Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
BMC Biol ; 21(1): 124, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37226238

ABSTRACT

BACKGROUND: The axonemal microtubules of primary cilium undergo a conserved protein posttranslational modification (PTM) - polyglutamylation. This reversible procedure is processed by tubulin tyrosine ligase-like polyglutamylases to form secondary polyglutamate side chains, which are metabolized by the 6-member cytosolic carboxypeptidase (CCP) family. Although polyglutamylation modifying enzymes have been linked to ciliary architecture and motility, it was unknown whether they also play a role in ciliogenesis. RESULTS: In this study, we found that CCP5 expression is transiently downregulated upon the initiation of ciliogenesis, but recovered after cilia are formed. Overexpression of CCP5 inhibited ciliogenesis, suggesting that a transient downregulation of CCP5 expression is required for ciliation initiation. Interestingly, the inhibitory effect of CCP5 on ciliogenesis does not rely on its enzyme activity. Among other 3 CCP members tested, only CCP6 can similarly suppress ciliogenesis. Using CoIP-MS analysis, we identified a protein that potentially interacts with CCP - CP110, a known negative regulator of ciliogenesis, whose degradation at the distal end of mother centriole permits cilia assembly. We found that both CCP5 and CCP6 can modulate CP110 level. Particularly, CCP5 interacts with CP110 through its N-terminus. Loss of CCP5 or CCP6 led to the disappearance of CP110 at the mother centriole and abnormally increased ciliation in cycling RPE-1 cells. Co-depletion of CCP5 and CCP6 synergized this abnormal ciliation, suggesting their partially overlapped function in suppressing cilia formation in cycling cells. In contrast, co-depletion of the two enzymes did not further increase the length of cilia, although CCP5 and CCP6 differentially regulate polyglutamate side-chain length of ciliary axoneme and both contribute to limiting cilia length, suggesting that they may share a common pathway in cilia length control. Through inducing the overexpression of CCP5 or CCP6 at different stages of ciliogenesis, we further demonstrated that CCP5 or CCP6 inhibited cilia formation before ciliogenesis, while shortened the length of cilia after cilia formation. CONCLUSION: These findings reveal the dual role of CCP5 and CCP6. In addition to regulating cilia length, they also retain CP110 level to suppress cilia formation in cycling cells, pointing to a novel regulatory mechanism for ciliogenesis mediated by demodifying enzymes of a conserved ciliary PTM, polyglutamylation.


Subject(s)
Carboxypeptidases , Cilia , Microtubule-Associated Proteins , HEK293 Cells , Humans , Carboxypeptidases/physiology , Microtubule-Associated Proteins/physiology , Cilia/physiology , Microtubules
2.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34333593

ABSTRACT

Peptides derived from proopiomelanocortin (POMC) are well-established neuropeptides and peptide hormones that perform multiple functions, including regulation of body weight. In humans and some animals, these peptides include α- and ß-melanocyte-stimulating hormone (MSH). In certain rodent species, no ß-MSH is produced from POMC because of a change in the cleavage site. Enzymes that convert POMC into MSH include prohormone convertases (PCs), carboxypeptidases (CPs), and peptidyl-α-amidating monooxygenase (PAM). Humans and mice with inactivating mutations in either PC1/3 or carboxypeptidase E (CPE) are obese, which was assumed to result from defective processing of POMC into MSH. However, recent studies have shown that selective loss of either PC1/3 or CPE in POMC-expressing cells does not cause obesity. These findings suggest that defects in POMC processing cannot alone account for the obesity observed in global PC1/3 or CPE mutants. We propose that obesity in animals lacking PC1/3 or CPE activity depends, at least in part, on deficient processing of peptides in non-POMC-expressing cells either in the brain and/or the periphery. Genetic background may also contribute to the manifestation of obesity.


Subject(s)
Carboxypeptidases/physiology , Mixed Function Oxygenases/physiology , Multienzyme Complexes/physiology , Obesity/etiology , Pro-Opiomelanocortin/physiology , Proprotein Convertases/physiology , Animals , Brain/metabolism , Disease Models, Animal , Humans , Mice , Mice, Obese , Obesity/metabolism , Obesity/pathology , Proprotein Convertase 2/physiology
3.
Sci Rep ; 11(1): 3948, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597582

ABSTRACT

The vascular adventitia contains numerous cell types including fibroblasts, adipocytes, inflammatory cells, and progenitors embedded within a complex extracellular matrix (ECM) network. In response to vascular injury, adventitial progenitors and fibroblasts become activated and exhibit increased proliferative capacity and differentiate into contractile cells that remodel the ECM. These processes can lead to vascular fibrosis and disease progression. Our previous work established that the ECM protein aortic carboxypeptidase-like protein (ACLP) promotes fibrotic remodeling in the lung and is activated by vascular injury. It is currently unknown what controls vascular adventitial cell differentiation and if ACLP has a role in this process. Using purified mouse aortic adventitia Sca1+ progenitors, ACLP repressed stem cell markers (CD34, KLF4) and upregulated smooth muscle actin (SMA) and collagen I expression. ACLP enhanced myocardin-related transcription factor A (MRTFA) activity in adventitial cells by promoting MRTFA nuclear translocation. Sca1 cells from MRTFA-null mice exhibited reduced SMA and collagen expression induced by ACLP, indicating Sca1 cell differentiation is regulated in part by the ACLP-MRTFA axis. We determined that ACLP induced vessel contraction and increased adventitial collagen in an explant model. Collectively these studies identified ACLP as a mediator of adventitial cellular differentiation, which may result in pathological vessel remodeling.


Subject(s)
Carboxypeptidases/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Adipocytes/metabolism , Adventitia/metabolism , Animals , Aorta/metabolism , Carboxypeptidases/physiology , Cell Differentiation , Collagen Type I/metabolism , Female , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription Factors/metabolism , Transcriptional Activation
4.
Eur J Neurosci ; 50(9): 3502-3519, 2019 11.
Article in English | MEDLINE | ID: mdl-31309630

ABSTRACT

Neuropeptides are processed from larger preproproteins by a dedicated set of enzymes. The molecular and biochemical mechanisms underlying preproprotein processing and the functional importance of processing enzymes are well-characterised in mammals, but little studied outside this group. In contrast to mammals, Drosophila melanogaster lacks a gene for carboxypeptidase E (CPE), a key enzyme for mammalian peptide processing. By combining peptidomics and neurogenetics, we addressed the role of carboxypeptidase D (dCPD) in global neuropeptide processing and selected peptide-regulated behaviours in Drosophila. We found that a deficiency in dCPD results in C-terminally extended peptides across the peptidome, suggesting that dCPD took over CPE function in the fruit fly. dCPD is widely expressed throughout the nervous system, including peptidergic neurons in the mushroom body and neuroendocrine cells expressing adipokinetic hormone. Conditional hypomorphic mutation in the dCPD-encoding gene silver in the larva causes lethality, and leads to deficits in starvation-induced hyperactivity and appetitive gustatory preference, as well as to reduced viability and activity levels in adults. A phylogenomic analysis suggests that loss of CPE is not common to insects, but only occurred in Hymenoptera and Diptera. Our results show that dCPD is a key enzyme for neuropeptide processing and peptide-regulated behaviour in Drosophila. dCPD thus appears as a suitable target to genetically shut down total neuropeptide production in peptidergic neurons. The persistent occurrence of CPD in insect genomes may point to important further CPD functions beyond neuropeptide processing which cannot be fulfilled by CPE.


Subject(s)
Carboxypeptidases/physiology , Drosophila/physiology , Locomotion/physiology , Protein Processing, Post-Translational/physiology , Survival Rate , Animals , Carboxypeptidases/genetics , Mutation/genetics , Neuropeptides/metabolism , Phylogeny , Protein Processing, Post-Translational/genetics
6.
PLoS One ; 10(4): e0126966, 2015.
Article in English | MEDLINE | ID: mdl-25915901

ABSTRACT

Variable DNA methylation in promoter regions has been implicated in altering transcriptional regulation. The current study analyzed the evolutionary origin and DNA methylation pattern of one of the promoters of Aebp2. According to the results, the first promoter of Aebp2 has been derived from retrotransposons independently in the primate and rodent lineages. DNA methylation analyses revealed that this promoter is unmethylated in sperm, methylated in mature oocytes, and partially methylated at embryonic day 10.5 (78.3%) and 14.5 (58.3%). This promoter also shows variable levels of DNA methylation among adult organs, ranging from the highest in spleen (~80%) to the lowest in tail (~50%). The results from the F1 hybrid of interspecific crossing further indicated that both alleles are equally methylated without any allele bias, also supported by its biallelic expression. Therefore, the partial methylation observed among somatic tissues is an outcome of the genome-wide resetting of DNA methylation during the implantation stage, but not of the inherited allelic methylation pattern preset during gametogenesis. Taken together, mammalian Aebp2 has adopted retrotransposons as its promoter, which displays partial DNA methylation pattern of allelic- or non-allelic origin during the different stages of development.


Subject(s)
Carboxypeptidases/physiology , DNA-Binding Proteins/physiology , Promoter Regions, Genetic/physiology , Repressor Proteins/physiology , Retroelements/physiology , Animals , DNA Methylation/physiology , Embryonic Development/physiology , Gametogenesis/physiology , Humans , Mice
7.
Wiley Interdiscip Rev RNA ; 6(3): 271-89, 2015.
Article in English | MEDLINE | ID: mdl-25630541

ABSTRACT

HIV TAR RNA-binding protein (TRBP) and Protein Activator of PKR (PACT) are double-stranded (ds) RNA-binding proteins that participate in both small regulatory RNA biogenesis and the response to viral dsRNA. Despite considerable progress toward understanding the structure-function relationship of TRBP and PACT, their specific roles in these seemingly distinct cellular pathways remain unclear. Both proteins are composed of three copies of the double-stranded RNA-binding domain, two of which interact with dsRNA, while the C-terminal copy mediates protein-protein interactions. PACT and TRBP are found in a complex with the endonuclease Dicer and facilitate processing of immature microRNAs. Their precise contribution to the Dicing step has not yet been defined: possibilities include precursor recruitment, rearrangement of dsRNA within the complex, loading the processed microRNA into the RNA-induced silencing complex, and distinguishing different classes of small dsRNA. TRBP and PACT also interact with the viral dsRNA sensors retinoic acid-inducible gene I (RIG-I) and double-stranded RNA-activated protein kinase (PKR). Current models suggest that PACT enables RIG-I to detect a wider range of viral dsRNAs, while TRBP and PACT exert opposing regulatory effects on PKR. Here, the evidence that implicates TRBP and PACT in regulatory RNA processing and viral dsRNA sensing is reviewed and discussed in the context of their molecular structure. The broader implications of a link between microRNA biogenesis and the innate antiviral response pathway are also considered.


Subject(s)
RNA, Double-Stranded/metabolism , RNA, Untranslated/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/physiology , Amino Acid Sequence , Carboxypeptidases/chemistry , Carboxypeptidases/metabolism , Carboxypeptidases/physiology , Models, Genetic , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , RNA, Viral/chemistry , RNA, Viral/immunology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Ribonuclease III/physiology , Structure-Activity Relationship
8.
J Biol Chem ; 289(44): 30668-30679, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25231992

ABSTRACT

Francisella tularensis is the etiological agent of tularemia, or rabbit fever. Although F. tularensis is a recognized biothreat agent with broad and expanding geographical range, its mechanism of infection and environmental persistence remain poorly understood. Previously, we identified seven F. tularensis proteins that induce a rapid encystment phenotype (REP) in the free-living amoeba, Acanthamoeba castellanii. Encystment is essential to the pathogen's long term intracellular survival in the amoeba. Here, we characterize the cellular and molecular function of REP34, a REP protein with a mass of 34 kDa. A REP34 knock-out strain of F. tularensis has a reduced ability to both induce encystment in A. castellanii and invade human macrophages. We determined the crystal structure of REP34 to 2.05-Å resolution and demonstrate robust carboxypeptidase B-like activity for the enzyme. REP34 is a zinc-containing monomeric protein with close structural homology to the metallocarboxypeptidase family of peptidases. REP34 possesses a novel topology and substrate binding pocket that deviates from the canonical funnelin structure of carboxypeptidases, putatively resulting in a catalytic role for a conserved tyrosine and distinct S1' recognition site. Taken together, these results identify REP34 as an active carboxypeptidase, implicate the enzyme as a potential key F. tularensis effector protein, and may help elucidate a mechanistic understanding of F. tularensis infection of phagocytic cells.


Subject(s)
Bacterial Proteins/chemistry , Carboxypeptidases/chemistry , Francisella tularensis/physiology , Amino Acid Sequence , Bacterial Proteins/physiology , Carboxypeptidases/physiology , Catalytic Domain , Cell Line , Host-Pathogen Interactions , Humans , Models, Molecular , Molecular Sequence Data , Monocytes/microbiology , Protein Structure, Secondary , Structural Homology, Protein , X-Ray Diffraction
9.
Mol Biol Cell ; 25(12): 1836-44, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24743595

ABSTRACT

Glutamylation is a functionally important tubulin posttranslational modification enriched on stable microtubules of neuronal axons, mitotic spindles, centrioles, and cilia. In vertebrates, balanced activities of tubulin glutamyl ligase and cytoplasmic carboxypeptidase deglutamylase enzymes maintain organelle- and cell type-specific tubulin glutamylation patterns. Tubulin glutamylation in cilia is regulated via restricted subcellular localization or expression of tubulin glutamyl ligases (ttlls) and nonenzymatic proteins, including the zebrafish TPR repeat protein Fleer/Ift70. Here we analyze the expression patterns of ccp deglutamylase genes during zebrafish development and the effects of ccp gene knockdown on cilia formation, morphology, and tubulin glutamylation. The deglutamylases ccp2, ccp5, and ccp6 are expressed in ciliated cells, whereas ccp1 expression is restricted to the nervous system. Only ccp5 knockdown increases cilia tubulin glutamylation, induces ciliopathy phenotypes, including axis curvature, hydrocephalus, and pronephric cysts, and disrupts multicilia motility, suggesting that Ccp5 is the principal tubulin deglutamylase that maintains functional levels of cilia tubulin glutamylation. The ability of ccp5 knockdown to restore cilia tubulin glutamylation in fleer/ift70 mutants and rescue pronephric multicilia formation in both fleer- and ift88-deficient zebrafish indicates that tubulin glutamylation is a key driver of ciliogenesis.


Subject(s)
Carboxypeptidases/physiology , Cilia/physiology , Glutamic Acid/metabolism , Tubulin/metabolism , Zebrafish Proteins/physiology , Animals , Gene Knockdown Techniques , Larva/cytology , Larva/enzymology , Microtubules/metabolism , Pronephros/cytology , Pronephros/enzymology , Protein Processing, Post-Translational , Zebrafish
10.
Genome Biol Evol ; 5(12): 2540-8, 2013.
Article in English | MEDLINE | ID: mdl-24336424

ABSTRACT

Plastids sequestered by sacoglossan sea slugs have long been a puzzle. Some sacoglossans feed on siphonaceous algae and can retain the plastids in the cytosol of their digestive gland cells. There, the stolen plastids (kleptoplasts) can remain photosynthetically active in some cases for months. Kleptoplast longevity itself challenges current paradigms concerning photosystem turnover, because kleptoplast photosystems remain active in the absence of nuclear algal genes. In higher plants, nuclear genes are essential for plastid maintenance, in particular, for the constant repair of the D1 protein of photosystem II. Lateral gene transfer was long suspected to underpin slug kleptoplast longevity, but recent transcriptomic and genomic analyses show that no algal nuclear genes are expressed from the slug nucleus. Kleptoplast genomes themselves, however, appear expressed in the sequestered state. Here we present sequence data for the chloroplast genome of Acetabularia acetabulum, the food source of the sacoglossan Elysia timida, which can maintain Acetabularia kleptoplasts in an active state for months. The data reveal what might be the key to sacoglossan kleptoplast longevity: plastids that remain photosynthetically active within slugs for periods of months share the property of encoding ftsH, a D1 quality control protease that is essential for photosystem II repair. In land plants, ftsH is always nuclear encoded, it was transferred to the nucleus from the plastid genome when Charophyta and Embryophyta split. A replenishable supply of ftsH could, in principle, rescue kleptoplasts from D1 photodamage, thereby influencing plastid longevity in sacoglossan slugs.


Subject(s)
Acetabularia/genetics , Carboxypeptidases/physiology , Gastropoda/physiology , Plant Proteins/physiology , Plastids/genetics , Proprotein Convertases/physiology , Algal Proteins , Animals , Carboxypeptidases/genetics , Chloroplasts/genetics , Genome, Plastid , Plant Proteins/genetics , Proprotein Convertases/genetics
11.
Blood ; 122(8): 1337-8, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23970356

ABSTRACT

In this issue of Blood, Adams et al provide evidence for an important novel function of prolylcarboxypeptidase (PRCP), or angiotensinase C, in endothelial cell physiology and angiogenesis.


Subject(s)
Carboxypeptidases/physiology , Endothelial Cells/enzymology , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Humans
12.
Blood ; 122(8): 1522-31, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23744584

ABSTRACT

Prolylcarboxypeptidase (PRCP) is associated with leanness, hypertension, and thrombosis. PRCP-depleted mice have injured vessels with reduced Kruppel-like factor (KLF)2, KLF4, endothelial nitric oxide synthase (eNOS), and thrombomodulin. Does PRCP influence vessel growth, angiogenesis, and injury repair? PRCP depletion reduced endothelial cell growth, whereas transfection of hPRCP cDNA enhanced cell proliferation. Transfection of hPRCP cDNA, or an active site mutant (hPRCPmut) rescued reduced cell growth after PRCP siRNA knockdown. PRCP-depleted cells migrated less on scratch assay and murine PRCP(gt/gt) aortic segments had reduced sprouting. Matrigel plugs in PRCP(gt/gt) mice had reduced hemoglobin content and angiogenic capillaries by platelet endothelial cell adhesion molecule (PECAM) and NG2 immunohistochemistry. Skin wounds on PRCP(gt/gt) mice had delayed closure and reepithelialization with reduced PECAM staining, but increased macrophage infiltration. After limb ischemia, PRCP(gt/gt) mice also had reduced reperfusion of the femoral artery and angiogenesis. On femoral artery wire injury, PRCP(gt/gt) mice had increased neointimal formation, CD45 staining, and Ki-67 expression. Alternatively, combined PRCP(gt/gt) and MRP-14(-/-) mice were protected from wire injury with less neointimal thickening, leukocyte infiltration, and cellular proliferation. PRCP regulates cell growth, angiogenesis, and the response to vascular injury. Combined with its known roles in blood pressure and thrombosis control, PRCP is positioned as a key regulator of vascular homeostasis.


Subject(s)
Carboxypeptidases/physiology , Endothelial Cells/enzymology , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Aorta/metabolism , Apoptosis , Calgranulin B/metabolism , Cattle , Cell Movement , Cell Proliferation , Cells, Cultured , Femoral Artery/pathology , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/pathology , Ki-67 Antigen/metabolism , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Wound Healing
13.
Plant J ; 75(5): 711-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23662629

ABSTRACT

Plant proteases and protease inhibitors are involved in plant developmental processes including those involving interactions with microbes. Here we show that a tandem between a Kunitz protease inhibitor (KPI106) and a serine carboxypeptidase (SCP1) controls arbuscular mycorrhiza development in the root cortex of Medicago truncatula. Both proteins are only induced during mycorrhiza formation and belong to large families whose members are also mycorrhiza-specific. Furthermore, the interaction between KPI106 and SCP1 analysed using the yeast two-hybrid system is specific, indicating that each family member might have a defined counterpart. In silico docking analysis predicted a putative P1 residue in KPI106 (Lys173) that fits into the catalytic pocket of SCP1, suggesting that KPI106 might inhibit the enzyme activity by mimicking the protease substrate. In vitro mutagenesis of the Lys173 showed that this residue is important in determining the strength and specificity of the interaction. The RNA interference (RNAi) inactivation of the serine carboxypeptidase SCP1 produces aberrant mycorrhizal development with an increased number of septated hyphae and degenerate arbuscules, a phenotype also observed when overexpressing KPI106. Protease and inhibitor are both secreted as observed when expressed in Nicotiana benthamiana epidermal cells. Taken together we envisage a model in which the protease SCP1 is secreted in the apoplast where it produces a peptide signal critical for proper fungal development within the root. KPI106 also at the apoplast would modulate the spatial and/or temporal activity of SCP1 by competing with the protease substrate.


Subject(s)
Carboxypeptidases/physiology , Medicago truncatula/microbiology , Mycorrhizae/enzymology , Peptides/physiology , Plant Proteins/physiology , Amino Acid Sequence , Binding Sites , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/genetics , Medicago truncatula/enzymology , Models, Molecular , Molecular Sequence Data , Mycorrhizae/genetics , Mycorrhizae/physiology , Peptides/genetics , Plant Proteins/genetics , Protein Structure, Tertiary , RNA Interference , Sequence Alignment
14.
Biochem J ; 448(3): 329-41, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22970852

ABSTRACT

The murein peptide amidase MpaA is a cytoplasmic enzyme that processes peptides derived from the turnover of murein. We have purified the enzyme from Escherichia coli and demonstrated that it efficiently hydrolyses the γ-D-glutamyl-diaminopimelic acid bond in the murein tripeptide (L-Ala-γ-D-Glu-meso-Dap), with Km and kcat values of 0.41±0.05 mM and 38.3±10 s-1. However, it is unable to act on the murein tetrapeptide (L-Ala-γ-D-Glu-meso-Dap-D-Ala). E. coli MpaA is a homodimer containing one bound zinc ion per chain, as judged by mass spectrometric analysis and size-exclusion chromatography. To investigate the structure of MpaA we solved the crystal structure of the orthologous protein from Vibrio harveyi to 2.17 Å (1Å=0.1 nm). Vh_MpaA, which has identical enzymatic and biophysical properties to the E. coli enzyme, has high structural similarity to eukaryotic zinc carboxypeptidases. The structure confirms that MpaA is a dimeric zinc metalloprotein. Comparison of the structure of MpaA with those of other carboxypeptidases reveals additional structure that partially occludes the substrate-binding groove, perhaps explaining the narrower substrate specificity of the enzyme compared with other zinc carboxypeptidases. In γ-proteobacteria mpaA is often located adjacent to mppA which encodes a periplasmic transporter protein previously shown to bind murein tripeptide. We demonstrate that MppA can also bind murein tetrapeptide with high affinity. The genetic coupling of these genes and their related biochemical functions suggest that MpaA amidase and MppA transporter form part of a catabolic pathway for utilization of murein-derived peptides that operates in γ-proteobacteria in addition to the established murein recycling pathways.


Subject(s)
Carboxypeptidases/chemistry , Carboxypeptidases/physiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/physiology , Gammaproteobacteria/chemistry , Gammaproteobacteria/physiology , Peptidoglycan/chemistry , Signal Transduction/physiology , Crystallography, X-Ray , Metabolism/physiology , Metalloproteins/chemistry , Metalloproteins/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Peptidoglycan/metabolism , Protein Multimerization , Zinc/chemistry
15.
J Biol Chem ; 287(46): 39171-81, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-22995915

ABSTRACT

Disruption of mammary stromal-epithelial communication leads to aberrant mammary gland development and induces mammary tumorigenesis. Macrophages have been implicated in carcinogenesis primarily by creating an inflammatory microenvironment, which promotes growth of the adjacent epithelial cells. Adipocyte enhancer-binding protein 1 (AEBP1), a novel proinflammatory mediator, promotes macrophage inflammatory responsiveness by inducing NF-κB activity, which has been implicated in tumor cell growth and survival by aberrant sonic hedgehog (Shh) expression. Here, we show that stromal macrophage AEBP1 overexpression results in precocious alveologenesis in the virgin AEBP1 transgenic (AEBP1(TG)) mice, and the onset of ductal hyperplasia was accelerated in AEBP1(TG) mice fed a high fat diet, which induces endogenous AEBP1 expression. Transplantation of AEBP1(TG) bone marrow cells into non-transgenic (AEBP1(NT)) mice resulted in alveolar hyperplasia with up-regulation of NF-κB activity and TNFα expression as displayed in the AEBP1(TG) mammary macrophages and epithelium. Shh expression was induced in AEBP1(TG) macrophages and RAW264.7 macrophages overexpressing AEBP1. The Shh target genes Gli1 and Bmi1 expression was induced in the AEBP1(TG) mammary epithelium and HC11 mammary epithelial cells co-cultured with AEBP1(TG) peritoneal macrophages. The conditioned AEBP1(TG) macrophage culture media promoted NF-κB activity and survival signal, Akt activation, in HC11 cells, whereas such effects were abolished by TNFα neutralizing antibody treatment. Furthermore, HC11 cells displayed enhanced proliferation in response to AEBP1(TG) macrophages and their conditioned media. Our findings highlight the role of AEBP1 in the signaling pathways regulating the cross-talk between mammary epithelium and stroma that could predispose the mammary tissue to tumorigenesis.


Subject(s)
Carboxypeptidases/genetics , Carboxypeptidases/physiology , Hedgehog Proteins/metabolism , Mammary Glands, Animal/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Bone Marrow Transplantation , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Hyperplasia , Inflammation , Macrophages/metabolism , Mammary Glands, Animal/pathology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Physiol Endocrinol Metab ; 302(12): E1502-10, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22454290

ABSTRACT

α-Melanocyte-stimulating hormone (α-MSH) is a critical regulator of energy metabolism. Prolyl carboxypeptidase (PRCP) is an enzyme responsible for its degradation and inactivation. PRCP-null mice (PRCP(gt/gt)) showed elevated levels of brain α-MSH, reduced food intake, and a leaner phenotype compared with wild-type controls. In addition, they were protected against diet-induced obesity. Here, we show that PRCP(gt/gt) animals have improved metabolic parameters compared with wild-type controls under a standard chow diet (SD) as well as on a high-fat diet (HFD). Similarly to when they are exposed to SD, PRCP(gt/gt) mice exposed to HFD for 13 wk showed a leaner phenotype due to decreased fat mass, increased energy expenditure, and locomotor activity. They also showed improved insulin sensitivity and glucose tolerance compared with WT controls and a significant reduction in fasting glucose levels. These improvements occured before changes in body weight and composition were evident, suggesting that the beneficial effect of PRCP ablation is independent of the adiposity levels. In support of a reduced gluconeogenesis, liver PEPCK and G-6-Pase mRNA levels were reduced significantly in PRCP(gt/gt) compared with WT mice. A significant decrease in liver weight and hepatic triglycerides were also observed in PRCP(gt/gt) compared with WT mice. Altogether, our data suggest that PRCP is an important regulator of energy and glucose homeostasis since its deletion significantly improves metabolic parameters in mice exposed to both SD and HFD.


Subject(s)
Carboxypeptidases/genetics , Carboxypeptidases/physiology , Diet, High-Fat/adverse effects , Obesity/metabolism , Animals , Body Composition/physiology , Body Weight/physiology , Energy Metabolism/physiology , Fatty Acids/metabolism , Gene Deletion , Gluconeogenesis/genetics , Gluconeogenesis/physiology , Glucose/pharmacology , Glucose Tolerance Test , Homeostasis/physiology , Hormones/blood , Leptin/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/physiology , Real-Time Polymerase Chain Reaction
18.
PLoS One ; 6(11): e27795, 2011.
Article in English | MEDLINE | ID: mdl-22114697

ABSTRACT

Adipocyte enhancer binding protein 1 (AEBP1) is a multifunctional protein that negatively regulates the tumor suppressor PTEN and IκBα, the inhibitor of NF-κB, through protein-protein interaction, thereby promoting cell survival and inflammation. Mice homozygous for a disrupted AEBP1 gene developed to term but showed defects in growth after birth. AEBP1(-/-) females display lactation defect, which results in the death of 100% of the litters nursed by AEBP1(-/-) dams. Mammary gland development during pregnancy appears normal in AEBP1(-/-) dams; however these mice exhibit expansion of the luminal space and the appearance of large cytoplasmic lipid droplets (CLDs) in the mammary epithelial cells at late pregnancy and parturition, which is a clear sign of failed secretory activation, and accumulation of milk proteins in the mammary gland, presumably reflecting milk stasis following failed secretory activation. Eventually, AEBP1(-/-) mammary gland rapidly undergoes involution at postpartum. Stromal restoration of AEBP1 expression by transplanting wild-type bone marrow (BM) cells is sufficient to rescue the mammary gland defect. Our studies suggest that AEBP1 is critical in the maintenance of normal tissue architecture and function of the mammary gland tissue and controls stromal-epithelial crosstalk in mammary gland development.


Subject(s)
Carboxypeptidases/physiology , Lactation/physiology , Mammary Glands, Animal/metabolism , Milk/chemistry , Repressor Proteins/physiology , Animals , Blotting, Western , Bone Marrow Transplantation , Epithelial Cells/cytology , Female , Mammary Glands, Animal/growth & development , Mice , Mice, Knockout , Pregnancy
19.
Blood ; 117(14): 3929-37, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21297000

ABSTRACT

Prolylcarboxypeptidase (PRCP) activates prekallikrein to plasma kallikrein, leading to bradykinin liberation, and degrades angiotensin II. We now identify PRCP as a regulator of blood vessel homeostasis. ß-Galactosidase staining in PRCP(gt/gt) mice reveals expression in kidney and vasculature. Invasive telemetric monitorings show that PRCP(gt/gt) mice have significantly elevated blood pressure. PRCP(gt/gt) mice demonstrate shorter carotid artery occlusion times in 2 models, and their plasmas have increased thrombin generation times. Pharmacologic inhibition of PRCP with Z-Pro-Prolinal or plasma kallikrein with soybean trypsin inhibitor, Pro-Phe-Arg-chloromethylketone or PKSI 527 also shortens carotid artery occlusion times. Aortic and renal tissues have uncoupled eNOS and increased reactive oxygen species (ROS) in PRCP(gt/gt) mice as detected by dihydroethidium or Amplex Red fluorescence or lucigenin luminescence. The importance of ROS is evidenced by the fact that treatment of PRCP(gt/gt) mice with antioxidants (mitoTEMPO, apocynin, Tempol) abrogates the hypertensive, prothrombotic phenotype. Mechanistically, our studies reveal that PRCP(gt/gt) aortas express reduced levels of Kruppel-like factors 2 and 4, thrombomodulin, and eNOS mRNA, suggesting endothelial cell dysfunction. Further, PRCP siRNA treatment of endothelial cells shows increased ROS and uncoupled eNOS and decreased protein C activation because of thrombomodulin inactivation. Collectively, our studies identify PRCP as a novel regulator of vascular ROS and homeostasis.


Subject(s)
Carboxypeptidases/genetics , Carotid Artery Thrombosis/genetics , Hypertension/genetics , RNA Interference/physiology , Vascular Diseases/genetics , Animals , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/physiology , Carotid Artery Thrombosis/complications , Cells, Cultured , Gene Knockdown Techniques , Humans , Hypertension/complications , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering/pharmacology , Thrombin Time , Time Factors , Vascular Diseases/complications , Vascular Diseases/physiopathology
20.
Cancer Res ; 71(4): 1219-28, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21303978

ABSTRACT

Even though it is among the most commonly methylated loci in multiple cancers, the retinoic acid-induced tumor suppressor retinoic acid receptor responder 1 (RARRES1) has no known function. We now show that RARRES1 is lost in many cancer cells, particularly those with a mesenchymal phenotype, and is a transmembrane carboxypeptidase inhibitor that interacts with ATP/GTP binding protein-like 2 (AGBL2), a cytoplasmic carboxypeptidase. Knockdown of AGBL2 results in a failure of the cell to detyrosinate the C-terminal EEY region of α-tubulin and indicates that it is a candidate for the long sought-after tubulin tyrosine carboxypeptidase important in the regulation of microtubule dynamics. In contrast, knockdown of RARRES1 increases the level of detyrosinated α-tubulin consistent with a role as the cognate inhibitor of AGBL2. We conclude that RARRES1, its interacting partners AGBL2, Eg5/KIF11, another EEY-bearing protein (EB1), and the microtubule tyrosination cycle are important in tumorigenesis and identify a novel area for therapeutic intervention.


Subject(s)
Carboxypeptidases/metabolism , Membrane Proteins/metabolism , Tubulin/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Carboxypeptidases/physiology , Cells, Cultured , Cytoplasm/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Binding/genetics , Protein Binding/physiology , Protein Conformation , Protein Processing, Post-Translational/genetics , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...