its role in lipid metabolism. Using EMSAs, luciferase reporter assays, and chromatin immunoprecipitation assays, we found that CRBP-III is a direct target of peroxisome proliferator-activated receptor-gamma (PPARgamma). Moreover, CRBP-III expression was induced in adipose tissue of mice after treatment with the PPARgamma agonist rosiglitazone. To examine a potential role of CRBP-III in regulating lipid metabolism in vivo, CRBP-III-deficient (C-III-KO) mice were maintained on a high-fat diet (HFD). Hepatic steatosis was decreased in HFD-fed C-III-KO compared with HFD-fed wild-type mice. These differences were partly explained by decreased serum free fatty acid levels and decreased free fatty acid efflux from adipose tissue of C-III-KO mice. In addition, the lack of CRBP-III was associated with reduced food intake, increased respiratory energy ratio, and altered body composition, with decreased adiposity and increased lean body mass. Furthermore, expression of genes involved in mitochondrial fatty acid oxidation in brown adipose tissue was increased in C-III-KO mice, and C-III-KO mice were more cold tolerant than wild-type mice fed an HFD. In summary, we demonstrate that CRBP-III is a PPARgamma target gene and plays a role in lipid and whole body energy metabolism.


Subject(s)
Lipid Metabolism/genetics , PPAR gamma/physiology , Retinol-Binding Proteins, Cellular/genetics , Retinol-Binding Proteins, Cellular/physiology , 3T3-L1 Cells , Adipose Tissue, Brown/metabolism , Animals , COS Cells , Chlorocebus aethiops , Diet, Atherogenic , Energy Metabolism/genetics , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Response Elements/physiology , Retinol-Binding Proteins, Cellular/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL