Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
J Mol Med (Berl) ; 95(5): 473-486, 2017 05.
Article in English | MEDLINE | ID: mdl-28160049

ABSTRACT

Prolylcarboxypeptidase (PRCP) is a carboxypeptidase that cleaves angiotensin II (AngII) forming Ang(1-7). The impact of genetic PRCP deficiency on AngII metabolism, blood pressure (BP), kidney histology, and cardiac phenotype was investigated in two lines of PRCP-deficient mice: KST302 derived in C57BL/6 background and GST090 derived in FVB/N background. The GST090 line had increased mean arterial pressure (MAP) (113.7 ± 2.07 vs. WT 105.0 ± 1.23 mmHg; p < 0.01) and left ventricular hypertrophy (LVH) (ratio of diastolic left ventricular posterior wall dimension to left ventricular diameter 0.239 ± 0.0163 vs. WT 0.193 ± 0.0049; p < 0.05). Mice in the KST302 line also had mild hypertension and LVH. Cardiac defects, increased glomerular size, and glomerular mesangial expansion were also observed. After infusion of AngII to mice in the KST302 line, both MAP and LVH increased, but the constitutive differences between the gene trap mice and controls were no longer observed. Plasma and cardiac AngII and Ang(1-7) were not significantly different between PRCP-deficient mice and controls. Thus, PRCP deficiency is associated with elevated blood pressure and cardiac alterations including LVH and cardiac defects independently of systemic or cardiac AngII and Ang(1-7). An ex vivo assay showed that recombinant PRCP, unlike recombinant ACE2, did not degrade AngII to form Ang(1-7) in plasma at pH 7.4. PRCP was localized in α-intercalated cells of the kidney collecting tubule. The low pH prevailing at this site and the acidic pH preference of PRCP suggest a role of this enzyme in regulating AngII degradation in the collecting tubule where this peptide increases sodium reabsorption and therfore BP. However, there are other potential mechanisms for increased BP in this model that need to be considered as well. PRCP converts AngII to Ang(1-7) but only at an acidic pH. Global PRCP deficiency causes heart and kidney alterations and a moderate rise in BP. PRCP is abundant in the kidney collecting tubules, where the prevailing pH is low. In collecting tubules, PRCP deficiency could result in impaired AngII degradation. Increased AngII at this nephron site stimulates Na reabsorption and increases BP. KEY MESSAGE: Prolylcarboxypeptidase (PRCP) converts AngII to Ang (1-7) but only at an acidic pH. Global PRCP deficiency causes heart and kidney alterations and a moderate rise in BP. PRCP is abundant in the kidney collecting tubules, where the prevailing pH is low. In collecting tubules, PRCP deficiency could result in impaired AngII degradation. Increased AngII at this nephron site stimulates Na reabsorption and increases BP.


Subject(s)
Angiotensin II/metabolism , Angiotensin I/metabolism , Blood Pressure/physiology , Carboxypeptidases/metabolism , Peptide Fragments/metabolism , Angiotensin II/blood , Animals , Blood Pressure/genetics , Carboxypeptidases/deficiency , Carboxypeptidases/genetics , Kidney/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules, Collecting/metabolism , Male , Mice , Mice, Mutant Strains , Myocardium/metabolism
2.
Cancer Res ; 75(19): 4235-43, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26238780

ABSTRACT

Thrombin-mediated proteolysis is a major determinant of metastasis, but is not universally important for primary tumor growth. Here, we report that colorectal adenocarcinoma represents one important exception whereby thrombin-mediated functions support both primary tumor growth and metastasis. In contrast with studies of multiple nongastrointestinal cancers, we found that the growth of primary tumors formed by murine and human colon cancer cells was reduced in mice by genetic or pharmacologic reduction of circulating prothrombin. Reduced prothrombin expression was associated with lower mitotic indices and invasion of surrounding tissue. Mechanistic investigations revealed that thrombin-driven colonic adenocarcinoma growth relied upon at least two targets of thrombin-mediated proteolysis, protease-activated receptor-1 (PAR-1) expressed by stromal cells and the extracellular matrix protein, fibrinogen. Colonic adenocarcinoma growth was reduced in PAR-1-deficient mice, implicating stromal cell-associated PAR-1 as one thrombin target important for tumor outgrowth. Furthermore, tumor growth was dramatically impeded in fibrinogen-deficient mice, offering the first direct evidence of a critical functional role for fibrinogen in malignant tumor growth. Tumors harvested from fibrinogen-deficient mice displayed a relative reduction in cell proliferative indices, as well as increased tumor necrosis and decreased tumor vascular density. Collectively, our findings established a functional role for thrombin and its targets PAR-1 and fibrinogen in the pathogenesis of colonic adenocarcinoma, supporting tumor growth as well as local invasion and metastasis.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Fibrinogen/physiology , Receptor, PAR-1/physiology , Thrombin/physiology , Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Afibrinogenemia/complications , Afibrinogenemia/genetics , Animals , Cell Division , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Disease Progression , Female , HCT116 Cells/transplantation , Heterografts , Humans , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/physiopathology , Prothrombin/analysis , Receptor, PAR-1/deficiency , Stromal Cells/metabolism , Thrombin/deficiency , Tumor Burden , Tumor Microenvironment
3.
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
4.
Blood ; 121(15): 3023-32, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23386129

ABSTRACT

Bradykinin B2 receptor-deleted mice (Bdkrb2(-/-)) have delayed carotid artery thrombosis times and prolonged tail bleeding time resulting from elevated angiotensin II (AngII) and angiotensin receptor 2 (AT2R) producing increased plasma nitric oxide (NO) and prostacyclin. Bdkrb2(-/-) also have elevated plasma angiotensin-(1-7) and messenger RNA and protein for its receptor Mas. Blockade of Mas with its antagonist A-779 in Bdkrb2(-/-) shortens thrombosis times (58 ± 4 minutes to 38 ± 4 minutes) and bleeding times (170 ± 13 seconds to 88 ± 8 seconds) and lowers plasma nitrate (22 ± 4 µM to 15 ± 5 µM), and 6-keto-PGF1α (259 ± 103 pg/mL to 132 ± 58 pg/mL). Bdkrb2(-/-) platelets express increased NO, guanosine 3',5'-cyclic monophosphate, and cyclic adenosine monophosphate with reduced spreading on collagen, collagen peptide GFOGER, or fibrinogen. In vivo A-779 or combined L-NAME and nimesulide treatment corrects it. Bdkrb2(-/-) platelets have reduced collagen-related peptide-induced integrin α2bß3 activation and P-selectin expression that are partially corrected by in vivo A-779, nimesulide, or L-NAME. Bone marrow transplantations show that the platelet phenotype and thrombosis time depends on the host rather than donor bone marrow progenitors. Transplantation of wild-type bone marrow into Bdkrb2(-/-) hosts produces platelets with a spreading defect and delayed thrombosis times. In Bdkrb2(-/-), combined AT2R and Mas overexpression produce elevated plasma prostacyclin and NO leading to acquired platelet function defects and thrombosis delay.


Subject(s)
Angiotensin I/blood , Blood Platelets/metabolism , Epoprostenol/blood , Nitric Oxide/blood , Peptide Fragments/blood , Platelet Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins/blood , Receptors, G-Protein-Coupled/blood , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Bleeding Time , Blood Platelets/drug effects , Bone Marrow Transplantation , Cyclic AMP/blood , Cyclic GMP/blood , Immunoblotting , Mice , Mice, 129 Strain , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptor, Angiotensin, Type 2/blood , Receptor, Bradykinin B2/deficiency , Receptor, Bradykinin B2/genetics , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Thrombosis/blood , Time Factors
5.
Int J Clin Exp Pathol ; 5(1): 1-11, 2012.
Article in English | MEDLINE | ID: mdl-22295141

ABSTRACT

VEGF and Angiopoietin (Ang)1 are growth factors that independently improve wound healing outcomes. Using a tet-repressible mouse model coupled with streptozotocin-induced diabetes, we examined wound healing in diabetic and nondiabetic mice engineered to overexpress keratinocyte-specific (K5) VEGF, Ang1 or Ang1-VEGF combined. All nondiabetic mice healed more rapidly than their diabetic counterparts; however overexpression of VEGF, Ang1 or the combination failed to improve wound closure under diabetic conditions. Conversely, under nondiabetic conditions, combining Ang1 and VEGF resulted in rapid wound closure. Molecular analyses of diabetic and nondiabetic K5-Ang1-VEGF skin revealed no differences in VEGF expression but an 80% decrease in Ang1 under diabetic conditions, suggesting an integral role for Ang1. Nondiabetic K5-Ang1 mice healed more quickly and had significant increases in granulation tissue and a 60% decrease in re-epithelialization 7 days after wounding. Furthermore, Ang1 stimulated primary mouse keratinocytes showed significantly less migration into a wound bed in an in vitro wound healing bioassay and had decreased pMAPK, pNFκB, pAkt, and pStat3 signaling. These data suggest that combined Ang1-VEGF overexpression cannot overcome diabetes-induced delays in wound healing but is efficacious under nondiabetic conditions possibly via Ang1-mediated delays in re-epithelialization and enhancement of granulation tissue formation, thereby allowing more rapid secondary intention healing.


Subject(s)
Angiopoietin-1/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Vascular Endothelial Growth Factor A/biosynthesis , Wound Healing/physiology , Angiopoietin-1/genetics , Animals , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Enzyme-Linked Immunosorbent Assay , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Mice, Transgenic , Neovascularization, Physiologic/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transgenes , Vascular Endothelial Growth Factor A/genetics
7.
Mol Cell Biol ; 32(7): 1202-13, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22269951

ABSTRACT

The induction of proinflammatory proteins in stimulated endothelial cells (EC) requires activation of multiple transcription programs. The homeobox transcription factor HOXA9 has an important regulatory role in cytokine induction of the EC-leukocyte adhesion molecules (ELAM) E-selectin and vascular cell adhesion molecule 1 (VCAM-1). However, the mechanism underlying stimulus-dependent activation of HOXA9 is completely unknown. Here, we elucidate the molecular mechanism of HOXA9 activation by tumor necrosis factor alpha (TNF-α) and show an unexpected requirement for arginine methylation by protein arginine methyltransferase 5 (PRMT5). PRMT5 was identified as a TNF-α-dependent binding partner of HOXA9 by mass spectrometry. Small interfering RNA (siRNA)-mediated depletion of PRMT5 abrogated stimulus-dependent HOXA9 methylation with concomitant loss in E-selectin or VCAM-1 induction. Chromatin immunoprecipitation analysis revealed that PRMT5 is recruited to the E-selectin promoter following transient HOXA9 binding to its cognate recognition sequence. PRMT5 induces symmetric dimethylation of Arg140 on HOXA9, an event essential for E-selectin induction. In summary, PRMT5 is a critical coactivator component in a newly defined, HOXA9-containing transcription complex. Moreover, stimulus-dependent methylation of HOXA9 is essential for ELAM expression during the EC inflammatory response.


Subject(s)
E-Selectin/genetics , Gene Expression Regulation , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nuclear Proteins/metabolism , Vascular Cell Adhesion Molecule-1/genetics , E-Selectin/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Methylation , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/immunology
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL