Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 150(7): 1123-1133, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34817877

ABSTRACT

Gene variants that encode pancreatic enzymes with impaired secretion can induce pancreatic acinar endoplasmic reticulum (ER) stress, cellular injury and pancreatitis. The role of such variants in pancreatic cancer risk has received little attention. We compared the prevalence of ER stress-inducing variants in CPA1 and CPB1 in patients with pancreatic ductal adenocarcinoma (PDAC cases), enrolled in the National Familial Pancreas Tumor Registry, to their prevalence in noncancer controls in the Genome Aggregation Database (gnomAD). Variants of unknown significance were expressed and variants with reduced secretion assessed for ER stress induction. In vitro assessments were compared with software predictions of variant function. Protein variant software was used to assess variants found in only one gnomAD control ("n-of-one" variants). A meta-analysis of prior PDAC case/control studies was also performed. Of the 1385 patients with PDAC, 0.65% were found to harbor an ER stress-inducing variant in CPA1 or CPB1, compared to 0.17% of the 64 026 controls (odds ratio [OR]: 3.80 [1.92-7.51], P = .0001). ER stress-inducing variants in the CPA1 gene were identified in 4 of 1385 PDAC cases vs 77 of 64 026 gnomAD controls (OR: 2.4 [0.88-6.58], P = .087), and variants in CPB1 were detected in 5 of 1385 cases vs 33 of 64 026 controls (OR: 7.02 [2.74-18.01], P = .0001). Meta-analysis demonstrated strong associations for pancreatic cancer and ER-stress inducing variants for both CPA1 (OR: 3.65 [1.58-8.39], P < .023) and CPB1 (OR: 9.51 [3.46-26.15], P < .001). Rare variants in CPB1 and CPA1 that induce ER stress are associated with increased odds of developing pancreatic cancer.


Subject(s)
Carboxypeptidase B/genetics , Carboxypeptidases A/genetics , Carcinoma, Pancreatic Ductal/etiology , Endoplasmic Reticulum Stress/physiology , Pancreatic Neoplasms/etiology , Carboxypeptidase B/physiology , Carboxypeptidases A/physiology , Carcinoma, Pancreatic Ductal/genetics , Case-Control Studies , Genetic Predisposition to Disease , Genetic Variation , Humans , Pancreatic Neoplasms/genetics , Risk
2.
Reprod Fertil Dev ; 33(6): 401-409, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33745502

ABSTRACT

Numerous long non-coding (lnc) RNAs are highly enriched or exclusively expressed in the mammalian testis, even in spermatids. Spermatid perinuclear RNA-binding protein (STRBP) can bind many RNAs, and loss of STRBP impairs male fertility. However, the functions of lncRNAs interacting with STRBP are unknown. In this study, the roles of one STRBP-interacting lncRNA, namely predicted gene 31453 (Gm31453), and its potential target gene encoding carboxypeptidase A5 (Cpa5) in spermatogenesis were determined using gene-knockout (KO) mice. Gm31453 and Cpa5 are located adjacent to each other on the same chromosome and are highly expressed in the testis. Gm31453 and Cpa5 are primarily expressed from secondary spermatocytes to elongated spermatids, implying their involvement in spermiogenesis. Although deletion of Gm31453 disturbed the expression of both its target and interacting gene, as indicated by decreased Cpa5 and increased Strbp mRNA levels, both Gm31453- and Cpa5-KO mice showed normal spermatogenesis and fertility, and had no detectable abnormalities in terms of testicular and epididymal development, sperm production morphology or motility, pregnancy rate or litter size. Thus, Gm31453 and Cpa5 are dispensable for spermatogenesis and male fertility in mice. Their involvement in spermatogenesis may be a fine-tuning role, regulating gene expression at the molecular level.


Subject(s)
Carboxypeptidases A/genetics , Fertility/genetics , Microtubule-Associated Proteins/genetics , RNA, Long Noncoding/physiology , RNA-Binding Proteins/genetics , Spermatogenesis/genetics , Animals , Carboxypeptidases A/physiology , Gene Expression , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/physiology , RNA, Long Noncoding/genetics , RNA-Binding Proteins/physiology , Sperm Motility , Spermatozoa/ultrastructure , Testis/metabolism
3.
Int J Exp Pathol ; 100(2): 133-138, 2019 04.
Article in English | MEDLINE | ID: mdl-31058377

ABSTRACT

Carboxypeptidase A4 (CPA4), a member of the metallo-carboxypeptidase family, is overexpressed in liver cancer and is associated with cancer progression. The role of CPA4 in hepatocellular carcinoma (HCC) remains unclear. In this study, we aimed to evaluate the relevance of CPA4 to the proliferation and expression of stem cell characteristics of hepatocellular carcinoma cells. Western blot analysis showed high CPA4 expression in the liver cancer cell line Bel7402 and low expression in HepG2 cells. Knock-down of CPA4 decreased cancer cell proliferation as detected by MTT and clone formation assays. The serum-free culture system revealed that downregulated CPA4 suppressed the sphere formation capacities of tumour cells. However, upregulated CPA4 increased the proliferation and sphere formation capacity. In addition, the protein expression of CD133, ALDH1 and CD44 also increased in cells with upregulated CPA4. In vivo, the overexpression of CPA4 in tumour cells that were subcutaneously injected into nude mice markedly increased the growth of the tumours. These data suggest that CPA4 expression leads to poor prognoses by regulating tumour proliferation and the expression of stem cell characteristics and may therefore serve as a potential therapeutic target of HCC.


Subject(s)
Carboxypeptidases A/physiology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/physiology , Animals , Carboxypeptidases A/deficiency , Carboxypeptidases A/genetics , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , Gene Knockdown Techniques/methods , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/enzymology , Male , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Up-Regulation
4.
Blood ; 119(15): 3585-94, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22368273

ABSTRACT

We used the opportunities afforded by the zebrafish to determine upstream pathways regulating mast cell development in vivo and identify their cellular origin. Colocalization studies demonstrated zebrafish notch receptor expression in cells expressing carboxypeptidase A5 (cpa5), a zebrafish mast cell-specific marker. Inhibition of the Notch pathway resulted in decreased cpa5 expression in mindbomb mutants and wild-type embryos treated with the γ-secretase inhibitor, Compound E. A series of morpholino knockdown studies specifically identified notch1b and gata2 as the critical factors regulating mast cell fate. Moreover, hsp70::GAL4;UAS::nicd1a transgenic embryos overexpressing an activated form of notch1, nicd1a, displayed increased cpa5, gata2, and pu.1 expression. This increase in cpa5 expression could be reversed and reduced below baseline levels in a dose-dependent manner using Compound E. Finally, evidence that cpa5 expression colocalizes with lmo2 in the absence of hematopoietic stem cells revealed that definitive mast cells initially delineate from erythromyeloid progenitors. These studies identify a master role for Notch signaling in vertebrate mast cell development and establish developmental origins of this lineage. Moreover, these findings postulate targeting the Notch pathway as a therapeutic strategy in mast cell diseases.


Subject(s)
Cell Lineage/genetics , Homeodomain Proteins/physiology , Mast Cells/physiology , Nerve Tissue Proteins/physiology , Receptor, Notch1/physiology , Zebrafish Proteins/physiology , Zebrafish/immunology , Animals , Animals, Genetically Modified , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Carboxypeptidases A/physiology , Cell Differentiation/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mast Cells/metabolism , Morpholinos/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
J Clin Invest ; 121(10): 4180-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926462

ABSTRACT

Mast cell degranulation is important in the pathogenesis of anaphylaxis and allergic disorders. Many animal venoms contain components that can induce mast cell degranulation, and this has been thought to contribute to the pathology and mortality caused by envenomation. However, we recently reported evidence that mast cells can enhance the resistance of mice to the venoms of certain snakes and that mouse mast cell-derived carboxypeptidase A3 (CPA3) can contribute to this effect. Here, we investigated whether mast cells can enhance resistance to the venom of the Gila monster, a toxic component of that venom (helodermin), and the structurally similar mammalian peptide, vasoactive intestinal polypeptide (VIP). Using 2 types of mast cell-deficient mice, as well as mice selectively lacking CPA3 activity or the chymase mouse mast cell protease-4 (MCPT4), we found that mast cells and MCPT4, which can degrade helodermin, can enhance host resistance to the toxicity of Gila monster venom. Mast cells and MCPT4 also can limit the toxicity associated with high concentrations of VIP and can reduce the morbidity and mortality induced by venoms from 2 species of scorpions. Our findings support the notion that mast cells can enhance innate defense by degradation of diverse animal toxins and that release of MCPT4, in addition to CPA3, can contribute to this mast cell function.


Subject(s)
Carboxypeptidases A/physiology , Mast Cells/enzymology , Scorpion Venoms/toxicity , Serine Endopeptidases/physiology , Vasoactive Intestinal Peptide/toxicity , Venoms/toxicity , Amino Acid Sequence , Animals , Carboxypeptidases A/deficiency , Carboxypeptidases A/genetics , Intercellular Signaling Peptides and Proteins , Lizards , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Molecular Sequence Data , Peptides/antagonists & inhibitors , Peptides/genetics , Peptides/toxicity , Scorpion Venoms/antagonists & inhibitors , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/genetics
6.
Trends Immunol ; 30(8): 401-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19643669

ABSTRACT

When mast cells are activated they can respond by releasing their secretory granule compounds, including mast cell-specific proteases of chymase, tryptase and carboxypeptidase A (MC-CPA) type. MC-CPA is a dominant protein component of the mast cell granule and the MC-CPA gene is extremely highly expressed. Despite this, relatively little has been known of its biological function. However, the recent generation of mouse strains lacking MC-CPA has opened up new possibilities for investigations related to this protease. This recent development has revealed a role for MC-CPA in regulating innate immunity responses, including the degradation of harmful substances such as the vasoconstrictive factor endothelin 1 and snake venom toxins. Here, we summarize the current knowledge of MC-CPA.


Subject(s)
Carboxypeptidases A , Immunity, Innate/physiology , Mast Cells/enzymology , Secretory Vesicles/metabolism , Animals , Carboxypeptidases A/chemistry , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Carboxypeptidases A/physiology , Humans , Mice , Rats , Substrate Specificity
7.
J Immunol ; 183(5): 3463-71, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19641140

ABSTRACT

MCP-1/CCL2 plays a critical role in monocyte recruitment into sites of immune responses and cancer. However, the role of other MCPs remains unclear. In this study, we generated a novel MCP-1-deficient (designated as MCP-1(Delta/Delta)) mouse model by deleting a 2.3-kb DNA fragment from the mouse genome using the Cre/loxP system. MCP-1 was not produced by LPS-activated MCP-1(Delta/Delta) macrophages; however, the production of MCP-3, coded by the immediate downstream gene, was significantly increased. In contrast, macrophages from another mouse line with a neo-gene cassette in intron 2 produced a significantly lower level of MCP-1 and MCP-3. Decreased MCP-3 production was also detected in previously generated MCP-1-deficient mice in which a neo-gene cassette was inserted in exon 2 (designated as MCP-1 knockout (KO)). Altered MCP-1 and/or MCP-3 production was also observed in vivo in each mouse model in response to i.p. injection of thioglycolate or zymosan. The up- and down-regulation of MCP-3 production in MCP-1(Delta/Delta) and MCP-1 KO mice, respectively, provided us with a unique opportunity to evaluate the role for MCP-3. Despite the increased MCP-3 production in MCP-1(Delta/Delta) mice, thioglycolate- or zymosan-induced monocyte/macrophage accumulation was still reduced by approximately 50% compared with wild-type mice, similar to the reduction detected in MCP-1 KO mice. Thus, up-regulated MCP-3 production did not compensate for the loss of MCP-1, and MCP-3 appears to be a less effective mediator of monocyte recruitment than MCP-1. Our results also indicate the presence of other mediators regulating the recruitment of monocytes in these models.


Subject(s)
Carboxypeptidases A/physiology , Cell Movement/immunology , Chemokine CCL2/physiology , Monocytes/cytology , Monocytes/immunology , Peritonitis/immunology , Thioglycolates/pharmacology , Zymosan/pharmacology , Animals , Carboxypeptidases A/antagonists & inhibitors , Carboxypeptidases A/biosynthesis , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Gene Deletion , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Peritonitis/chemically induced , Peritonitis/pathology , Thioglycolates/administration & dosage , Up-Regulation/immunology , Zymosan/administration & dosage
8.
Exp Physiol ; 93(5): 613-21, 2008 May.
Article in English | MEDLINE | ID: mdl-18356559

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2), a homologue of angiotensin-converting enzyme (ACE), converts angiotensin (Ang) I to Ang(1-9) and Ang II to Ang(1-7), but does not directly process Ang I to Ang II. Cardiac function is compromised in ACE2 null mice; however, the importance of ACE2 in the processing of angiotensin peptides within the murine heart is not known. We determined the metabolism of angiotensins in wild-type (WT), ACE (ACE(-/-)) and ACE2 null mice (ACE2(-/-)). Angiotensin II was converted almost exclusively to Ang(1-7) in the cardiac membranes of WT and ACE(-/-) strains, although generation of Ang(1-7) was greater in the ACE(-/-) mice (27.4 +/- 4.1 versus 17.5 +/- 3.2 nmol(-1) mg h(-1) for WT). The ACE2 inhibitor MLN4760 significantly attenuated Ang II metabolism and the subsequent formation of Ang(1-7) in both strains. In the ACE2(-/-) hearts, Ang II metabolism and the generation of Ang(1-7) were significantly attenuated; however, the ACE2 inhibitor reduced the residual Ang(1-7)-forming activity in this strain. Angiotensin I was primarily converted to Ang(1-9) (WT, 28.9 +/- 3.1 nmol(-1) mg h(-1); ACE(-/-), 49.8 +/- 5.3 nmol(-1) mg h(-1); and ACE2(-/-), 35.9 +/- 5.4 nmol(-1) mg h(-1)) and to smaller quantities of Ang(1-7) and Ang II. Although the ACE2 inhibitor had no effect on Ang(1-9) formation, the carboxypeptidase A inhibitor benzylsuccinate essentially abolished the formation of Ang(1-9) and increased the levels of Ang I in cardiac membranes. In conclusion, our studies in the murine heart suggest that ACE2 is the primary pathway for the metabolism of Ang II and the subsequent formation of Ang(1-7), a peptide that, in contrast to Ang II, exhibits both antifibrotic and antiproliferative actions.


Subject(s)
Angiotensins/metabolism , Carboxypeptidases A/physiology , Myocardium/metabolism , Peptidyl-Dipeptidase A/physiology , Angiotensin I/biosynthesis , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Carboxypeptidases A/antagonists & inhibitors , Cell Proliferation/drug effects , Fibrosis/prevention & control , Heart/drug effects , Imidazoles/pharmacology , Immunohistochemistry , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Membranes/enzymology , Mice , Mice, Knockout , Myocardium/enzymology , Peptide Fragments/biosynthesis , Peptidyl-Dipeptidase A/genetics , Protease Inhibitors/pharmacology , Succinates/pharmacology
9.
J Exp Med ; 204(11): 2629-39, 2007 Oct 29.
Article in English | MEDLINE | ID: mdl-17923505

ABSTRACT

Mast cells are protective against snake venom sarafotoxins that belong to the endothelin (ET) peptide family. The molecular mechanism underlying this recently recognized innate defense pathway is unknown, but secretory granule proteases have been invoked. To specifically disrupt a single protease function without affecting expression of other proteases, we have generated a mouse mutant selectively lacking mast cell carboxypeptidase A (Mc-cpa) activity. Using this mutant, we have now identified Mc-cpa as the essential protective mast cell enzyme. Mass spectrometry of peptide substrates after cleavage by normal or mutant mast cells showed that removal of a single amino acid, the C-terminal tryptophan, from ET and sarafotoxin by Mc-cpa is the principle molecular mechanism underlying this very rapid mast cell response. Mast cell proteases can also cleave ET and sarafotoxin internally, but such "nicking" is not protective because intramolecular disulfide bridges maintain peptide function. We conclude that mast cells attack ET and sarafotoxin exactly at the structure required for toxicity, and hence sarafotoxins could not "evade" Mc-cpa's substrate specificity without loss of toxicity.


Subject(s)
Carboxypeptidases A/physiology , Endothelins/pharmacology , Mast Cells/drug effects , Mast Cells/physiology , Viper Venoms/pharmacology , Animals , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Carboxypeptidases A/deficiency , Carboxypeptidases A/genetics , Gene Expression Regulation , Mast Cells/enzymology , Mice , Mice, Knockout , Vasoconstrictor Agents/pharmacology
10.
Mol Cell Biol ; 25(14): 6199-210, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988029

ABSTRACT

Mast cell carboxypeptidase A (Mc-cpa) is a highly conserved secretory granule protease. The onset of expression in mast cell progenitors and lineage specificity suggest an important role for Mc-cpa in mast cells. To address the function of Mc-cpa, we generated Mc-cpa-null mice. Mc-cpa-/- mast cells lacked carboxypeptidase activity, revealing that Mc-cpa is a nonredundant enzyme. While Mc-cpa-/- peritoneal mast cells were ultrastructurally normal and synthesized normal amounts of heparin, they displayed striking histochemical and biochemical hallmarks of immature mast cells. Wild-type peritoneal mast cells had a mature phenotype characterized by differential histochemical staining with proteoglycan-reactive dyes (cells do not stain with alcian blue but stain with safranin and with berberine) and a high side scatter to forward scatter ratio by flow cytometry and were detergent resistant. In contrast, Mc-cpa-/- peritoneal mast cells, like immature bone marrow-derived cultured mast cells, stained with alcian blue normally or weakly and either did not stain with safranin and berberine or stained weakly, had a low side scatter to forward scatter ratio, and were detergent sensitive. This phenotype was partially ameliorated with age. Thus, histochemistry and flow cytometry, commonly used to measure mast cell maturation, deviated from morphology in Mc-cpa-/- mice. The Mc-cpa-/- mast cell phenotype was not associated with defects in degranulation in vitro or passive cutaneous anaphylaxis in vivo. Collectively, Mc-cpa plays a crucial role for the generation of phenotypically mature mast cells.


Subject(s)
Carboxypeptidases A/physiology , Mast Cells/enzymology , Mast Cells/ultrastructure , Animals , Antibodies/immunology , Berberine/pharmacology , Carboxypeptidases A/analysis , Carboxypeptidases A/genetics , Heparin/immunology , Heparin/metabolism , Histocytochemistry , Mast Cells/drug effects , Mice , Mice, Mutant Strains , Monocyte Chemoattractant Proteins/metabolism , Phenotype , Proteoglycans/metabolism , Serine Endopeptidases/metabolism , Tryptases
11.
J Am Chem Soc ; 126(24): 7452-3, 2004 Jun 23.
Article in English | MEDLINE | ID: mdl-15198587

ABSTRACT

A model for the natural enzyme carboxypeptidase A was prepared by molecular imprinting in synthetic polymers. An unusually high activity and efficiency for carbonate hydrolysis could be obtained by imprinting with a stable transition-state analogue template and introducing an amidinium group and a Cu2+ ion-binding site in a defined orientation to each other into the active site. With substrates having a very similar structure to the template, extraordinarily high enhancements of rates of 110 000-fold were obtained of catalyzed to uncatalyzed reaction kcat/kuncat . The efficiency kcat/Km of the molecularly imprinted catalysts compared to that of the nonimprinted control polymers containing the same functional groups was 790-fold higher, a clear indication of a very efficient imprinting procedure.


Subject(s)
Amides/chemistry , Carboxypeptidases A/chemistry , Carboxypeptidases A/physiology , Copper/chemistry , Molecular Mimicry , Binding Sites/physiology , Catalysis , Kinetics , Models, Chemical , Molecular Structure , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...