Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 26(1): 233-242, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28011710

ABSTRACT

Chromogranins are pro-hormone secretory proteins released from neuroendocrine cells, with effects on control of blood pressure. We conducted a genome-wide association study for plasma catestatin, the catecholamine release inhibitory peptide derived from chromogranin A (CHGA), and other CHGA- or chromogranin B (CHGB)-related peptides, in 545 US and 1252 Australian subjects. This identified loci on chromosomes 4q35 and 5q34 affecting catestatin concentration (P = 3.40 × 10-30 for rs4253311 and 1.85 × 10-19 for rs2731672, respectively). Genes in these regions include the proteolytic enzymes kallikrein (KLKB1) and Factor XII (F12). In chromaffin cells, CHGA and KLKB1 proteins co-localized in catecholamine storage granules. In vitro, kallikrein cleaved recombinant human CHGA to catestatin, verified by mass spectrometry. The peptide identified from this digestion (CHGA360-373) selectively inhibited nicotinic cholinergic stimulated catecholamine release from chromaffin cells. A proteolytic cascade involving kallikrein and Factor XII cleaves chromogranins to active compounds both in vivo and in vitro.


Subject(s)
Biomarkers/metabolism , Catecholamines/metabolism , Chromaffin Cells/metabolism , Chromogranin A/blood , Genetic Loci/genetics , Hypertension/genetics , Peptide Fragments/blood , Adolescent , Adrenal Glands/metabolism , Adult , Aged , Animals , Australia , Biomarkers/analysis , Cells, Cultured , Factor XII/genetics , Factor XII/metabolism , Female , Genome-Wide Association Study , Humans , Hypertension/blood , Kallikreins/genetics , Kallikreins/metabolism , Male , Mice , Middle Aged , Rats , United States , Young Adult
2.
Microcirculation ; 26(6): e12548, 2019 08.
Article in English | MEDLINE | ID: mdl-30946505

ABSTRACT

OBJECTIVE: The mechanisms underlying cell and organ dysfunctions in hypertension are uncertain. The spontaneously hypertensive rat (SHR) has elevated levels of unchecked degrading proteases compared to the control Wistar Kyoto (WKY) rat. The extracellular proteases destroy membrane receptors leading to cell dysfunctions, including arteriolar constriction and elevated blood pressure. Our goal was to identify potential sources of the uncontrolled enzymatic activity. METHODS: Zymographic and digital immunohistochemical measurements in SHR pancreas and intestine were obtained as part of the digestive system with high levels of degrading enzymes. OBJECTIVE: The results showed that SHRs have significantly higher protease activity than WKY in pancreas (22.04 ± 9.01 vs 13.02 ± 3.92 casein fluorescence intensity unit; P < 0.05) and pancreatic venules (0.011 ± 0.003 vs 0.005 ± 0.003 trypsin absorbance; P < 0.05) as well as in venous blood (71.07 ± 13.92 vs 36.44 ± 16.59 casein fluorescence intensity unit; P < 0.05). The enzymatic activity is contributed by trypsin and chymotrypsin. Furthermore, a decrease of these enzyme activity levels achieved during a short-term fasting period is associated with a reduction in systolic blood pressurein SHR (135 ± 8 mm Hg vs 124 ± 7 mm Hg; P < 0.05). CONCLUSIONS: The results suggest the pancreas of the SHR is a potential source for serine proteases leaking into the circulation and contributing to its protease activity.


Subject(s)
Caloric Restriction , Chymotrypsin/metabolism , Hypertension/epidemiology , Pancreas/enzymology , Trypsin/metabolism , Animals , Hypertension/pathology , Male , Pancreas/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY
3.
J Am Coll Nutr ; 38(6): 485-492, 2019 08.
Article in English | MEDLINE | ID: mdl-30964398

ABSTRACT

Objective: To examine resting and postprandial peripheral protease activity in healthy controls and individuals with type 2 diabetes mellitus (T2DM) and pre-T2DM. Methods: Individuals with T2DM or pre-T2DM and healthy controls (mean age 55.8 years) were studied before and for a span of 300 minutes following a single high-calorie McDonald's breakfast. Metalloproteases-2/-9 (MMP-2/-9), elastase, and trypsin activities were assessed in whole blood before and following the meal using a novel high-precision electrophoretic platform. Also assessed were circulating levels of inflammatory biomarkers and insulin receptor density on peripheral blood mononuclear cells (PBMCs) in relationship to protease activity. Results: Premeal MMP-2/-9 and elastase activity levels in T2DM and in pre-T2DM participants were significantly elevated as compared to controls. The T2DM group showed a significant increase in elastase activity 15 minutes after the meal; elastase activity continued to increase to the 30-minute time point (p < 0.01). In control participants, MMP-2/-9, elastase, and trypsin were significantly increased at 15 minutes after the meal (p < 0.05) and returned to premeal values within a period of approximately 30 to 60 minutes post meal. PBMCs incubated for 1 hour with plasma from T2DM and pre-T2DM participants had significantly lower levels of insulin receptor density compared to those incubated with plasma from control participants (p < 0.001). Conclusions: The results of this study suggest that individuals with T2DM and pre-T2DM have higher resting systemic protease activity than nonsymptomatic controls. A single high-calorie/high-carbohydrate meal results in further elevations of protease activity in the systemic circulation of T2DM and pre-T2DM, as well as in healthy controls. The protease activity in turn can lead to a downregulation of insulin receptor density, potentially supporting a state of insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2 , Peptide Hydrolases/blood , Postprandial Period/physiology , Receptor, Insulin , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Male , Middle Aged , Receptor, Insulin/blood , Receptor, Insulin/metabolism , Rest/physiology
4.
J Clin Monit Comput ; 31(1): 167-175, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26686691

ABSTRACT

The nature of hemodynamic instability typical of circulatory shock is not well understood, but an improved interpretation of its dynamic features could help in the management of critically ill patients. The objective of this work was to introduce new metrics for the analysis of arterial blood pressure (ABP) in order to characterize the risk of catastrophic outcome in splanchnic arterial occlusion (SAO) shock. Continuous ABP (fs = 1 kHz) was measured in rats during experimental SAO shock, which induced a fatal pressure drop (FPD) in ABP. The FPD could either be slow (SFPD) or fast (FFPD), with the latter causing cardiovascular collapse. Time series of mean arterial pressure, systolic blood pressure and heart period were derived from ABP. The sample asymmetry-based algorithm Heart Rate Characteristics was adapted to compute the Heart Period Characteristics (HPC) and the Blood Pressure Characteristics (BPC). Baroreflex sensitivity (BRS) was assessed by means of a bivariate model. The approach to FPD of the animals who collapsed (FFPD) was characterized by higher BRS in the low frequency band versus SFPD animals (0.36 ± 0.15 vs. 0.19 ± 0.12 ms/mmHg, p value = 0.0196), bradycardia as indicated by the HPC (0.76 ± 0.57 vs. 1.94 ± 1.27, p value = 0.0179) and higher but unstable blood pressure as indicated by BPC (3.02 ± 2.87 vs. 1.47 ± 1.29, p value = 0.0773). The HPC and BPC indices demonstrated promise as potential clinical markers of hemodynamic instability and impending cardiovascular collapse, and this animal study suggests their test in data from intensive care patients.


Subject(s)
Blood Pressure Determination , Blood Pressure , Shock/physiopathology , Splanchnic Circulation , Algorithms , Animals , Baroreflex , Cardiovascular Diseases/physiopathology , Critical Care , Heart/physiopathology , Heart Rate , Hemodynamics , Humans , Intensive Care Units , Male , Models, Statistical , Rats , Rats, Wistar , Risk , Time Factors , Treatment Outcome
5.
Electrophoresis ; 37(15-16): 2248-56, 2016 08.
Article in English | MEDLINE | ID: mdl-27271700

ABSTRACT

Present coagulation assays fail to detect mild coagulation disorders, while thrombin-generation (TG) assays solve this problem. However, most of them only work with threated blood samples, which makes them labor intensive, time consuming, unreliable, and expensive. We have developed a TG electrophoretic assay that uses a thrombin specific charge-changing fluorescent peptide substrate, electrophoretic separation, and requires a drop of blood. The limit of detection of the assay was 1.97 nM in phosphate buffer saline and 6.82 nM in citrated whole blood. The assay was used to determine the TG in whole blood from healthy volunteers (n = 6, one aspirin user), over 30 min, after the blood was drawn; the TG increased from a baseline level of 2 × 10(6) RFU to 1.2 × 10(13) RFU. The lag time between the blood draw and initial burst of TG was 6 min for the volunteers (n = 5) and 15 min for the aspirin user. Specificity of the assay was evaluated by reacting our substrate with the heparinized blood samples and other proteases. The TG electrophoretic assay was designed and tested in the whole human blood, requiring no sample preparation, 5 µL of blood, 45 min, and it detected differences in coagulation patterns between a volunteer taking aspirin and non-aspirin users.


Subject(s)
Blood Coagulation Tests/methods , Electrophoresis/methods , Aspirin/pharmacology , Blood Coagulation/drug effects , Humans , Limit of Detection , Sensitivity and Specificity , Thrombin Time
6.
J Pediatr Gastroenterol Nutr ; 62(2): 317-27, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26230900

ABSTRACT

OBJECTIVES: Parents of children with autism spectrum disorders (ASDs) often report gastrointestinal (GI) dysfunction in their children. The objectives of the present study were to determine whether infants at high risk for developing ASD (ie, siblings of children diagnosed as having ASD) show greater prevalence of GI problems and whether this prevalence is associated with diet and age at weaning from breast milk. METHODS: Using questionnaires, diet history and GI problems were tracked prospectively and retrospectively in 57 high-risk infants and for comparison in 114 low-risk infants (infants from families without ASD history). RESULTS: In low-risk infants, prevalence of GI symptoms, in aggregate, did not vary with diet or age of weaning. By contrast, high-risk infants with GI symptoms were weaned earlier than those without symptoms (P < 0.04), and high-risk infants showed greater prevalence of GI symptoms, in aggregate, on a no breast milk diet than on an exclusive breast milk diet (P < 0.017). Constipation, in particular, was more prevalent in high-risk infants compared with low-risk infants (P = 0.01), especially on a no breast milk diet (P = 0.002). High-risk infants who completed weaning earlier than 6 months showed greater prevalence of constipation (P = 0.001) and abdominal distress (P = 0.004) than those fully weaned after 6 months. CONCLUSIONS: The greater prevalence of GI symptoms in high-risk infants suggests that GI dysfunction during early infant development may be a part of the ASD endophenotype. Late weaning and exclusive breast milk were associated with protection against GI symptoms in high-risk infants.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Breast Feeding , Constipation/prevention & control , Diet , Milk, Human , Weaning , Adult , Autism Spectrum Disorder/complications , Autistic Disorder/complications , Child, Preschool , Constipation/complications , Gastrointestinal Diseases/complications , Gastrointestinal Diseases/prevention & control , Humans , Infant , Middle Aged , Phenotype , Surveys and Questionnaires , Young Adult
7.
J Am Soc Nephrol ; 26(12): 3072-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25855776

ABSTRACT

Diabetic kidney disease (DKD) is a microvascular complication that leads to kidney dysfunction and ESRD, but the underlying mechanisms remain unclear. Podocyte Wnt-pathway activation has been demonstrated to be a trigger mechanism for various proteinuric diseases. Notably, four-and-a-half LIM domains protein 2 (FHL2) is highly expressed in urogenital systems and has been implicated in Wnt/ß-catenin signaling. Here, we used in vitro podocyte culture experiments and a streptozotocin-induced DKD model in FHL2 gene-knockout mice to determine the possible role of FHL2 in DKD and to clarify its association with the Wnt pathway. In human and mouse kidney tissues, FHL2 protein was abundantly expressed in podocytes but not in renal tubular cells. Treatment with high glucose or diabetes-related cytokines, including angiotensin II and TGF-ß1, activated FHL2 protein and Wnt/ß-catenin signaling in cultured podocytes. This activation also upregulated FHL2 expression and promoted FHL2 translocation from cytosol to nucleus. Genetic deletion of the FHL2 gene mitigated the podocyte dedifferentiation caused by activated Wnt/ß-catenin signaling under Wnt-On, but not under Wnt-Off, conditions. Diabetic FHL2(+/+) mice developed markedly increased albuminuria and thickening of the glomerular basement membrane compared with nondiabetic FHL2(+/+) mice. However, FHL2 knockout significantly attenuated these DKD-induced changes. Furthermore, kidney samples from patients with diabetes had a higher degree of FHL2 podocyte nuclear translocation, which was positively associated with albuminuria and progressive renal function deterioration. Therefore, we conclude that FHL2 has both structural and functional protein-protein interactions with ß-catenin in the podocyte nucleus and that FHL2 protein inhibition can mitigate Wnt/ß-catenin-induced podocytopathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Podocytes/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Albuminuria/etiology , Angiotensin II/pharmacology , Animals , Cell Dedifferentiation/genetics , Cells, Cultured , Diabetic Nephropathies/complications , Diabetic Nephropathies/pathology , Gene Knockout Techniques , Glomerular Basement Membrane/pathology , Glucose/pharmacology , Humans , LIM-Homeodomain Proteins/drug effects , LIM-Homeodomain Proteins/genetics , Male , Mice , Muscle Proteins/drug effects , Muscle Proteins/genetics , Podocytes/drug effects , Protein Transport , Transcription Factors/drug effects , Transcription Factors/genetics , Transforming Growth Factor beta1/pharmacology , Wnt Signaling Pathway/drug effects
8.
Hum Mol Genet ; 22(18): 3624-40, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23674521

ABSTRACT

Hypertension is a common hereditary syndrome with unclear pathogenesis. Chromogranin A (Chga), which catalyzes formation and cargo storage of regulated secretory granules in neuroendocrine cells, contributes to blood pressure homeostasis centrally and peripherally. Elevated Chga occurs in spontaneously hypertensive rat (SHR) adrenal glands and plasma, but central expression is unexplored. In this report, we measured SHR and Wistar-Kyoto rat (control) Chga expression in central and peripheral nervous systems, and found Chga protein to be decreased in the SHR brainstem, yet increased in the adrenal and the plasma. By re-sequencing, we systematically identified five promoter, two coding and one 3'-untranslated region (3'-UTR) polymorphism at the SHR (versus WKY or BN) Chga locus. Using HXB/BXH recombinant inbred (RI) strain linkage and correlations, we demonstrated genetic determination of Chga expression in SHR, including a cis-quantitative trait loci (QTLs) (i.e. at the Chga locus), and such expression influenced biochemical determinants of blood pressure, including a cascade of catecholamine biosynthetic enzymes, catecholamines themselves and steroids. Luciferase reporter assays demonstrated that the 3'-UTR polymorphism (which disrupts a microRNA miR-22 motif) and promoter polymorphisms altered gene expression consistent with the decline in SHR central Chga expression. Coding region polymorphisms did not account for changes in Chga expression or function. Thus, we hypothesized that the 3'-UTR and promoter mutations lead to dysregulation (diminution) of Chga in brainstem cardiovascular control nuclei, ultimately contributing to the pathogenesis of hypertension in SHR. Accordingly, we demonstrated that in vivo administration of miR-22 antagomir to SHR causes substantial (∼18 mmHg) reductions in blood pressure, opening a novel therapeutic avenue for hypertension.


Subject(s)
Chromogranin A/genetics , Chromogranin A/metabolism , Hypertension/genetics , MicroRNAs/genetics , Promoter Regions, Genetic , 3' Untranslated Regions , Adrenal Glands/metabolism , Animals , Blood Pressure/genetics , Brain Stem/metabolism , Cell Line, Tumor , Chromogranin A/blood , Chromogranin A/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genetic Linkage , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Male , MicroRNAs/metabolism , PC12 Cells , Polymorphism, Genetic , Protein Structure, Secondary , Quantitative Trait Loci , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sequence Alignment , Transcription, Genetic
9.
J Biol Chem ; 288(1): 598-607, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23155052

ABSTRACT

Matrix metalloproteinase-1 (MMP-1) is a collagenase that is highly active in extracellular matrix and vascular remodeling, angiogenesis, and tumor progression. Vascular endothelial growth factor receptor-2 (VEGFR2), the main receptor for VEGF-A, is expressed on endothelial cells and promotes cell survival, proliferation, and other functions. Although MMP-1 and VEGFR2 co-exist in many normal and pathophysiological conditions, the effect of MMP-1 on cellular VEGFR2 that can promote the above processes is unknown. In this study we test the hypothesis that stimulation of endothelial cells with MMP-1 increases their levels of VEGFR2. The increased VEGFR2 is then available to bind VEGF-A, resulting in increased response. Indeed we found that endothelial cells incubated with active MMP-1 had higher mRNA and protein levels of VEGFR2. Furthermore, VEGF-A-dependent phosphorylation of intracellular signaling molecules and endothelial proliferation were elevated after MMP-1 treatment. MMP-1 caused activation of the nuclear factor-κB (NF-κB) pathway (p65/RelA) in endothelial cells, and this response was dependent upon activation of protease activated receptor-1 (PAR-1). Chromatin immunoprecipitation was used to confirm NF-κB-mediated active transcription of the VEGFR2 (KDR) gene. Elevation in VEGFR2 after MMP-1 stimulation was inhibited by PAR-1 knockdown and NF-κB specific inhibition. We conclude that MMP-1 promotes VEGFR2 expression and proliferation of endothelial cells through stimulation of PAR-1 and activation of NF-κB. These results suggest a mechanism by which MMP-1 may prime or sensitize endothelial cell functions.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gene Expression Regulation, Enzymologic , Matrix Metalloproteinase 1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Animals , Cattle , Cell Proliferation , Endothelial Cells/cytology , Humans , Microscopy, Fluorescence/methods , Models, Biological , NF-kappa B/metabolism , Signal Transduction , Up-Regulation
10.
Microvasc Res ; 95: 131-42, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25172177

ABSTRACT

Metabolic disease is accompanied by a range of cellular defects ("comorbidities") whose origin is uncertain. To investigate this pathophysiological phenomenon we used the Spontaneously Hypertensive Rat (SHR), which besides an elevated arterial blood pressure also has many other comorbidities, including a defective glucose and lipid metabolism. We have shown that this model of metabolic disease has elevated plasma matrix metalloproteinase (MMP) activity, which cleaves the extracellular domain of membrane receptors. We hypothesize here that the increased MMP activity also leads to abnormal cleavage of the scavenger receptor and fatty acid transporter CD36. To test this idea, chronic pharmaceutical MMP inhibition (CGS27023A) of the SHR and its normotensive control, the Wistar Kyoto Rat (WKY), was used to determine if inhibition of MMP activity serves to maintain CD36 receptor density and function. Surface density of CD36 on macrophages from the heart, spleen, and liver was determined in WKY, SHR, CGS-treated WKY (CGS WKY), and CGS-treated SHR (CGS SHR) by immunohistochemistry with an antibody against the CD36 ectodomain. The extracellular CD36 density was lower in SHR heart and spleen macrophages compared to that in the WKY. MMP inhibition by CGS served to restore the reduced CD36 density on SHR cardiac and splanchnic macrophages to levels of the WKY. To examine CD36 function, culture assays with murine macrophages (RAW 264.7) after incubation in fresh WKY or SHR plasma were used to test for adhesion of light-weight donor red blood cell (RBC) by CD36. This form of RBC adhesion to macrophages was reduced after incubation in SHR compared WKY plasma. Analysis of the supernatant macrophage media by Western blot shows a higher level of CD36 extracellular protein fragments following exposure to SHR plasma compared to WKY. MMP inhibition in the SHR plasma compared to untreated plasma, served to increase the RBC adhesion to macrophages and decrease the number of receptor fragments in the macrophage media. In conclusion, these studies bring to light that plasma in the SHR model of metabolic disease has an unchecked MMP degrading activity which causes cleavage of a variety of membrane receptors, including CD36, which attenuates several cellular functions typical for the metabolic disease, including RBC adhesion to the scavenger receptor CD36. In addition to other cell dysfunctions chronic MMP inhibition restores CD36 in the SHR.


Subject(s)
CD36 Antigens/metabolism , Hypertension/enzymology , Macrophages/enzymology , Matrix Metalloproteinases/metabolism , Myocardium/enzymology , Spleen/enzymology , Animals , Arterial Pressure , CD36 Antigens/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation , Erythrocytes/metabolism , Hypertension/immunology , Hypertension/physiopathology , Macrophages/immunology , Male , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardium/immunology , Proteolysis , Rats, Inbred SHR , Rats, Inbred WKY , Spleen/drug effects , Spleen/immunology
11.
J Pediatr Gastroenterol Nutr ; 59(3): 365-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24840512

ABSTRACT

OBJECTIVES: Fat is digested in the intestine into free fatty acids (FFAs), which are detergents and therefore toxic to cells at micromolar concentration. The mucosal barrier protects cells in the adult intestine, but this barrier may not be fully developed in premature infants. Lipase-digested infant formula, but not fresh human milk, has elevated FFAs and is cytotoxic to intestinal cells, and therefore could contribute to intestinal injury in necrotizing enterocolitis (NEC), but even infants exclusively fed breast milk may develop NEC. Our objective was to determine whether stored milk and milk from donor milk (DM) banks could also become cytotoxic, especially after digestion. METHODS: We exposed cultured rat intestinal epithelial cells or human neutrophils to DM and milk collected fresh and stored at 4°C or -20°C for up to 12 weeks and then treated for 2 hours (37°C) with 0.1 or 1 mg/mL pancreatic lipase and/or trypsin and chymotrypsin. RESULTS: DM and milk stored 3 days (at 4°C or -20°C) and then digested were cytotoxic. Storage at -20°C for 8 and 12 weeks resulted in an additional increase in cytotoxicity. Protease digestion decreased, but did not eliminate cell death. CONCLUSIONS: Present storage practices may allow milk to become cytotoxic and contribute to intestinal damage in NEC.


Subject(s)
Digestion , Fatty Acids, Nonesterified/metabolism , Food Storage , Lipase/metabolism , Milk, Human/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Chymotrypsin/metabolism , Epithelial Cells , Fatty Acids, Nonesterified/pharmacology , Humans , Intestinal Mucosa/cytology , Milk Banks , Milk, Human/chemistry , Neutrophils , Rats , Temperature , Time Factors , Trypsin/metabolism
12.
Microvasc Res ; 90: 154-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24060804

ABSTRACT

Oxygen free radical and matrix metalloproteinases-9 (MMP-9) play an important pathophysiological role in the development of chronic hypertension. MMP-9 activities are regulated at different levels. We hypothesize that as mediators of the expression of MMP-9 the transcription factors like nuclear factor kappa B (NF-κB), c-fos and retinoic acid receptors-α (RAR-α) with binding sites to the MMP-9 promoter are overexpressed in the spontaneously hypertensive rat (SHR) in a process that is regulated by oxygen free radicals. Transcription factor NF-κB, c-fos and RAR-α expression levels were determined by immunohistochemistry in renal, cardiac and mesentery microcirculation of the SHR and its normotensive control, the Wistar Kyoto (WKY) rat. The animals were treated with a superoxide scavenger (Tempol) for eight weeks. The elevated plasma levels of thiobarbituric acid reactive substances and MMP-9 levels in the SHR were significantly decreased by Tempol treatment (P<0.05). The NF-κB, c-fos and RAR-α expression levels in renal glomerular, heart and mesentery microvessels were enhanced in the SHR and could also be reduced by Tempol compared to untreated animals (P<0.05). The enhanced MMP-9 levels in SHR microvessels co-express with transcription factors. These results suggest that elevated NF-κB, c-fos and RAR-α expressions and MMP-9 activity in the SHR are superoxide-dependent.


Subject(s)
Hypertension/enzymology , Matrix Metalloproteinase 9/metabolism , Microvessels/enzymology , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Blood Pressure , Disease Models, Animal , Free Radical Scavengers/pharmacology , Gene Expression Regulation, Enzymologic , Hypertension/genetics , Hypertension/physiopathology , Matrix Metalloproteinase 9/genetics , Microvessels/drug effects , Microvessels/physiopathology , NF-kappa B/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors , Up-Regulation
13.
Pediatr Res ; 72(6): 560-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23007028

ABSTRACT

BACKGROUND: Premature infants fed formula are more likely to develop necrotizing enterocolitis (NEC) than those who are breastfed, but the mechanisms of intestinal necrosis in NEC and protection by breast milk are unknown. We hypothesized that after lipase digestion, formula, but not fresh breast milk, contains levels of unbound free fatty acids (FFAs) that are cytotoxic to intestinal cells. METHODS: We digested multiple term and preterm infant formulas or human milk with pancreatic lipase, proteases (trypsin and chymotrypsin), lipase + proteases, or luminal fluid from a rat small intestine and tested FFA levels and cytotoxicity in vitro on intestinal epithelial cells, endothelial cells, and neutrophils. RESULTS: Lipase digestion of formula, but not milk, caused significant death of neutrophils (ranging from 47 to 99% with formulas vs. 6% with milk) with similar results in endothelial and epithelial cells. FFAs were significantly elevated in digested formula vs. milk and death from formula was significantly decreased with lipase inhibitor pretreatment, or treatments to bind FFAs. Protease digestion significantly increased FFA binding capacity of formula and milk but only enough to decrease cytotoxicity from milk. CONCLUSION: FFA-induced cytotoxicity may contribute to the pathogenesis of NEC.


Subject(s)
Cell Death , Enterocolitis, Necrotizing/etiology , Infant Food , Milk, Human , Animals , Cattle , Enterocolitis, Necrotizing/pathology , Humans , In Vitro Techniques , Infant, Newborn , Infant, Premature
14.
Curr Hypertens Rep ; 14(1): 88-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22081429

ABSTRACT

One of the major challenges for hypertension research is to identify the mechanisms that cause the comorbidities encountered in many hypertensive patients, as seen in the metabolic syndrome. An emerging body of evidence suggests that human and experimental hypertensives may exhibit uncontrolled activity of proteinases, including the family of matrix metalloproteinases, recognized for their ability to restructure the extracellular matrix proteins and to play a role in hypertrophy. We propose a new hypothesis that provides a molecular framework for the comorbidities of hypertension, diabetes, capillary rarefaction, immune suppression, and other cell and organ dysfunctions due to early and uncontrolled extracellular receptor cleavage by active proteinases. The proteinase and signaling activity in hypertensives requires further detailed analysis of the proteinase expression, the mechanisms causing proenzyme activation, and identification of the proteinase substrate. This work may open the opportunity for reassessment of old interventions and development of new interventions to manage hypertension and its comorbidities.


Subject(s)
Extracellular Matrix Proteins/metabolism , Hypertension/enzymology , Matrix Metalloproteinases/metabolism , Metabolic Syndrome/enzymology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Comorbidity , Humans , Hypertension/complications , Hypertension/physiopathology , Hypertrophy/enzymology , Immune Tolerance/physiology , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Microcirculation/physiology , Models, Biological , Renin-Angiotensin System/physiology , Vasoconstriction/physiology
15.
BMC Physiol ; 12: 7, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22720777

ABSTRACT

BACKGROUND: Observations in our laboratory provide evidence of vascular islands, defined as disconnected endothelial cell segments, in the adult microcirculation. The objective of this study was to determine if vascular islands are involved in angiogenesis during microvascular network growth. RESULTS: Mesenteric tissues, which allow visualization of entire microvascular networks at a single cell level, were harvested from unstimulated adult male Wistar rats and Wistar rats 3 and 10 days post angiogenesis stimulation by mast cell degranulation with compound 48/80. Tissues were immunolabeled for PECAM and BRDU. Identification of vessel lumens via injection of FITC-dextran confirmed that endothelial cell segments were disconnected from nearby patent networks. Stimulated networks displayed increases in vascular area, length density, and capillary sprouting. On day 3, the percentage of islands with at least one BRDU-positive cell increased compared to the unstimulated level and was equal to the percentage of capillary sprouts with at least one BRDU-positive cell. At day 10, the number of vascular islands per vascular area dramatically decreased compared to unstimulated and day 3 levels. CONCLUSIONS: These results show that vascular islands have the ability to proliferate and suggest that they are able to incorporate into the microcirculation during the initial stages of microvascular network growth.


Subject(s)
Capillaries/cytology , Endothelial Cells/cytology , Microcirculation/physiology , Microvessels/cytology , Neovascularization, Physiologic/physiology , Animals , Capillaries/growth & development , Cell Growth Processes/physiology , Male , Mast Cells/physiology , Mesentery/blood supply , Neovascularization, Pathologic/physiopathology , Rats , Rats, Wistar
16.
Prev Nutr Food Sci ; 27(2): 172-179, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35919566

ABSTRACT

Obesity involving adipose tissue growth and development are associated with angiogenesis and extracellular matrix remodeling. Rice bran has antioxidant and cardioprotective properties, and can act as a food supplement with potential health benefits, such as lowering blood pressure, hepatic steatosis, and inflammation. Therefore, we hypothesized that rice bran extract (RBE) can regulate adipose tissue growth and obesity. Male Institute of Cancer Research mice were fed with a high-fat diet (HFD) for 8 weeks and then supplemented with 220 and 1,100 mg/kg/d RBE while the low-fat diet group (control) were not. In addition to body weight, adipose tissue mass, and vessel density, we evaluated the mRNA expression of angiogenic factors such as matrix metalloproteinases, Mmp-2, Mmp-9, and the vascular endothelial growth factor (Vegf) in visceral and subcutaneous adipose tissues using real-time polymerase chain reaction. Administration of RBE to HFD-induced obese mice reduced the body weight and adipose tissue mass compared with untreated mice. It also decreased blood vessel density in the adipose tissue. Furthermore, RBE downregulated Vegf and Mmp-2 mRNA levels in visceral fat tissue. These results demonstrate that RBE, at high concentrations, significantly reduces adipose tissue mass and prevents obesity development in HFD-induced obese mice, which might be partly mediated via an anti-angiogenic mechanism.

17.
Eur J Trauma Emerg Surg ; 48(3): 1579-1588, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33483765

ABSTRACT

PURPOSE: Trauma and hemorrhagic shock (T/HS) is a major cause of morbidity and mortality. Existing treatment options are largely limited to source control and fluid and blood repletion. Previously, we have shown that enteral protease inhibition improves outcomes in experimental models of T/HS by protecting the gut from malperfusion and ischemia. However, enteral protease inhibition was achieved invasively, by laparotomy and direct injection of tranexamic acid (TXA) into the small intestine. In this study, we tested a minimally invasive method of enteral protease inhibitor infusion in experimental T/HS that can be readily adapted for clinical use. METHODS: Wistar rats were exsanguinated to a mean arterial blood pressure (MABP) of 40 mmHg, with laparotomy to induce trauma. Hypovolemia was maintained for 120 min and was followed by reperfusion of shed blood. Animals were monitored for an additional 120 min. A modified orogastric multi-lumen tube was developed to enable rapid enteral infusion of a protease inhibitor solution while simultaneously mitigating risk of reflux aspiration into the airways. The catheter was used to deliver TXA (T/HS + TXA) or vehicle (T/HS) continuously into the proximal small intestine, starting 20 min into the ischemic period. RESULTS: Rats treated with enteral protease inhibition (T/HS + TXA) displayed improved outcomes compared to control animals (T/HS), including significantly improved MABP (p = 0.022) and lactate (p = 0.044). Mass spectrometry-based analysis of the plasma peptidome after T/HS indicated mitigation of systemic proteolysis in T/HS + TXA. CONCLUSION: Minimally invasive, continuous enteral protease inhibitor delivery improves outcomes in T/HS and is readily translatable to the clinical arena.


Subject(s)
Shock, Hemorrhagic , Tranexamic Acid , Animals , Disease Models, Animal , Humans , Intestine, Small , Ischemia , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Rats , Rats, Wistar , Shock, Hemorrhagic/drug therapy , Tranexamic Acid/therapeutic use
18.
Am J Physiol Cell Physiol ; 301(2): C451-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21525434

ABSTRACT

Continuous exposure of polymorphonuclear leukocytes (PMNLs) to circulatory hemodynamics points to fluid flow as a biophysical regulator of their activity. Specifically, fluid flow-derived shear stresses deactivate leukocytes via actions on the conformational activities of proteins on the cell surface. Because membrane properties affect activities of membrane-bound proteins, we hypothesized that changes in the physical properties of cell membranes influence PMNL sensitivity to fluid shear stress. For this purpose, we modified PMNL membranes and showed that the cellular mechanosensitivity to shear was impaired whether we increased, reduced, or disrupted the organization of cholesterol within the lipid bilayer. Notably, PMNLs with enriched membrane cholesterol exhibited attenuated pseudopod retraction responses to shear that were recovered by select concentrations of benzyl alcohol (a membrane fluidizer). In fact, PMNL responses to shear positively correlated (R(2) = 0.96; P < 0.0001) with cholesterol-related membrane fluidity. Moreover, in low-density lipoprotein receptor-deficient (LDLr(-/-)) mice fed a high-fat diet (a hypercholesterolemia model), PMNL shear-responses correlated (R(2) = 0.5; P < 0.01) with blood concentrations of unesterified (i.e., free) cholesterol. In this regard, the shear-responses of PMNLs gradually diminished and eventually reversed as free cholesterol levels in blood increased during 8 wk of the high-fat diet. Collectively, our results provided evidence that cholesterol is an important component of the PMNL mechanotransducing capacity and elevated membrane cholesterol impairs PMNL shear-responses at least partially through its impact on membrane fluidity. This cholesterol-linked perturbation may contribute to dysregulated PMNL activity (e.g., chronic inflammation) related to hypercholesterolemia and causal for cardiovascular pathologies (e.g., atherosclerosis).


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Hypercholesterolemia/metabolism , Mechanotransduction, Cellular , Membrane Fluidity , Neutrophils/metabolism , Animals , Benzyl Alcohol/pharmacology , Cell Adhesion , Cell Membrane/drug effects , Cell Movement , Cholesterol/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Filipin/pharmacology , Humans , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Male , Mechanotransduction, Cellular/drug effects , Membrane Fluidity/drug effects , Mice , Mice, Knockout , Neutrophils/drug effects , Pseudopodia/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Stress, Mechanical , Time Factors , Up-Regulation , beta-Cyclodextrins/pharmacology
19.
Microcirculation ; 18(3): 228-37, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21418372

ABSTRACT

A complication of the spontaneously hypertensive rat (SHR) is microvascular rarefaction, defined by the loss of microvessels. However, the molecular mechanisms involved in this process remain incompletely identified. Recent work in our laboratory suggests that matrix metalloproteinases (MMPs) may play a role by cleavage of the vascular endothelial growth factor receptor 2 (VEGFR-2). In order to further delineate the role for MMPs in microvascular rarefaction, the objective of the current study was to examine the relationship in the same tissue between MMP activity, VEGFR-2 cleavage and rarefaction. Using an in vivo microzymographic technique, we show significantly enhanced levels of MMP-1, -1/-9, -7, and -8 activities, but not MMP-2 and -3 activities, along mesenteric microvessels of the SHR compared to its normotensive control, Wistar Kyoto rat. Based on immunohistochemical methods, the SHR exhibited a decreased labeling of the extracellular, but not the intracellular, domain of VEGFR-2 along mesenteric microvessels. Chronic MMP inhibition served to attenuate VEGFR-2 cleavage and microvascular network rarefaction in the SHR mesentery. These results spatially link MMP-induced VEGFR-2 cleavage and rarefaction in the mesentery of the SHR and thus support the hypothesis that MMPs serve as regulators of microvascular dysfunction in hypertension.


Subject(s)
Hypertension/physiopathology , Matrix Metalloproteinases/metabolism , Microvessels/pathology , Splanchnic Circulation , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Hydrolysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY
SELECTION OF CITATIONS
SEARCH DETAIL