Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(12): e0291701, 2023.
Article in English | MEDLINE | ID: mdl-38064470

ABSTRACT

Since the first case of COVID-19, Brazil has undergone infection waves with distinct characteristics. The description of new variants has alerted the emergence of more contagious or virulent viruses. The variant of concern Gamma emerged in Brazil and caused an epidemic wave, but its spread outside the country was limited. We report the clinical-epidemiological profile of hospitalized patients with COVID-19 by comparing two periods. A retrospective cohort study was performed. The primary outcome was to assess individuals with COVID-19 admitted in wards and intensive care units at the academic hospital of the Federal University of Parana (CHC-UFPR) between March 2020 and July 2021, correlating demographic, clinical-epidemiologic, and survival data with the most prevalent viral variant found in each period. We used Kaplan-Meier analysis to estimate the probability of survival and ROC curves to evaluate laboratory tests to find a cutoff point for poor outcomes. Data from 2,887 individuals were analyzed, 1,495 and 1,392 from the first and second periods, respectively. Hospitalization predominated among males in both periods, and the median age was significantly lower in the second one. The frequency of comorbidities was similar. Various demographic factors, clinical assessments, and laboratory tests were examined in relation to greater severity. When comparing the two periods, we observed predominance of the Wild virus during the first wave and the Gamma variant during the second, with no significant difference in outcomes. The findings suggest that despite the association of many factors with increased severity, the temporal variation between the two periods did not result in a notable divergence in the measured outcomes. The COVID-19 pandemic has lasted for a long time, with periods marked by peaks of cases, often caused by the emergence of viral variants, resulting in higher infection rates and rapid dissemination but, for variant Gamma, no apparent greater virulence.


Subject(s)
COVID-19 , Patient Admission , Humans , Male , Brazil/epidemiology , COVID-19/epidemiology , Pandemics , Retrospective Studies , SARS-CoV-2 , Tertiary Care Centers , Female
2.
Cardiovasc Res ; 118(4): 1004-1019, 2022 03 16.
Article in English | MEDLINE | ID: mdl-33757121

ABSTRACT

The human transcriptome comprises a complex network of coding and non-coding RNAs implicated in a myriad of biological functions. Non-coding RNAs exhibit highly organized spatial and temporal expression patterns and are emerging as critical regulators of differentiation, homeostasis, and pathological states, including in the cardiovascular system. This review defines the current knowledge gaps, unmet methodological needs, and describes the challenges in dissecting and understanding the role and regulation of the non-coding transcriptome in cardiovascular disease. These challenges include poor annotation of the non-coding genome, determination of the cellular distribution of transcripts, assessment of the role of RNA processing and identification of cell-type specific changes in cardiovascular physiology and disease. We highlight similarities and differences in the hurdles associated with the analysis of the non-coding and protein-coding transcriptomes. In addition, we discuss how the lack of consensus and absence of standardized methods affect reproducibility of data. These shortcomings should be defeated in order to make significant scientific progress and foster the development of clinically applicable non-coding RNA-based therapeutic strategies to lessen the burden of cardiovascular disease.


Subject(s)
Cardiovascular Diseases , RNA, Long Noncoding , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/genetics , Cardiovascular Diseases/therapy , Humans , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reproducibility of Results , Transcriptome
3.
J Transl Med ; 18(1): 408, 2020 10 31.
Article in English | MEDLINE | ID: mdl-33129318

ABSTRACT

COronaVIrus Disease 19 (COVID-19) is caused by the infection of the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2). Although the main clinical manifestations of COVID-19 are respiratory, many patients also display acute myocardial injury and chronic damage to the cardiovascular system. Understanding both direct and indirect damage caused to the heart and the vascular system by SARS-CoV-2 infection is necessary to identify optimal clinical care strategies. The homeostasis of the cardiovascular system requires a tight regulation of the gene expression, which is controlled by multiple types of RNA molecules, including RNA encoding proteins (messenger RNAs) (mRNAs) and those lacking protein-coding potential, the noncoding-RNAs. In the last few years, dysregulation of noncoding-RNAs has emerged as a crucial component in the pathophysiology of virtually all cardiovascular diseases. Here we will discuss the potential role of noncoding RNAs in COVID-19 disease mechanisms and their possible use as biomarkers of clinical use.


Subject(s)
Cardiovascular Diseases/complications , Coronavirus Infections/complications , Pneumonia, Viral/complications , RNA, Untranslated , Angiotensin-Converting Enzyme 2 , Animals , Arrhythmias, Cardiac/complications , Betacoronavirus , COVID-19 , Cardiomegaly/complications , Cardiovascular Diseases/genetics , Gene Expression Profiling , Gene Expression Regulation , Homeostasis , Humans , Inflammation/complications , Mice , Pandemics , Peptidyl-Dipeptidase A/genetics , Renin-Angiotensin System , SARS-CoV-2 , Transcriptome
4.
Cell Mol Biol (Noisy-le-grand) ; 61(1): 30-5, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25817343

ABSTRACT

Insulin is known to regulate blood—glucose level and promote its utilization as an energy source in cardiac tissues under normal physiological conditions as well as stimulates signaling pathways that involved cell growth and proliferation. Although recently insulin generated free radicals via NAD(P)H has been documented but the molecular mechanism is still under investigation. The aim of present study is to elucidate the reactive oxygen species (ROS) dependent possible role of insulin in cardiac abnormalities, including hypertrophy by regulation of antioxidants enzyme (SOD) activity. In the current study, 60 cardiac patients and 50 healthy individuals as well as the rat model with insulin administration were under investigation. Oxidant, anti—oxidant biochemical assays, hypertrophic marker expression via immunobloting and histopathology were performed. We observed statistically significant elevation of the reactive oxygen species level in the serum of patients as well as in the insulin administrated rat model, a mild expression of cardiac marker in experimental models along with abnormal histopathology of hearts. However, super oxide dismutase free radical scavenger activity was down regulated upon insulin treatment compared to control rats. Conclusively, the present study showed that over expression of insulin might stimulate cardiac hypertrophic signal via up regulation of free radicals and down regulation of antioxidants enzymes including SOD activity.


Subject(s)
Cardiomegaly/physiopathology , Insulin/physiology , Reactive Oxygen Species/metabolism , Up-Regulation/physiology , Adult , Aged , Animals , Case-Control Studies , Disease Models, Animal , Female , Humans , Insulin/pharmacology , Male , Middle Aged , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Superoxide Dismutase/metabolism
5.
Br J Pharmacol ; 164(2b): 570-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21486281

ABSTRACT

BACKGROUND AND PURPOSE: Statins, a major component of the prevention of cardiovascular disease, aid progenitor cell functions in vivo and in vitro. Statins bearing a NO-releasing moiety were developed for their enhanced anti-inflammatory/anti-thrombotic properties. Here, we investigated if the NO-donating atorvastatin (NCX 547) improved the functions of circulating angiogenic cells (CACs). EXPERIMENTAL APPROACH: Circulating angiogenic cells (CACs) were prepared from peripheral blood monocytes of healthy volunteers and type-2 diabetic patients and were cultured in low (LG) or high glucose (HG) conditions, in presence of atorvastatin or NCX 547 (both at 0.1 µM) or vehicle. Functional assays (outgrowth, proliferation, viability, senescence and apoptosis) were performed in presence of the endothelial NOS inhibitor L-NIO, the NO scavenger c-PTIO or vehicle. KEY RESULTS: Culturing in HG conditions lowered NO in CACs, inhibited outgrowth, proliferation, viability and migration, and induced cell senescence and apoptosis. NCX 547 fully restored NO levels and functions of HG-cultured CACs, while atorvastatin prevented only apoptosis in CACs. The activity of Akt, a pro-survival kinase, was increased by atorvastatin in LG-cultured but not in HG-cultured CACs, whereas NCX 547 increased Akt activity in both conditions. L-NIO partially blunted and c-PTIO prevented NCX 547-induced improvements in CAC functions. Finally, NCX 547 improved outgrowth and migration of CACs prepared from patients with type 2 diabetes. CONCLUSIONS AND IMPLICATIONS: NCX 547 was more effective than atorvastatin in preserving functions of CACs. This property adds to the spectrum of favourable actions that would make NO-releasing statins more effective agents for treating cardiovascular disease.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Nitrates/pharmacology , Nitric Oxide Donors/pharmacology , Pyrroles/pharmacology , Stem Cells/drug effects , Aged , Anticholesteremic Agents/pharmacology , Apoptosis/drug effects , Benzoates/pharmacology , Cardiovascular Agents/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Female , Humans , Imidazoles/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Nitric Oxide Synthase Type III/antagonists & inhibitors , Ornithine/analogs & derivatives , Ornithine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , Stem Cells/metabolism
6.
Br J Cancer ; 103(9): 1422-31, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20859288

ABSTRACT

BACKGROUND: Human tissue kallikrein (hK1) generates vasodilator kinins from kininogen and promotes angiogenesis by kinin-dependent and kinin-independent mechanisms. Here, we investigate the expression and functional relevance of hK1 in human gastrointestinal stromal tumour (GIST). METHODS: Vascularisation and hK1 expression of GIST samples were assessed by immunohistochemistry. In two GIST cell lines, hK1 expression was assessed by PCR, and hK1 protein levels and activity were measured by ELISA and an amidolytic assay, respectively. The effect of hK1 silencing, inhibition or overexpression on GIST cell proliferation, migration and paracrine induction of angiogenesis was studied. Finally, local and systemic levels of hK1 were assessed in mice injected with GIST cells. RESULTS: Human tissue kallikrein was detected in 19 out of 22 human GIST samples. Moreover, GIST cells express and secrete active hK1. Titration of hK1 demonstrated its involvement in GIST invasive behaviour, but not proliferation. Furthermore, hK1 released by GIST cells promoted endothelial cell migration and network formation through kinin-dependent mechanisms. Gastrointestinal stromal tumour implantation in nude mice resulted in local and systemic hK1 expression proportional to tumour dimension. CONCLUSIONS: Human tissue kallikrein is produced and released by GIST and participates in tumour invasion. Further studies are needed to validate hK1 as a diagnostic biomarker and therapeutic target in GIST.


Subject(s)
Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Neoplasm Invasiveness , Tissue Kallikreins/physiology , Animals , Cell Line, Tumor , Cell Movement , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Tissue Kallikreins/blood , Tissue Kallikreins/metabolism
7.
Cell Death Differ ; 15(2): 299-311, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17992191

ABSTRACT

Neurotrophins (NTs) control neuron survival and regeneration. Recent research showed that NTs possess cardiovascular actions. In this study, we investigated the hypothesis that the NT nerve growth factor (NGF) prevents cardiomyocyte apoptosis. We demonstrated that cultured rat neonatal cardiomyocytes (RNCMs) produce NGF and express its trkA (tropomyosin-related receptor A (NGF high-affinity receptor)) receptor. RNCMs given a neutralizing antibody for NGF or the trkA inhibitor K252a underwent apoptosis, thus suggesting that NGF is an endogenous prosurvival factor for cardiomyocytes. Adenovirus (Ad)-mediated NGF overexpression protected RNCMs from apoptosis induced by either hypoxia/reoxygenation or angiotensin II (AngII). Similarly, recombinant NGF inhibited AngII-induced apoptosis in isolated rat adult cardiomyocytes. Finally, in a rat model of myocardial infarction, NGF gene transfer promoted cardiomyocyte survival. In RNCMs, recombinant NGF induced trkA phosphorylation, followed by Ser473 phosphorylation and nuclear translocation of phospho-protein kinase B (Akt). In response to Akt activation, Forkhead transcription factors Foxo-3a and Foxo-1 were phosphorylated and excluded from the nucleus. The prosurvival effect of adenoviral vector carrying the human NGF gene was inhibited in vitro by K252a, LY294002 (a pan-phosphatidyl inositol 3-kinase - PI3K - inhibitor), an Akt small interfering RNA, and adenoviruses carrying a dominant negative mutant form of Akt (Ad.DN.Akt) or an Akt-resistant Foxo-3a (Ad.AAA-Foxo-3a). These results newly demonstrate the cardiac prosurvival action of NGF and provide mechanistic information on the signaling pathway, which encompasses trkA, PI3K-Akt, and Foxo.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Receptor, trkA/metabolism , Angiotensin II/metabolism , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Cell Survival , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Humans , Indole Alkaloids/pharmacology , Morpholines/pharmacology , Myocytes, Cardiac/drug effects , Nerve Growth Factor/genetics , Nerve Tissue Proteins/metabolism , PC12 Cells , Phosphorylation , Rats , Signal Transduction , Transfection
8.
Br J Pharmacol ; 150(7): 873-82, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17351667

ABSTRACT

BACKGROUND AND PURPOSE: Mature endothelial cells and their progenitors are dysfunctional in diabetes, resulting in deficient neovascularisation following arterial occlusion. This study aimed to evaluate the therapeutic activity of a nitric oxide (NO) releasing statin in the setting of experimental diabetes and peripheral ischaemia. EXPERIMENTAL APPROACH: The effects of NCX 6550, an NO-releasing pravastatin derivative, on angiogenesis in ischaemic limbs was studied in normoglycaemic mice or mice made diabetic by treatment with streptozotocin (STZ). Control mice received an equimolar dosage of the parent statin compound, pravastatin. The therapeutic action of NCX 6550 was also tested in mice lacking the gene for endothelial nitric oxide synthase (eNOS). KEY RESULTS: In normoglycaemic or STZ-diabetic CD1 mice, only NCX 6550 stimulated skeletal muscle revascularisation. In addition, NCX 6550 induced greater improvement in limb reperfusion and salvage, than pravastatin. The number of circulating endothelial progenitor cells was decreased in STZ-diabetic mice, this defect being prevented by NCX 6550 and, to a lesser extent by pravastatin. In vitro, high glucose concentrations reduced the migratory capacity of endothelial progenitor EPCs, which was partly reversed by preincubation with pravastatin and completely reversed by NCX 6550. The postischaemic recovery of eNOS knockout mice was severely impaired as a consequence of depressed angiogenesis and this recovery was improved by treatment with NCX 6550, but not with pravastatin. CONCLUSIONS AND IMPLICATIONS: These findings indicate that incorporation of a bioactive NO moiety improves the therapeutic profile of statins for the treatment of peripheral vascular disease.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Nitrates/therapeutic use , Pravastatin/analogs & derivatives , Pravastatin/therapeutic use , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Hindlimb/blood supply , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/pathology , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics
9.
J Biomed Mater Res B Appl Biomater ; 81(2): 462-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17034001

ABSTRACT

Microfabrication techniques were combined with fractal algorithms to realize polymeric scaffolds resembling capillary networks. The scaffolds were seeded with human endothelial cells in monoculture as well as in coculture with human fibroblasts. To enhance the process of angiogenesis, endothelial cells were transfected with an adenoviral vector carrying the gene for human tissue kallikrien. The results demonstrate that both the presence of a structured scaffold as well as fibroblasts in coculture contribute synergically to the promotion of a metabolically active network. The fractal scaffolds have several possible applications for example in vascularized tissue engineering and therapeutic angiogenesis. A broader implication of these results is that cell-extra cellular matrix and cell-cell interactions cooperate dynamically both at a biochemical as well as microstructural level.


Subject(s)
Biocompatible Materials/chemistry , Capillaries/growth & development , Neovascularization, Physiologic , Polymers/chemistry , Tissue Engineering/methods , Algorithms , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fractals , Genetic Vectors , Humans , Materials Testing , Polyesters/chemistry , Tetrazolium Salts , Thiazoles , Tissue Kallikreins/genetics , Transfection
10.
Vascul Pharmacol ; 45(5): 281-301, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17010676

ABSTRACT

Ischemic disease represents the new epidemic worldwide. Animal models of ischemic disease are useful because they can help us to understand the underlying pathogenetic mechanisms and develop new therapies. The present review article summarizes the results of a consensus conference on the status and future development of experimentation in the field of cardiovascular medicine using murine models of peripheral and myocardial ischemia. The starting point was to recognize the limits of the approach, which mainly derive from species- and disease-related differences in cardiovascular physiology. For instance, the mouse heart beats at a rate 10 times faster than the human heart. Furthermore, healing processes are more rapid in animals, as they rely on mechanisms that may have lost relevance in man. The main objective of the authors was to propose general guidelines, diagnostic end points and relevance to clinical problems.


Subject(s)
Animal Experimentation , Disease Models, Animal , Extremities/blood supply , Graft Occlusion, Vascular/physiopathology , Ischemia/physiopathology , Myocardial Ischemia/physiopathology , Animal Experimentation/ethics , Animal Experimentation/legislation & jurisprudence , Animals , Atherosclerosis/surgery , Comorbidity , Consensus , Diabetes Mellitus, Type 1/physiopathology , Endpoint Determination , Graft Occlusion, Vascular/therapy , Guidelines as Topic , Humans , Ischemia/therapy , Mice , Myocardial Ischemia/therapy , Regenerative Medicine , Reproducibility of Results , Severity of Illness Index , Species Specificity , Veins/transplantation , Wound Healing
11.
Diabetologia ; 49(2): 405-20, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16416271

ABSTRACT

AIMS/HYPOTHESIS: Benfotiamine, a vitamin B1 analogue, reportedly prevents diabetic microangiopathy. The aim of this study was to evaluate whether benfotiamine is of benefit in reparative neovascularisation using a type I diabetes model of hindlimb ischaemia. We also investigated the involvement of protein kinase B (PKB)/Akt in the therapeutic effects of benfotiamine. METHODS: Streptozotocin-induced diabetic mice, given oral benfotiamine or vehicle, were subjected to unilateral limb ischaemia. Reparative neovascularisation was analysed by histology. The expression of Nos3 and Casp3 was evaluated by real-time PCR, and the activation state of PKB/Akt was assessed by western blot analysis and immunohistochemistry. The functional importance of PKB/Akt in benfotiamine-induced effects was investigated using a dominant-negative construct. RESULTS: Diabetic muscles showed reduced transketolase activity, which was corrected by benfotiamine. Importantly, benfotiamine prevented ischaemia-induced toe necrosis, improved hindlimb perfusion and oxygenation, and restored endothelium-dependent vasodilation. Histological studies revealed the improvement of reparative neovascularisation and the inhibition of endothelial and skeletal muscle cell apoptosis. In addition, benfotiamine prevented the vascular accumulation of advanced glycation end products and the induction of pro-apoptotic caspase-3, while restoring proper expression of Nos3 and Akt in ischaemic muscles. The benefits of benfotiamine were nullified by dominant-negative PKB/Akt. In vitro, benfotiamine stimulated the proliferation of human EPCs, while inhibiting apoptosis induced by high glucose. In diabetic mice, the number of circulating EPCs was reduced, with the deficit being corrected by benfotiamine. CONCLUSIONS/INTERPRETATION: We have demonstrated, for the first time, that benfotiamine aids the post-ischaemic healing of diabetic animals via PKB/Akt-mediated potentiation of angiogenesis and inhibition of apoptosis. In addition, benfotiamine combats the diabetes-induced deficit in endothelial progenitor cells.


Subject(s)
Apoptosis/drug effects , Diabetic Angiopathies/drug therapy , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Thiamine/analogs & derivatives , Animals , Blotting, Western , Body Weight , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/physiopathology , Dietary Supplements , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Enzyme Activation/drug effects , Hemodynamics/drug effects , Immunohistochemistry , Ischemia/physiopathology , Ischemia/prevention & control , Male , Mice , Mice, Inbred Strains , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Stem Cells/drug effects , Stem Cells/pathology , Stem Cells/physiology , Thiamine/pharmacology , Thiamine/therapeutic use
12.
Ann Rheum Dis ; 64(3): 382-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15708892

ABSTRACT

BACKGROUND: In systemic sclerosis (SSc) the lack of an angiogenic response to hypoxia may be due to inappropriate synthesis of angiogenic and angiostatic factors. Tissue kallikrein (t-kallikrein), regulating the kallikrein-kinin system and acting on the microcirculation, is a potent angiogenic agent, and kallistatin is its natural inhibitor. OBJECTIVE: To evaluate, in patients with SSc, t-kallikrein and kallistatin levels and their correlation with clinical features and measures of microvascular involvement. PATIENTS AND METHODS: Serum levels of t-kallikrein and kallistatin (ELISA) and t-kallikrein skin expression (immunohistochemistry) were studied in patients with SSc, and evaluated for subset (dSSc or lSSc), clinical and immunological features, and microvascular involvement (ulcers, telangiectasias, nailfold videocapillaroscopy). RESULTS: Circulating levels of t-kallikrein were higher in SSc than in controls (p<0.001). T-kallikrein did not differ between lSSc and dSSc, although it was higher in lSSc than in controls (p<0.001).T-kallikrein levels were higher in patients with early and active capillaroscopic pattern than in those with late pattern (p = 0.019 and 0.023). Patients with giant capillaries and capillary microhaemorrhages had higher t-kallikrein concentrations than patients with architectural derangement (p = 0.04). No differences in kallistatin levels were detected between patients with SSc and controls, or between lSSc and dSSc. In early SSc skin, the presence of t-kallikrein was found in endothelial and in perivascular inflammatory cells, while no staining in skin of advanced SSc was detected. CONCLUSION: T-kallikrein levels are increased in patients with SSc, particularly in lSSc, and are associated with early and active capillaroscopic patterns. T-kallikrein may play a part in SSc microvascular changes.


Subject(s)
Scleroderma, Systemic/blood , Tissue Kallikreins/blood , Adult , Aged , Autoantibodies/blood , Capillaries/pathology , Carrier Proteins/blood , Endothelium, Vascular/metabolism , Female , Humans , Immunoenzyme Techniques , Male , Microcirculation , Microscopic Angioscopy/methods , Middle Aged , Scleroderma, Systemic/pathology , Serpins/blood , Skin/metabolism , Tissue Kallikreins/antagonists & inhibitors
13.
Arch Mal Coeur Vaiss ; 97(6): 679-87, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15283043

ABSTRACT

Angiogenesis is essential for the repair of wounds and tissues damaged by ischemia. The regenerative process is tightly regulated by master angiogenic factors, cytokines and the downstream mediator NO. In addition, modulators of vascular growth, such as COX-2-generated prostanoids, contribute to the process by stabilizing the hypoxia-inducible factor and stimulating the expression of VEGF. Recently, we discovered that human tissue kallikrein, a member of the serine proteinase superfamily, possesses potent angiogenic effects. It has been categorized as a pleiotropic angiogenic agent acting via enzymatic cleavage of kininogen and subsequent release of kinin peptides. Kinins bind G-protein coupled receptors, subtype B1 and B2, and exert proliferative effects on endothelial cells via an IP3K-Akt-NO mediated mechanism independent of VEGF. In addition, kinins stimulate the release of angiogenic prostacyclin. Gene transfer of human tissue kallikrein rescues ischemic tissues in otherwise normal mice, as well as in hypertensive or diabetic animals. In addition, prophylactic gene delivery of tissue kallikrein to diabetic skeletal muscles prevents the development of microangiopathy and stimulates collateralization, thus protecting from the consequences of supervening arterial occlusion.


Subject(s)
Coagulants/pharmacology , Coagulants/therapeutic use , Gene Transfer Techniques , Kallikreins/pharmacology , Kallikreins/therapeutic use , Neovascularization, Physiologic/drug effects , Animals , Diabetes Complications , Disease Models, Animal , Humans , Hypertension/complications , Kallikreins/genetics , Mice , Myocardial Ischemia/therapy , Myocardial Ischemia/veterinary , Stroke/therapy , Vascular Endothelial Growth Factor A
14.
Diabetologia ; 47(6): 1055-63, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15184980

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes increases the risk of peripheral ischaemia and impairs recovery once ischaemia occurs, probably because the healing process is hampered by diabetes-induced endothelial dysfunction. In normoglycaemic mice subjected to limb ischaemia, blockade of nerve growth factor (NGF) compromises reparative angiogenesis. In the present study, we evaluated if expressional alterations of endogenous NGF system components are associated with diabetes-related impairment in neovascularisation. In addition, we tested whether the correction of NGF liabilities benefits post-ischaemic healing of Type 1 diabetic animals. METHODS: Unilateral hindlimb ischaemia was produced in streptozotocin-induced Type 1 diabetic mice. Purified murine NGF (20 microg daily for 14 days) or PBS were injected into ischaemic adductors. Non-diabetic mice given PBS served as controls. Hindlimb blood flow was analysed sequentially for up to 14 days. At necroscopy, adductors were removed for quantification of microvessel density, endothelial cell apoptosis and NGF receptor expression. NGF content was determined by ELISA three days after ischaemia. In vitro, we tested whether NGF protects endothelial cells from apoptosis induced by high glucose and whether vascular endothelial growth factor-A (VEGF-A) is involved in this beneficial effect. RESULTS: Muscles removed from Type 1 diabetic mice showed reduced NGF content and up-regulation of the NGF p75 receptor. NGF supplementation promoted capillarisation and arteriogenesis, reduced apoptosis, and accelerated blood flow recovery. NGF stimulated VEGF-A production by human endothelial cells incubated in high-glucose medium and conferred resistance against high-glucose-induced apoptosis via a VEGF-A-mediated mechanism. CONCLUSIONS/INTERPRETATION: NGF protects endothelial cells from apoptosis induced by Type 1 diabetes and facilitates reparative neovascularisation. The findings may open up new therapeutic options for the treatment of diabetic complications.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Hindlimb/drug effects , Hindlimb/injuries , Ischemia/complications , Ischemia/drug therapy , Nerve Growth Factor/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/physiology , Capillaries/drug effects , Capillaries/pathology , Capillaries/physiopathology , Cell Survival , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Drug Administration Schedule , Drug Evaluation, Preclinical/methods , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression , Hindlimb/blood supply , Ischemia/physiopathology , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Reperfusion , Retinal Vessels/physiology , Retinal Vessels/ultrastructure , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/physiology , Vascular Endothelial Growth Factor A/metabolism
15.
Diabetologia ; 47(6): 1047-54, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15164170

ABSTRACT

AIMS/HYPOTHESIS: The neurotrophin nerve growth factor (NGF) is pro-angiogenic and facilitates wound repair. The present study was conducted to (i) assess the statement of NGF system components in diabetic wounds and (ii) evaluate whether NGF supplementation could prevent impairment of wound neoangiogenesis by diabetes. METHODS: Skin wounds were produced in the interscapular region of streptozotocin-induced diabetic mice. NGF (1 microg per day in PBS) or vehicle was applied onto the ulcers for 3 days after punching. Non-diabetic mice were used as controls. RESULTS: In wounds of untreated diabetic mice, endogenous levels of immunoreactive NGF were lower than those in wounds of non-diabetic mice ( p<0.01). Immunohistochemical analysis showed down-regulation of tyrosine kinase receptor-A (TrkA) and up-regulation of p75 receptor in granulation tissue microvasculature. Local NFG administration prevented diabetes-induced expressional alterations, enhanced reparative capillarisation ( p<0.01), and accelerated wound closure ( p<0.01). This was associated with a three-fold increase in endothelial cell proliferation ( p<0.01), while apoptosis was reduced by 50% ( p<0.05). Quantitative RT-PCR documented a 5.5-fold increase in the expression of vascular endothelial growth factor-A (VEGF-A) by exogenous NGF in diabetic tissues ( p<0.01). In in vitro preparations of human endothelial cells from derma, NGF increased the release of immunoreactive VEGF-A, and reduced high-glucose-induced apoptosis ( p<0.05), the latter effect being inhibited by a VEGF-A receptor-2 antagonist. CONCLUSIONS/INTERPRETATION: Diabetic ulcers display distinct alterations in reparative angiogenesis and in the expression of NGF and its receptors. NGF supplementation corrects endogenous liabilities, facilitates vascular regeneration, and suppresses endothelial apoptosis seemingly via VEGF-A. Our findings unravel new mechanisms responsible for NGF reparative action.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus, Type 1/complications , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Nerve Growth Factor/therapeutic use , Skin Ulcer/complications , Skin Ulcer/drug therapy , Administration, Cutaneous , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression/drug effects , Glucose/adverse effects , Glucose/antagonists & inhibitors , Glucose/metabolism , Humans , Italy , Male , Mice , Neovascularization, Physiologic/physiology , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Skin Ulcer/pathology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , Wound Healing/physiology
16.
Trends Pharmacol Sci ; 22(9): 478-84, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11543876

ABSTRACT

Kinins, the biological end-products of the kallikrein-kininogen system, influence many aspects of the cellular function. Interest in this peptidergic system has been renewed recently by the discovery that kinins exert cardiovascular protective effects and promote post-ischaemic recovery by stimulating vascular growth. Pharmacological and genetic studies indicate that induction of kallikrein and kinin receptors by ischaemia is functionally relevant in the natural host response that permits perfusion recovery and tissue healing. Furthermore, potentiation of the generation of kinins by continuous supply of tissue kallikrein promotes reparative angiogenesis through stimulation of the release of nitric oxide and prostaglandins. Strategies that activate kinin receptors might be applicable to the treatment of occlusive vascular disease, whereas kinin receptor antagonists could represent therapeutic reagents against pathological angiogenesis in cancer and chronic inflammatory conditions.


Subject(s)
Ischemia/therapy , Kallikrein-Kinin System , Neovascularization, Physiologic/physiology , Receptors, Cell Surface/physiology , Animals , Asialoglycoprotein Receptor , Humans , Kallikrein-Kinin System/genetics , Kallikrein-Kinin System/physiology
17.
Br J Pharmacol ; 133(7): 951-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487503

ABSTRACT

Ischaemic diseases are characterized by an impaired supply of blood resulting from narrowed or blocked arteries that starve tissues of needed nutrients and oxygen. Coronary-atherosclerosis induced myocardial infarction is one of the leading causes of mortality in developed countries. Ischaemic disease also affects the lower extremities. Considerable advances in both surgical bypassing and percutaneous revascularization techniques have been reached. However, many patients cannot benefit from these therapies because of the extension of arterial occlusion and/or microcirculation impairment. Consequently, the need for alternative therapeutic strategies is compelling. An innovative approach consists of stimulating collateral vessel growth, a natural host defence response that intervenes upon occurrence of critical reduction in tissue perfusion (Isner & Asahara, 1999). This review will debate the relevance of therapeutic angiogenesis for promotion of tissue repair. The following issues will receive attention: (a) vascular growth patterns, (b) delivery systems for angiogenesis gene transfer, (c) achievements of therapeutic angiogenesis in myocardial and peripheral ischaemia, and (d) future directions to improve effectiveness and safety of vascular gene therapy.


Subject(s)
Genetic Therapy/methods , Ischemia/therapy , Neovascularization, Physiologic , Animals , Forecasting , Genetic Therapy/trends , Humans , Ischemia/genetics , Models, Biological
18.
Hypertension ; 38(1): 136-41, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11463774

ABSTRACT

Angiogenesis represents a compensatory response targeted to preserve the integrity of tissues subjected to ischemia. The aim of the present study was to examine whether reparative angiogenesis is impaired in spontaneously hypertensive rats (SHR), as a function of progression of hypertension. In addition, the potential of gene therapy with human tissue kallikrein (HK) in revascularization was challenged in SHR and normotensive Wistar-Kyoto rats (WKY) that underwent excision of the left femoral artery. Expression of vascular endothelial growth factor and HK was upregulated in ischemic hindlimb of WKY but not of SHR. Capillary density was increased in ischemic adductor muscle of WKY (from 266+/-20 to 633+/-73 capillaries/mm(2) at 28 days, P<0.001), whereas it remained unchanged in SHR (from 276+/-20 to 354+/-48 capillaries/mm(2), P=NS), thus compromising perfusion recovery as indicated by reduced plantar blood flow ratio (0.61+/-0.08 versus 0.92+/-0.07 in WKY at 28 days, P<0.05). In separate experiments, saline or 5x10(9) pfu adenovirus containing the HK gene (Ad.CMV-cHK) or the beta-galactosidase gene (Ad.CMV-LacZ) was injected intramuscularly at 7 days after the induction of ischemia. Ad.CMV-cHK augmented capillary density and accelerated hemodynamic recovery in both strains, but these effects were more pronounced in SHR (P<0.01). Our results indicate that native angiogenic response to ischemia is impaired in SHR, possibly as a result of defective modulation of endothelial cell mitogens. Supplementation with kallikrein, one of the growth factors found to be deficient in SHR, restores physiological angiogenic response utilitarian for tissue healing. Our discoveries may have important implications in vascular medicine for therapeutic benefit.


Subject(s)
Kallikreins/physiology , Muscles/physiology , Neovascularization, Physiologic/physiology , Animals , Gene Transfer Techniques , Hindlimb/blood supply , Humans , Injections, Intramuscular , Ischemia/genetics , Ischemia/physiopathology , Kallikreins/genetics , Kallikreins/pharmacology , Male , Perfusion , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Regional Blood Flow
19.
Peptides ; 22(6): 915-22, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11390021

ABSTRACT

The involvement of kinins, calcitonin gene-related peptide (CGRP), and tachykinins during mesenteric post-ischemic reperfusion was studied in anesthetized rats by using antagonists for bradykinin (BK) B1, BK B2, CGRP1, or tachykinin NK1 receptor, or by capsaicin-induced desensitization. B1, B2, or CGRP1 receptor antagonists or desensitization attenuated the transient hypotension and plasma protein and leukocyte infiltration of intestinal wall observed during post-ischemic reperfusion. These effects were abolished by the combination of B2 and CGRP1 blockade as well as by B2 antagonism in capsaicinized rats, while NK1 blockade was ineffective. Our results suggest that kinins and CGRP contribute to systemic vasodilatation and microvascular leakage during mesenteric reperfusion. Pharmacological blockade of these systems could help preventing hypotension and intestinal injury consequent to reperfusion.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Intestinal Mucosa/metabolism , Kinins/physiology , Reperfusion Injury/metabolism , Reperfusion , Animals , Bradykinin Receptor Antagonists , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Capsaicin/pharmacology , Duodenum/pathology , Inflammation/metabolism , Ischemia/metabolism , Male , Mesenteric Arteries/drug effects , Neurokinin-1 Receptor Antagonists , Rats , Rats, Wistar , Receptor, Bradykinin B1 , Receptor, Bradykinin B2 , Tachykinins/biosynthesis , Time Factors
20.
Kidney Int ; 59(6): 2233-42, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380826

ABSTRACT

BACKGROUND: Renal kallikrein has been linked with inheritance of arterial hypertension and with sensitivity to drug nephrotoxicity. Identification of a cause--effect relationship between low kallikrein and intermediate phenotypes has been hampered by the lack of adequate animal models. METHODS: Kallikrein was measured in tissues obtained from rats inbred for low urinary kallikrein excretion (LKR) and wild-type controls. Blood pressure and indices of myocardial contractility were recorded via an intraventricular cannula connected to a transducer. The functional relevance of endogenous angiotensin II (Ang II) in LKR was explored by determining the effect of Ang II subtype 1 (AT(1)) receptor blockade on glomerular filtration rate, renal blood flow, and urinary sodium excretion. In addition, sensitivity to gentamycin-induced nephrotoxicity was evaluated. RESULTS: Kallikrein activity was reduced by 60% in the kidney of LKR (P < 0.01), whereas it was increased in the heart (P < 0.05) and was unaltered in the pancreas, liver, and salivary glands. Heart rate and myocardial contractility were reduced, and the mean blood pressure (MBP) was increased in LKR as compared with controls (P < 0.05). LKR exhibited polydipsia, polyuria, glomerular hyperfiltration, and reduced fractional sodium excretion under basal conditions and impaired renal vasodilation in response to volume expansion. These functional alterations were significantly attenuated by AT(1) receptor blockade. Gentamycin reduced the glomerular filtration rate in LKR, but not in controls. CONCLUSIONS: In LKR, unopposed activity of Ang II appears to be responsible for increased glomerular hydrostatic pressure and augmented tubular reabsorption. Balance between the kallikrein-kinin and renin-angiotensin systems is essential for normal renal function.


Subject(s)
Kallikreins/genetics , Kallikreins/urine , Kidney/chemistry , Kidney/physiology , Angiotensin Receptor Antagonists , Animals , Anti-Bacterial Agents/toxicity , Blood Volume , Body Weight , Gentamicins/toxicity , Heart Rate , Kallikreins/analysis , Kidney Concentrating Ability/physiology , Kidney Diseases/chemically induced , Kidney Diseases/urine , Male , Osmolar Concentration , Phenotype , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Water Deprivation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...