Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Life Sci ; 329: 121973, 2023 Sep 15.
Article En | MEDLINE | ID: mdl-37482211

AIMS: Tissue kallikrein-related peptidase8 (KLK8) has been found to mitigate acute myocardial ischemia-reperfusion (IR) injury. However, the effect of KLK8 on cardiac remodeling in response to IR injury has not been determined. MATERIALS AND METHODS: KLK8 overexpressing transgenic rat (KLK8-TG) was used as the animal model. IR injury was induced by ligating the left anterior descending coronary artery for 1 h and subsequent reperfusion. The functional and morphological changes of the heart were examined 14 days after the injury. Neonatal rat cardiac fibroblasts (CFs) were used to investigate the molecular mechanisms in vitro. KEY FINDINGS: KLK8 overexpression enhanced cardiac diastolic dysfunction, fibrosis, and hypertrophy after IR injury, indicating that KLK8 accentuated cardiac remodeling in response to IR injury. Moreover, KLK8 overexpression increased epidermal growth factor (EGF) release and promoted the phosphorylation of EGF receptor (EGFR) and ERK1/2 in the heart after IR injury. It was interesting to find that both EGFR antagonist (AG 1478) and MEK inhibitor (PD98059) attenuated the KLK8-induced proliferation and activation of CFs in vitro, indicating that EGFR signaling might mediate the pro-fibrotic action of KLK8. SIGNIFICANCE: KLK8 plays a crucial role in cardiac remodeling after myocardial infarction. KLK8 accentuates cardiac fibrosis after IR injury, possibly mediated by EGFR signaling in CFs.


Myocardial Reperfusion Injury , Rats , Animals , Myocardial Reperfusion Injury/metabolism , Tissue Kallikreins/genetics , Tissue Kallikreins/metabolism , Tissue Kallikreins/pharmacology , Ventricular Remodeling , ErbB Receptors/metabolism , Fibrosis , Fibroblasts/metabolism , Myocardium/metabolism
2.
Domest Anim Endocrinol ; 81: 106748, 2022 10.
Article En | MEDLINE | ID: mdl-35842984

Estradiol-17ß (E2) increases kallikrein in rodent and human reproductive tissues. Kallikrein specific activity is increased in the porcine uterus when conceptus E2 is secreted at maternal recognition of pregnancy. When kallikrein acts on kininogen to liberate bradykinin, angiogenic and vasoactive factors are released. The uterus of ovariectomized ewes administered E2 undergoes rapid vascular changes via different patterns of angiogenic and vasoactive factors. Our hypothesis was that E2 would increase the specific activity and protein secretion of tissue kallikrein in endometrial explants culture media (ECM) and ewes exposed to E2 would have uterine arteries that would be more sensitive to the vasodilatory effects of bradykinin. Ovariectomized ewes received 100 mg of E2 implants for 0, 12, 24, or 48 h. After treatment, uterine weights were determined, and caruncles were processed for ECM. Uterine weights and uterine weight per ewe body weight were significantly greater in the 12 and 24 h ewes compared with the 0 h ewes, with the 48 h ewes being similar to the 24 h ewes. There were no statistically significant differences in caruncular tissue kallikrein protein secretion among the treatment groups. There was a tendency (P = 0.09) for duration of E2 exposure to influence tissue kallikrein specific activity where kallikrein activity was greater (P ≤ 0.05) in the 12 and 48 h ewes compared with the 0 h ewes, with 24 h ewes being intermediate (unprotected F test). Uterine arteries from ewes with E2 for 24 and 48 h had more sensitivity to bradykinin, via the bradykinin receptor 2, than uterine arteries from ewes with 0 or 12 h E2 exposure. We fail to reject our hypothesis as E2 did elicit a positive response in tissue kallikrein specific activity and bradykinin response. Further investigations are needed to determine how kallikrein and bradykinin may be involved in vascular remodeling of the ovine uterus.


Bradykinin , Estradiol , Animals , Bradykinin/metabolism , Bradykinin/pharmacology , Cell Proliferation , Estradiol/metabolism , Estradiol/pharmacology , Female , Humans , Kallikreins/metabolism , Kallikreins/pharmacology , Pregnancy , Sheep , Swine , Tissue Kallikreins/metabolism , Tissue Kallikreins/pharmacology , Transcription Factors/metabolism , Uterus/metabolism
3.
Int Immunopharmacol ; 105: 108547, 2022 Apr.
Article En | MEDLINE | ID: mdl-35066448

BACKGROUND: Tissue kallikrein offers a wide spectrum of biological activity in the protection against various types of injury. However, information on its role in tacrolimus (TAC)-induced renal injury is limited. OBJECTIVES: This study aimed to assess the beneficial effects of pancreatic kininogenase (PK) in a rat model of chronic TAC nephrotoxicity and in vitro. METHODS: Sprague Dawley rats were treated daily with either TAC or PK or a combination of the two for four weeks. The influence of PK on renal injury was examined in terms of renal function, histopathology, cytokine expression, oxidative stress, intracellular organelles, programmed cell death, and PI3K/AKT signaling. Human kidney proximal tubular (HK-2) cells and mouse mesangial (SV40 MES13) cells treated with TAC and PK were also studied. RESULTS: PK treatment improved renal function and histopathology. This effect was paralleled by downregulation of proinflammatory and profibrotic cytokine expression. TAC-induced oxidative stress was closely associated with endoplasmic reticulum stress and mitochondrial dysfunction, resulting in excessive programmed cell death (apoptosis and autophagy) that was significantly abrogated by concurrent PK interference with PI3K/AKT signaling. PK also stimulated bradykinin receptor 1 (B1R) and B2R mRNA synthesis and increased bioactive nitric oxide (NO) and cAMP concentrations in TAC-treated kidneys. Blockade of either B1R or B2R eliminated the renoprotective effects of PK. In HK-2 and SV40 MES13 cells, PK decreased TAC-induced overproduction of intracellular reactive oxygen species and inhibited apoptotic cells, whereas cell viability was improved. Moreover, activated PI3K/AKT signaling in HK-2 cells was inhibited by PK and the PI3K inhibitor, LY294002. CONCLUSIONS: These findings indicate that PK treatment protects against chronic TAC nephrotoxicity via inhibition of PI3K/AKT signaling.


Phosphatidylinositol 3-Kinases , Tacrolimus , Animals , Apoptosis , Kidney , Mice , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Bradykinin/metabolism , Tacrolimus/pharmacology , Tissue Kallikreins/metabolism , Tissue Kallikreins/pharmacology
4.
Oxid Med Cell Longev ; 2020: 6834236, 2020.
Article En | MEDLINE | ID: mdl-32190176

OBJECTIVE: To investigate the protective effects and mechanisms of human tissue kallikrein 1 (hKLK1) on type 1 diabetes mellitus- (DM-) induced erectile dysfunction in rats. Materials and Methods. The homozygous transgenic rats (TGR) harboring the hKLK1 gene and age-matched wild-type Sprague Dawley rats (WTR) were involved, and intraperitoneal injection of streptozotocin was utilized to induce diabetes in rats. Forty-eight-week-old male rats were randomly divided into a WTR group, TGR group, diabetic WTR group (WTDM), diabetic TGR group (TGDM), and TGDM with HOE140 group (TGDMH), with eight rats in each group. Twelve weeks later, the erectile response of all rats was detected by cavernous nerve electric stimulation, and corpus cavernosums were harvested to evaluate the levels of cavernous oxidative stress (OS), apoptosis, fibrosis, and involved pathways. Moreover, cavernous smooth muscle cells (CSMC) and endothelial cells (EC) were primarily isolated to build a coculture system for a series of in vitro verification. RESULTS: The hKLK1 gene and age-matched wild-type Sprague Dawley rats (WTR) were involved, and intraperitoneal injection of streptozotocin was utilized to induce diabetes in rats. Forty-eight-week-old male rats were randomly divided into a WTR group, TGR group, diabetic WTR group (WTDM), diabetic TGR group (TGDM), and TGDM with HOE140 group (TGDMH), with eight rats in each group. Twelve weeks later, the erectile response of all rats was detected by cavernous nerve electric stimulation, and corpus cavernosums were harvested to evaluate the levels of cavernous oxidative stress (OS), apoptosis, fibrosis, and involved pathways. Moreover, cavernous smooth muscle cells (CSMC) and endothelial cells (EC) were primarily isolated to build a coculture system for a series of. CONCLUSIONS: hKLK1 preserves erectile function of DM rats through its antitissue excessive OS, apoptosis, and fibrosis effects, as well as activation of the PI3K/AKT/eNOS/cGMP pathway in the penis. Moreover, hKLK1 promotes relaxation and prevents high glucose-induced injuries of CSMC mediated by EC-CSMC crosstalk.


Diabetes Mellitus, Experimental/complications , Erectile Dysfunction/drug therapy , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tissue Kallikreins/therapeutic use , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Body Weight , Calcium/metabolism , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental/pathology , Electric Stimulation , Erectile Dysfunction/complications , Erectile Dysfunction/pathology , Fasting/blood , Fibrosis , Glucose/toxicity , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , Penis/drug effects , Penis/pathology , Rats, Sprague-Dawley , Rats, Transgenic , Signal Transduction/drug effects , Streptozocin , Tissue Kallikreins/pharmacology
5.
Pain Res Manag ; 2019: 6393150, 2019.
Article En | MEDLINE | ID: mdl-31428213

Migraine is one of the most common neurological disorders which poses significant socioeconomic burden worldwide. Neuroinflammation and oxidative stress both play important roles in the pathogenesis of migraine. Human urinary kallidinogenase (UK) is a tissue kallikrein derived from human urine. Increasing evidence suggests that UK may protect against ischemic stroke, but UK's treatment potential against migraine remains to be explored. Immortal BV-2 murine microglial cells were treated with UK (125 nM, 250 nM, and 500 nM) and then given lipopolysaccharides (LPS, 1000 ng/mL). Cell viability of BV-2 cells was tested by the CCK-8 assay. Expressions of tumor necrosis factor-α (TNFα), prostaglandin E2 (PGE2), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were examined with the ELISA method and western blot. Intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) were measured to determine oxidative stress. Our results showed that LPS administration increased the levels of proinflammatory cytokines (TNFα, PGE2, IL-6, and IL-1ß) and oxidative stress (ROS and MDA) when compared with the control group and decreased significantly upon introduction with UK. Taken together, UK treatment reduced LPS-induced neuroinflammation and oxidative stress in a dose-dependent manner, which might be a potential treatment of migraine.


Microglia/drug effects , Oxidative Stress/drug effects , Tissue Kallikreins/pharmacology , Animals , Cell Survival/drug effects , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , Migraine Disorders/metabolism , Migraine Disorders/physiopathology
6.
J Neurochem ; 139(2): 208-220, 2016 10.
Article En | MEDLINE | ID: mdl-27248356

Previous studies have demonstrated that tissue kallikrein (TK) protects against cerebral ischemia injury mainly through inhibition of apoptosis via bradykinin B2 receptor (B2R). In this study, we proposed that autophagy induction contributed to the neuroprotective mechanism of TK. To validate this hypothesis, we investigated TK-induced autophagy and its signaling mechanisms in human SH-SY5Y cells exposed to oxygen and glucose deprivation (OGD). We found that TK treatment enhanced autophagy induction, reflected by augmented LC3 conversion and Beclin1 expression, decreased p62 levels and increased monomeric red fluorescent protein-LC3 puncta formation. Green fluorescent protein-monomeric red fluorescent protein-LC3 adenovirus assay indicated that TK maintained autophagic flux. Moreover, bafilomycin A1 (Baf.A1) caused obvious LC3-II accumulation either in the presence or absence of TK. Autophagy inhibition by Beclin1 knockdown or Baf.A1 treatment abrogated the neuroprotective effects of TK. Mitogen-activated protein kinase kinase 1/2 (MEK1/2)/extracellular signal-regulated kinase (ERK)1/2 and AMP-activated protein kinase (AMPK)/tuberous sclerosis complex 2 (TSC2)/mammalian target of rapamycin (mTOR) signaling were induced by OGD stress and enhanced by TK. MEK/ERK inhibitor U0126 alone elevated autophagy in OGD conditions, but impaired TK-induced autophagy. Blockade of AMPK/TSC2/mTOR signaling by AMPK inhibitor compound C and shRNA mediated the knockdown of AMPK α1 and TSC2 but abolished autophagy in SH-SY5Y cells exposed to OGD treated either with or without TK. Moreover, B2R expression was up-regulated by OGD exposure. B2R knockdown attenuated autophagy and suppressed MEK1/2/ERK1/2 and AMPK/TSC2/mTOR signaling in OGD conditions in either the presence or absence of TK. In sum, we revealed the significance of B2R-mediated MEK/ERK and AMPK signaling in autophagy induction under OGD stress, and proposed novel mechanisms involved in the neuropotective function of TK through B2R-dependent regulation of autophagy. We propose the depicted model for the neuroprotective mechanism of tissue kallikrein (TK) during OGD stress: TK enhances bradykinin B2 receptor (B2R)-mediated MEK1/2/ERK1/2 and AMPK/TSC2/mTOR signaling, thus inducing protective autophagy. The findings reported in this study should provide new evidence for the pro-survival role of B2R-mediated autophagy in cerebral ischemia.


Autophagy/drug effects , Cell Hypoxia/drug effects , Glucose/deficiency , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptor, Bradykinin B2/drug effects , Tissue Kallikreins/pharmacology , Beclin-1/biosynthesis , Cell Survival/drug effects , Humans , MAP Kinase Signaling System/drug effects , Microtubule-Associated Proteins/biosynthesis , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics
7.
Mol Med Rep ; 13(2): 1389-94, 2016 Feb.
Article En | MEDLINE | ID: mdl-26677174

Previous studies by our group showed that tissue kallikrein (TK) exerts neuroprotective effects during cerebral ischemia. Autophagy is an important adaptive response to cellular stress during nutrient deprivation, and ß-catenin in known to repress autophagy. The present study investigated the possible involvement of autophagy and ß-catenin signaling in the protective effects of TK under nutrient deprivation-induced stress conditions. TK was shown to promote the survival and inhibit the death of SH-SY5Y cells under serum starvation and enhanced autophagic activity in a concentration-dependent manner, as indicated by augmented light chain (LC)3-II levels and Beclin-1 expression. The autophagy inhibitors 3-methyladenine and NH4Cl abolished the protective effects of TK. Of note, although serum starvation alone and TK treatment increased p62 protein levels and mRNA expression, incubation with the lysosome inhibitor NH4Cl increased the accumulation of LC3-II and p62 protein, indicating normal autophagic flux. It was also observed that ß-catenin expression was significantly downregulated by TK treatment. TK stimulated the interaction between LC3 and ß-catenin, and NH4Cl abolished the effects of TK on ß-catenin levels in serum-starved cells, suggesting the autophagic degradation of ß-catenin, which may have led to the enhancement of autophagy. In conclusion, the findings of the present study demonstrated that TK promoted cell survival and ß-catenin degradation in serum-starved SH-SY5Y cells via increasing autophagy, which indicated the therapeutic potential of TK under nutrient deprivation-associated stress conditions.


Autophagy/drug effects , Proteolysis/drug effects , Tissue Kallikreins/pharmacology , beta Catenin/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Culture Media, Serum-Free , Humans , RNA-Binding Proteins/metabolism , Up-Regulation/drug effects
8.
PLoS One ; 9(9): e107213, 2014.
Article En | MEDLINE | ID: mdl-25259810

The kallikrein-kinin system (KKS) comprises a cascade of proteolytic enzymes and biogenic peptides that regulate several physiological processes. Over-expression of tissue kallikrein-1 and modulation of the KKS shows beneficial effects on insulin sensitivity and other parameters relevant to type 2 diabetes mellitus. However, much less is known about the role of kallikreins, in particular tissue kallikrein-1, in type 1 diabetes mellitus (T1D). We report that chronic administration of recombinant human tissue kallikrein-1 protein (DM199) to non-obese diabetic mice delayed the onset of T1D, attenuated the degree of insulitis, and improved pancreatic beta cell mass in a dose- and treatment frequency-dependent manner. Suppression of the autoimmune reaction against pancreatic beta cells was evidenced by a reduction in the relative numbers of infiltrating cytotoxic lymphocytes and an increase in the relative numbers of regulatory T cells in the pancreas and pancreatic lymph nodes. These effects may be due in part to a DM199 treatment-dependent increase in active TGF-beta1. Treatment with DM199 also resulted in elevated C-peptide levels, elevated glucagon like peptide-1 levels and a reduction in dipeptidyl peptidase-4 activity. Overall, the data suggest that DM199 may have a beneficial effect on T1D by attenuating the autoimmune reaction and improving beta cell health.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/immunology , Immunomodulation/drug effects , Recombinant Proteins/pharmacology , Tissue Kallikreins/pharmacology , Animals , Autoimmunity/drug effects , Biomarkers , Blood Glucose/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/drug therapy , Female , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Recombinant Proteins/administration & dosage , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tissue Kallikreins/administration & dosage , Transforming Growth Factor beta1/metabolism
9.
PLoS One ; 9(8): e103981, 2014.
Article En | MEDLINE | ID: mdl-25100328

Modulation of the kallikrein-kinin system (KKS) has been shown to have beneficial effects on glucose homeostasis and several other physiological responses relevant to the progression of type 2 diabetes mellitus (T2D). The importance of bradykinin and its receptors in mediating these responses is well documented, but the role of tissue kallikrein-1, the protease that generates bradykinin in situ, is much less understood. We developed and tested DM199, recombinant human tissue kallikrein-1 protein (rhKLK-1), as a potential novel therapeutic for T2D. Hyperinsulinemic-euglycemic clamp studies suggest that DM199 increases whole body glucose disposal in non-diabetic rats. Single-dose administration of DM199 in obese db/db mice and ZDF rats, showed an acute, dose-dependent improvement in whole-body glucose utilization. Sub-acute dosing for a week in ZDF rats improved glucose utilization, with a concomitant rise in fasting insulin levels and HOMA1-%B scores. After cessation of sub-acute dosing, fasting blood glucose levels were significantly lower in ZDF rats during a drug wash-out period. Our studies show for the first time that DM199 administration results in acute anti-hyperglycemic effects in several preclinical models, and demonstrate the potential for further development of DM199 as a novel therapeutic for T2D.


Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacokinetics , Tissue Kallikreins/pharmacology , Animals , Blood Glucose , CHO Cells , Cricetinae , Cricetulus , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Mice, Obese , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Time Factors
10.
Eur J Pharmacol ; 738: 74-82, 2014 Sep 05.
Article En | MEDLINE | ID: mdl-24877690

Ocular ischemic syndrome is likely stem from retinal ischemia, and which causes visual disorder. The pathological mechanism of ocular ischemic syndrome is still unknown, therefore the optimal treatment for ocular ischemic syndrome remains to be established. Then, this study aimed to evaluate the effects of tissue-derived kallidinogenase in retinal ischemia protection in mice. In the present study, the effects of tissue-derived kallidinogenase (1 or 10 µg/kg, i.v.) on ischemia/reperfusion-induced retinal damage in mice were examined by histological, electrophysiological, and permeability analyses. In addition, we assessed phosphorylation of endothelial nitric oxide synthase (eNOS) and nuclear factor-kappa B (NF-κB), which is closely-involved in ischemic injury and permeability. Moreover, the neuroprotective effect of kallidinogenase in an in vitro model of ischemia induced by oxygen-glucose deprivation or hypoxia was examined. The results indicated that kallidinogenase significantly prevented the decrease in ganglion cell number induced by ischemia/reperfusion. Electroretinogram measurements showed that kallidinogenase significantly prevented the ischemia/reperfusion-induced reductions in a- and b-wave amplitudes seen 5 days after ischemia/reperfusion. Moreover, kallidinogenase significantly inhibited the permeability increase induced by ischemia/reperfusion. Similar to the results in vivo, kallidinogenase significantly inhibited the retinal ganglion cell death induced by oxygen-glucose deprivation. Also, kallidinogenase significantly suppressed the hypoxia-induced increase in permeability. However, these effects observed in vitro disappeared when an eNOS inhibitor was used concurrently. These findings suggest that kallidinogenase may prevent ischemia/reperfusion-induced retinal damage, might be through eNOS activation.


Ischemia/prevention & control , Retina/drug effects , Retina/physiopathology , Tissue Kallikreins/pharmacology , Animals , Cell Hypoxia/drug effects , Enzyme Activation/drug effects , Male , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxygen/metabolism , Permeability/drug effects , Phosphorylation/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
11.
Cell Signal ; 26(2): 220-32, 2014 Feb.
Article En | MEDLINE | ID: mdl-24211626

Tissue kallikrein (TK) was previously shown to take most of its biological effects through bradykinin receptors. In this study, we assumed that TK mediated neurite outgrowth was independent of bradykinin receptors. To test the hypothesis, we investigated TK-induced neurite outgrowth and its signaling mechanisms in cultured primary neurons and human SH-SY5Y cells. We found that TK stimulation could increase the number of processes and mean process length of primary neurons, which were blocked by epidermal growth factor receptor (EGFR) inhibitor or down-regulation, small interfering RNA for flotillin-2 and extracellular signal-regulated kinase (ERK) 1/2 inhibitor. Moreover, TK-induced neurite outgrowth was associated with EGFR and ERK1/2 activation, which were inhibited by EGFR antagonist or RNA interference and flotillin-2 knockdown. Interestingly, inhibition of bradykinin receptors had no significant effects on EGFR and ERK1/2 phosphorylation. In the present research, our data also suggested that EGFR and flotillin-2 formed constitutive complex that translocated to around the nuclei in the TK stimulation. In sum, our findings provided evidence that TK could promote neurite outgrowth via EGFR, flotillin-2 and ERK1/2 signaling pathway in vitro.


ErbB Receptors/metabolism , Membrane Proteins/metabolism , Neurites/drug effects , Tissue Kallikreins/pharmacology , Animals , Cells, Cultured , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Quinazolines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology
12.
Regul Pept ; 181: 30-6, 2013 Feb 10.
Article En | MEDLINE | ID: mdl-23318501

In order to understand the mechanisms of interaction between tonin-angiotensin and renin-angiotensin systems (RAS) we evaluated, "in vivo" and "in vitro", in Wistar rats, cardiovascular and electrocardiographic parameters after tonin administration. Arterial pressure (AP) and electrocardiogram (ECG) were recorded in awake animals before and after tonin administration. Langendorff technique was used to analyze cardiac function in isolated heart in the presence of tonin and video motion edge detection system was used to evaluate the effect of tonin upon contractile function of isolated rat ventricular cardiomyocytes. After tonin infusion rats presented significantly higher diastolic and mean arterial pressure (MAP) and heart rate (HR) as compared with control. The ECG analysis revealed shorter RR interval, increase in the low-frequency (LF) range of the heart rate variability (HRV) power (%) and decrease in the high-frequency (HF) of HRV power (%). Isolated hearts perfused with tonin presented an increase in the arterial coronary pressure (ACP) and decline in the ventricular systolic tension (ST), maximal (dT/dt+) and minimal (dT/dt) contractility. The rates of contraction and relaxation of isolated ventricular cardiomyocytes were significantly increased due to the presence of tonin. The angiotensin II (Ang II) levels in the coronary sinus effluent increased in the presence of tonin in a dose-dependent manner and the effect of tonin upon ACP was completely blocked by candesartan. Tonin is able to generate the vasoconstrictor peptide Ang II in the isolated heart of the rat and the cardiovascular response induced by tonin was completely blocked by candesartan, an indication that the action of Ang II on Ang II type 1 (AT1) receptors is the major mechanism of the heart effects. Tonin affects cardiomyocyte contractile function which may be due to interference with Ca(2+) handling.


Angiotensin II/metabolism , Heart/drug effects , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/drug effects , Tissue Kallikreins/pharmacology , Angiotensin II/agonists , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arterial Pressure/drug effects , Benzimidazoles/pharmacology , Biphenyl Compounds , Calcium/metabolism , Cells, Cultured , Electrocardiography , Heart Rate/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/agonists , Tetrazoles/pharmacology
13.
Cell Signal ; 24(11): 2205-15, 2012 Nov.
Article En | MEDLINE | ID: mdl-22575735

Previous studies have demonstrated that human tissue kallikrein (TK) gene delivery protects against mouse cerebral ischemia/reperfusion (I/R) injury through bradykinin B2 receptor (B2R) activation. We have also reported that exogenous TK administration can suppress glutamate- or acidosis-induced neurotoxicity through the extracellular signal-regulated kinase1/2 (ERK1/2) pathway. To further explore the neuroprotection mechanisms of TK, in the present study we performed immunoprecipitation analysis and identified a scaffolding protein Homer1b/c using MALDI-TOF MS analysis. Here, we tested the hypothesis that TK reduces cell injury induced by oxygen and glucose deprivation/reoxygenation (OGD/R) through activating Homer1b/c. We found that TK increased the expression of Homer1b/c in a concentration- and time-dependent manner. Moreover, TK facilitated the translocation of Homer1b/c to the plasma membrane under OGD/R condition by confocal microscope assays. We also observed that overexpression of Homer1b/c showed the neuroprotection against OGD/R-induced cell injury by enhancing cell survival, reducing LDH release, caspase-3 activity and cell apoptosis. However, the knockdown of Homer1b/c by small interfering RNA showed the opposite effects, indicating that Homer1b/c had protective effects against OGD/R-induced neuronal injury. More interestingly, TK exerted its much more significantly neuroprotective effects after Homer1b/c overexpression, whereas it exerted its reduced effects after Homer1b/c knockdown. In addition, TK pretreatment increased the phosphorylation of the ERK1/2 and Akt-GSK3ß through Homer1b/c activation. The beneficial effects of Homer1b/c were abolished by the ERK1/2 or PI3K antagonist. Therefore, we propose novel signaling mechanisms involved in the anti-hypoxic function of TK through activation of Homer1b/c-ERK1/2 and Homer1b/c-PI3K-Akt signaling pathways.


Carrier Proteins/metabolism , Cell Hypoxia/drug effects , Tissue Kallikreins/pharmacology , Apoptosis , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Caspase 3/metabolism , Cell Line , Cell Membrane/metabolism , Flavonoids/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homer Scaffolding Proteins , Humans , Immunoprecipitation , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Up-Regulation/drug effects
14.
Biochem Biophys Res Commun ; 407(2): 283-7, 2011 Apr 08.
Article En | MEDLINE | ID: mdl-21376701

Systemic or local delivery of human tissue kallikrein gene (hTK) has been shown to be an effective strategy to alleviate cerebral ischemia/reperfusion (I/R) injury, and tissue kallikrein (TK) administration can suppress glutamate- or acidosis-mediated neurotoxicity in vitro. In the present study, the role of TK in hypoxia/reoxygenation (H/R) induced neuronal cell death was investigated. We found that TK administration could remarkably alleviate H/R-induced neuronal injury by reduction of LDH release and promotion of neuron viability. The protective effects of TK could be counteracted by bradykinin B2 receptor (B2R) antagonist HOE140, which could suppress up-regulation of TK on the ERK signal pathway under H/R condition. These results indicate that TK plays an important role in preventing neurons from H/R damage at least partially through the TK-B2R-ERK1/2 pathway.


Brain Ischemia/pathology , Cerebral Cortex/pathology , Cytoprotection , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/pathology , Reperfusion Injury/pathology , Tissue Kallikreins/physiology , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Brain Ischemia/enzymology , Cells, Cultured , Cerebral Cortex/enzymology , Humans , Neurons/enzymology , Rats , Reperfusion Injury/enzymology , Tissue Kallikreins/genetics , Tissue Kallikreins/pharmacology
15.
Exp Cell Res ; 316(3): 376-89, 2010 Feb 01.
Article En | MEDLINE | ID: mdl-19879874

Biological functions of tissue kallikrein (TK, KLK1) are mainly mediated by kinin generation and subsequent kinin B2 receptor activation. In this study, we investigated the potential role of TK and its signaling pathways in cultured human keratinocyte migration and in a rat skin wound healing model. Herein, we show that TK promoted cell migration and proliferation in a concentration- and time-dependent manner. Inactive TK or kinin had no significant effect on cell migration. Interestingly, cell migration induced by active TK was not blocked by icatibant or L-NAME, indicating an event independent of kinin B2 receptor and nitric oxide formation. TK's stimulatory effect on cell migration was inhibited by small interfering RNA for proteinase-activated receptor 1 (PAR(1)), and by PAR(1) inhibitor. TK-induced migration was associated with increased phosphorylation of epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK), which was blocked by inhibition of protein kinase C (PKC), Src, EGFR and ERK. TK-induced cell migration and EGFR phosphorylation were blocked by metalloproteinase (MMP) inhibitor, heparin, and antibodies against EGFR external domain, heparin-binding EGF-like growth factor (HB-EGF) and amphiregulin (AR). Local application of TK promoted skin wound healing in rats, whereas icatibant and EGFR inhibitor blocked TK's effect. Skin wound healing was further delayed by aprotinin and neutralizing TK antibody. This study demonstrates a novel role of TK in skin wound healing and uncovers new signaling pathways mediated by TK in promoting keratinocyte migration through activation of the PAR(1)-PKC-Src-MMP pathway and HB-EGF/AR shedding-dependent EGFR transactivation.


Cell Movement/drug effects , ErbB Receptors/metabolism , Keratinocytes/cytology , Keratinocytes/enzymology , Receptor, PAR-1/metabolism , Signal Transduction/drug effects , Tissue Kallikreins/pharmacology , Animals , Cell Proliferation/drug effects , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Keratinocytes/drug effects , Kinins/metabolism , Ligands , Matrix Metalloproteinases/metabolism , Nitric Oxide/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Receptor, Bradykinin B2/metabolism , Skin/drug effects , Skin/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Wound Healing/drug effects
16.
Exp Neurol ; 219(2): 453-65, 2009 Oct.
Article En | MEDLINE | ID: mdl-19576887

Human tissue kallikrein (hTK) gene transfer has been shown to protect neurons against cerebral ischemia/reperfusion (I/R) injury, and exogenous tissue kallikrein (TK) administration can enhance neurogenesis and angiogenesis following focal cortical infarction. Previous studies have reported that acidosis is a common feature of ischemia and plays a critical role in brain injury. However, little is known about the role of TK in ischemia-acidosis-induced injury, which is partially caused by the activation of acid-sensing ion channels (ASICs). Here we report that pretreatment of cultured cortical neurons with TK reduced cell death induced by either acidosis or oxygen and glucose deprivation-acidosis/reoxygenation (OGD-A/R). Immunocytochemical staining revealed that TK largely prevented OGD-A/R-induced neuronal morphological changes. We also observed that TK treatment protected cultured neurons from acidosis and OGD-A/R insults. TK exerted the neuroprotective effects by reducing production of reactive oxygen species (ROS), stabilizing the mitochondrial membrane potential (MMP) and inhibiting caspase-3 activation, and thereby attenuating oxidative stress and apoptosis. In addition, we found that activation of the extracellular signal-regulated kinase1/2 (ERK1/2) signaling cascade but not the PI3K/Akt signaling pathway was required for the survival-promoting effect of TK on neurons exposed to OGD-A/R. Moreover, blockade of ASICs had effects similar to TK administration, suggesting direct or indirect involvement of ASICs in TK protection. In conclusion, TK has antioxidant characteristics and is capable of alleviating ischemia-acidosis/reperfusion-induced injury, inhibiting apoptosis and promoting cell survival in vitro through activating the ERK1/2 signaling pathways. Therefore, TK represents a promising therapeutic strategy for ischemic stroke.


Cerebral Cortex/cytology , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tissue Kallikreins/pharmacology , Acid Sensing Ion Channels , Acidosis/drug therapy , Analysis of Variance , Animals , Animals, Newborn , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Central Nervous System Stimulants/toxicity , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glucose/deficiency , Humans , Hydrogen-Ion Concentration , Hypoxia/drug therapy , In Situ Nick-End Labeling/methods , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Picrotoxin/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sodium Channels/metabolism , Urine/chemistry
17.
Cardiovasc Res ; 80(3): 354-64, 2008 Dec 01.
Article En | MEDLINE | ID: mdl-18689794

AIMS: We investigated the role of the Akt-glycogen synthase kinase (GSK)-3beta signalling pathway in mediating the protective effects of tissue kallikrein on myocardial injury by promoting angiogenesis and blood flow in rats after myocardial infarction (MI). METHODS AND RESULTS: Human tissue kallikrein gene in an adenoviral vector, with or without co-administration of dominant-negative Akt (Ad.DN-Akt) or constitutively active GSK-3beta (Ad.GSK-3betaS9A), was injected into rat myocardium after MI. The expression of recombinant human kallikrein in rat heart significantly improved cardiac function and reduced infarct size 10 days after gene delivery. Kallikrein administration significantly increased myocardial blood flow as well as capillary and arteriole densities in the infarcted myocardium. Kallikrein increased cardiac Akt and GSK-3beta phosphorylation in conjunction with decreased GSK-3beta activity and the upregulation of vascular endothelial growth factor (VEGF) and VEGF receptor-2 (VEGFR-2). All of kallikrein's effects on the myocardium were abrogated by Ad.DN-Akt and Ad.GSK-3betaS9A. Moreover, in cultured human aortic endothelial cells, tissue kallikrein stimulated capillary tube formation and promoted cell migration; however, these effects were blocked by Ad.DN-Akt, Ad.GSK-3betaS9A, icatibant (a kinin B2 receptor antagonist), Tki (a VEGF receptor tyrosine kinase inhibitor), and a neutralizing VEGF antibody. In addition, tissue kallikrein decreased GSK-3beta activity via the phosphatidylinositol 3-kinase-Akt pathway and enhanced VEGF and VEGFR-2 expression in endothelial cells. CONCLUSION: These data provide the first direct evidence that tissue kallikrein protects against acute-phase MI by promoting neovascularization, restoring regional blood flow and improving cardiac function through the kinin B2 receptor-Akt-GSK-3beta and VEGF signalling pathways.


Glycogen Synthase Kinase 3/metabolism , Heart/drug effects , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tissue Kallikreins/metabolism , Tissue Kallikreins/pharmacology , Adenoviridae/genetics , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Glycogen Synthase Kinase 3 beta , Heart/physiology , Humans , Male , Myocardial Infarction/metabolism , Myocardium/metabolism , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Tissue Kallikreins/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Biol Chem ; 389(6): 725-30, 2008 Jun.
Article En | MEDLINE | ID: mdl-18627294

Adenovirus-mediated kallikrein delivery has been shown to promote blood vessel growth in the limb under both ischemic and normoperfused conditions. Here we investigated whether a continuous supply of kallikrein and kinin peptide can induce neovascularization in a rat model of hindlimb ischemia. Rats underwent femoral artery ligation and localized injection of tissue kallikrein, bradykinin or B1 receptor agonist, followed by infusion of proteins by osmotic minipump. Regional blood flow was monitored weekly by laser Doppler perfusion imaging. Three weeks after surgery, rats receiving kallikrein and kinins showed a significant increase in the perfusion ratio of ischemic vs. normoperfused limb compared to control rats. Similarly, a microsphere assay showed that kallikrein and kinins significantly increased regional blood flow without altering blood pressure. Moreover, kallikrein and kinins significantly augmented capillary and arteriole densities, as quantified by immunostaining with CD-31 and smooth muscle alpha-actin. Both tissue kallikrein and bradykinin increased hemoglobin content in Matrigel implants in mice, providing further evidence of the angiogenic properties. Kinins, when delivered subcutaneously via Matrigel in rats, also increased regional perfusion. This is the first demonstration that local application of tissue kallikrein protein or kinin peptide has therapeutic value in the treatment of ischemic disease by promoting neovascularization.


Hindlimb/blood supply , Hindlimb/physiopathology , Ischemia/physiopathology , Kinins/pharmacology , Neovascularization, Physiologic/drug effects , Tissue Kallikreins/pharmacology , Animals , Arterioles/growth & development , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , Capillaries/growth & development , Collagen , Drug Combinations , Hemoglobins/metabolism , Hindlimb/drug effects , Injections , Kinins/administration & dosage , Laminin , Lasers , Male , Microspheres , Peptides/administration & dosage , Peptides/pharmacology , Perfusion , Proteoglycans , Rats , Tissue Kallikreins/administration & dosage
19.
Mol Cancer Res ; 6(6): 1043-51, 2008 Jun.
Article En | MEDLINE | ID: mdl-18567807

A major characteristic of prostate cancer is the elevation of serum levels of prostate-specific antigen (hK3) and hK2, which are tumor markers that correlate with advancing stages of disease. Including hK4, these three kallikrein serine proteases are almost exclusively produced by the prostate. Prostate cancer cells have been recently shown to overexpress protease-activated receptors (PAR), which can be potentially activated by kallikreins and can regulate tumor growth. Here, we show that recombinant hK2 and hK4 activate ERK1/2 signaling of DU-145, PC-3, and LNCaP prostate cancer cells, which express both PAR1 and PAR2. These kallikreins also stimulate the proliferation of DU-145 cells. Pretreatment of hK2 and hK4 with the serine protease inhibitor, aprotinin, blocks the responses in DU-145 cells, and small interfering RNA against PAR1 and PAR2 also inhibits ERK1/2 signaling. To determine which PAR is activated by hK2 and hK4, a cell line that expresses a single PAR, a PAR1 knockout mouse lung fibroblast cell line transfected with PAR1 (KOLF-PAR1) or PAR2 (KOLF-PAR2) was used. hK4 activates both PAR1 and PAR2, whereas hK2 activates PAR2. hK4 generates more phosphorylated ERK1/2 than hK2. These data indicate that prostatic kallikreins (hK2 and hK4) directly stimulate prostate cancer cell proliferation through PAR1 and/or PAR2 and may be potentially important targets for future drug therapy for prostate cancer.


Kallikreins/pharmacology , Prostatic Neoplasms/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Tissue Kallikreins/pharmacology , Animals , Aprotinin/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , MAP Kinase Signaling System , Male , Mice , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-2/agonists , Receptor, PAR-2/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Tissue Kallikreins/antagonists & inhibitors , Tissue Kallikreins/metabolism
20.
Regul Pept ; 140(1-2): 12-20, 2007 Apr 05.
Article En | MEDLINE | ID: mdl-17196272

We investigated the effect of tissue kallikrein infusion on cardiac protection at acute and sub-acute phases after myocardial infarction (MI). Immediately after MI, rats were infused with purified tissue kallikrein, with or without icatibant (a kinin B2 receptor antagonist). Intramyocardial injection of kallikrein reduced myocardial infarct size and inhibited cardiomyocyte apoptosis at 1 day after MI associated with increased nitric oxide levels, Akt and glycogen synthase kinase-3beta phosphorylation and decreased caspase-3 activation. Kallikrein infusion for 7 days improved cardiac function, normalized left ventricular wall thickness and decreased monocyte/macrophage infiltration in the infarct heart. Kallikrein treatment reduced NADH oxidase expression and activity, superoxide formation and malondialdehyde levels, and reduced MAPK and Ikappa-Balpha phosphorylation, NF-kappaB activation and MCP-1 and VCAM-1 expression. Kallikrein's effects were all blocked by icatibant. These results indicate that kallikrein through kinin B2 receptor activation prevents apoptosis, inflammation and ventricular remodeling by increased nitric oxide formation and suppression of oxidative stress-mediated signaling pathways.


Apoptosis/drug effects , Inflammation/drug therapy , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Tissue Kallikreins/pharmacology , Ventricular Remodeling/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Cell Nucleus/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Polymerase Chain Reaction/methods , Protein Transport/drug effects , Rats , Rats, Wistar , Tissue Kallikreins/administration & dosage , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
...