Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Br J Dermatol ; 190(1): 70-79, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37672660

ABSTRACT

BACKGROUND: Multiple treatment options are available for the management of psoriasis, but clinical response varies among individual patients and no biomarkers are available to facilitate treatment selection for improved patient outcomes. OBJECTIVES: To utilize retrospective data to conduct a pharmacogenetic study to explore the potential genetic pathways associated with drug response in the treatment of psoriasis. METHODS: We conducted a retrospective pharmacogenetic study using self-evaluated treatment response from 1942 genotyped patients with psoriasis. We examined 6 502 658 genetic markers to model their associations with response to six treatment options using linear regression, adjusting for cohort variables and demographic features. We further utilized an integrative approach incorporating epigenomics, transcriptomics and a longitudinal clinical cohort to provide biological implications for the topmost signals associated with drug response. RESULTS: Two novel markers were revealed to be associated with treatment response: rs1991820 (P = 1.30 × 10-6) for anti-tumour necrosis factor (TNF) biologics; and rs62264137 (P = 2.94 × 10-6) for methotrexate, which was also associated with cutaneous mRNA expression levels of two known psoriasis-related genes KLK7 (P = 1.0 × 10-12) and CD200 (P = 5.4 × 10-6). We demonstrated that KLK7 expression was increased in the psoriatic epidermis, as shown by immunohistochemistry, as well as single-cell RNA sequencing, and its responsiveness to anti-TNF treatment was highlighted. By inhibiting the expression of KLK7, we further illustrated that keratinocytes have decreased proinflammatory responses to TNF. CONCLUSIONS: Our study implicates the genetic regulation of cytokine responses in predicting clinical drug response and supports the association between pharmacogenetic loci and anti-TNF response, as shown here for KLK7.


Subject(s)
Psoriasis , Humans , Kallikreins/genetics , Kallikreins/therapeutic use , Pharmacogenetics , Pharmacogenomic Testing , Psoriasis/drug therapy , Psoriasis/genetics , Psoriasis/pathology , Retrospective Studies , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/genetics
2.
Tissue Cell ; 78: 101874, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994918

ABSTRACT

Right ventricular (RV) hypertrophy and further heart failure are major co-morbidities, resulting in the premature death of patients with hypoxic pulmonary hypertension (HPH). The regulatory effects of kallikrein-related peptidase (KLK) family members on cardiac function have been extensively studied. However, to the best of the authors' knowledge, the regulatory effects of KLK8 on RV hypertrophy caused by HPH have yet to be reported. The aim of the present study was to assess KLK8 expression in the RV tissue of HPH-modeled rats, and to further explore the effects and underlying mechanism of KLK8 in regulating the hypertrophy of hypoxia-induced H9c2 cardiomyocytes. In HPH model rats, increases in the right ventricle hypertrophy index, the right ventricular systolic pressure, cardiac output, as well as pulmonary artery wall thickness were observed. Western blot analysis revealed that KLK8 expression and MAPK/p53 signaling activity were enhanced in the RVs of rats in an RV HPH rat model. In hypoxia-induced H9c2 cardiomyocytes, KLK8 overexpression promoted cardiomyocyte hypertrophy, whereas KLK8 silencing showed the opposite results. KLK8 overexpression increased the expression levels of ventricular hypertrophy markers, including atrial natriuretic peptide, brain natriuretic peptide and myosin heavy chain 7, which were blocked upon addition of the p38 MAPK inhibitor, SB202190. Conversely, KLK8 silencing caused a decrease in the expression levels of the ventricular hypertrophy markers, which were further reduced via inhibition of the p38 MAPK/p53 signaling pathway. Taken together, the results of the present study have shown that KLK8 may subtly regulate RV hypertrophy, and therefore KLK8 may be a promising therapeutic target for treating HPH-induced RV hypertrophy.


Subject(s)
Atrial Natriuretic Factor , Hypertrophy, Right Ventricular , Animals , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/etiology , Hypoxia/metabolism , Kallikreins/metabolism , Kallikreins/pharmacology , Kallikreins/therapeutic use , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/pharmacology , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/pharmacology , Natriuretic Peptide, Brain/therapeutic use , Rats , Serine Endopeptidases/metabolism , Serine Endopeptidases/pharmacology , Serine Endopeptidases/therapeutic use , Signal Transduction , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
J Integr Neurosci ; 21(3): 80, 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35633161

ABSTRACT

BACKGROUND: Post-stroke cognitive impairment (PSCI) is a common symptom of stroke and affects the quality of life and prognosis of stroke survivors. In our study, we evaluated the efficacy of Human urinary kallidinogenase (HUK) on cognitive function in acute ischemic stroke (AIS) patients, and discussed the role of cystatin C (CysC) in improving PSCI. METHODS: We enrolled a retrospective cohort with prospective follow-up. From August 2020 to May 2021, 130 patients completed the final follow-up. Among them, 61 patients received HUK combined with basic treatment, which we defined as the HUK group, and 69 patients received basic treatment, which we defined as the control group. We compared the changes of CysC, urea nitrogen and creatinine levels after one week of treatment between the two groups. Cognitive function was assessed by Montreal Cognitive Assessment (MoCA) at 3-month after AIS. RESULTS: No significant differences in demographic data and Laboratory tests between two groups before treatment. A total of 67 patients (51.5%) were diagnosed as PSCI at 3-month follow-up, among which, 25 patients were in the HUK group and 42 patients were in the control group. Compared with the control group (60.9%), the incidence of PSCI was significantly lower in the HUK group (41.0%). In addition, the serum CysC level after a week of treatment significantly decreased from baseline in HUK group (p = 0.037), in comparison, the serum CysC level in the control group was basically unchanged (p = 0.951). There was a significant negative correlation between MoCA score and the level of CysC after treatment (p = 0.003, r = -0.373). CONCLUSIONS: HUK can reduce the risk of PSCI at 3-month in AIS patients. The decrease of serum CysC level may be one of the mechanisms by which HUK reduces the incidence of PSCI.


Subject(s)
Cognitive Dysfunction , Ischemic Stroke , Kallikreins , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Humans , Incidence , Ischemic Stroke/complications , Kallikreins/therapeutic use , Prospective Studies , Retrospective Studies
4.
Cancer Biol Ther ; 23(1): 136-138, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35129066

ABSTRACT

The vast majority of the human proteome is yet to be functionally characterized thus hindering ongoing investigations on potential drug resistance mechanisms and advanced treatment options. Chemical proteomics is a powerful solution for enzyme profiling and the development of next generation cancer therapeutics previously deemed undruggable by small molecules. Within this field, activity-based protein profiling (ABPP) is a specialized technology capable of discriminating enzyme interactions that occur within complex, biological environments. In a recent publication by Lovell et al, the kallikrein-related peptidase (KLK) family of serine proteases that is highly implicated in the progression of prostate cancer (PCa) was subject to ABPP to elucidate enzymatic activities in the presence of enzalutamide. This is the first report of ABPP in PCa and of activity-based chemical probes selective for individual KLKs. Further, the study reveals androgen receptor-dependent activity among KLK proteins, particularly in mediating the invasion of the bone microenvironment.


Subject(s)
Kallikreins , Prostatic Neoplasms , Humans , Kallikreins/chemistry , Kallikreins/metabolism , Kallikreins/therapeutic use , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Tumor Microenvironment
5.
Drugs Today (Barc) ; 58(2): 59-67, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35188140

ABSTRACT

Orladeyo, a once-daily oral formulation of berotralstat (formerly BCX-7353), is a novel oral small-molecule drug developed by BioCryst Pharmaceuticals for the prevention of hereditary angioedema (HAE) attacks. It was first approved by the U.S. Food and Drug Administration (FDA) in 2020, and in 2021 also gained approval for marketing in Japan and the European Union. Preclinical and phase I studies showed promising efficacy and safety, and several multicenter international Angioedema Prophylaxis (APeX) phase II and III trials have since been initiated to further evaluate berotralstat. The ongoing phase III APeX-2 trial showed a 67% reduction in HAE attacks at the standard 150-mg dosing. Mild to moderate gastrointestinal side effects are most commonly seen and minimal serious adverse effects have been reported. Other first-line therapies for HAE prophylaxis rely on burdensome subcutaneous or intravenous routes. Thus far berotralstat has shown to be effective and well tolerated for HAE prophylaxis with the convenience of once-daily oral dosing.


Subject(s)
Angioedemas, Hereditary , Pharmaceutical Preparations , Angioedemas, Hereditary/drug therapy , Humans , Kallikreins/therapeutic use , Multicenter Studies as Topic , Pyrazoles/therapeutic use , United States
6.
J Clin Immunol ; 42(3): 597-605, 2022 04.
Article in English | MEDLINE | ID: mdl-35040012

ABSTRACT

Netherton syndrome (NS) is a rare, severe type of ichthyosis, often lethal in neonates, for which there is no therapy. Spink5-/- mice recapitulate major NS hallmarks and die homogeneously within 5 h from birth due to severe epidermal barrier defect leading to dehydration. Spink5-/-Klk5-/- mice survive neonatal lethality, indicating that KLK5 could be a drug target for NS. Nevertheless, after a week, these mice developed epidermal inflammation and signs of barrier defect leading to lethality. Here we tested whether anti-TNFα strategy in combination with anti-KLK5 could provide a long-term effective therapy for NS. Deletion of Tnfa in Spink5-/- suppressed the inflammatory phenotype but did not rescue neonatal lethality of Spink5-/- indicating that anti-TNFα therapy alone would not be sufficient to treat NS. Interestingly, in Spink5-/-Klk5-/-Tnfa-/- mice, NS features were rescued, and mice lived normally for 16-18 months. For the first time, evidence is provided that a combination of anti-TNFα and anti-KLK5 therapeutics represents an effective therapeutic strategy for NS. Notably, anti-TNFα factors are marketed and used widely, while LMW KLK5 inhibitors are being developed.


Subject(s)
Netherton Syndrome , Animals , Inflammation , Kallikreins/genetics , Kallikreins/therapeutic use , Mice , Netherton Syndrome/drug therapy , Netherton Syndrome/genetics , Phenotype , Protease Inhibitors/therapeutic use
7.
J. investig. allergol. clin. immunol ; 31(1): 1-16, 2021. tab, graf
Article in English | IBECS | ID: ibc-202251

ABSTRACT

Hereditary angioedema due to C1-esterase inhibitor deficiency (C1-INH-HAE) is a rare autosomal dominant disease.In the last decade, new drugs and new indications for old drugs have played a role in the management of C1-INH-HAE. This review examines current therapy for C1-INH-HAE and provides a brief summary of drugs that are under development. Increased knowledge of the pathophysiology of C1-INH-HAE has been crucial for advances in the field, with inhibition of the kallikrein-kinin system (plasma kallikrein, activated factor XII) as a key area in the discovery of new drugs, some of which are already marketed for treatment of C1-INH-HAE.Pharmacological treatment is based on 3 pillars: treatment of acute angioedema attacks (on-demand treatment), short-term (preprocedure) prophylaxis, and long-term prophylaxis.The 4 drugs that are currently available for the treatment of acute angioedema attacks (purified plasma-derived human C1 esterase inhibitor concentrate, icatibant acetate, ecallantide, recombinant human C1 esterase inhibitor) are all authorized for self-administration, except ecallantide.Purified plasma-derived human C1 esterase inhibitor concentrate is the treatment of choice for short-term prophylaxis.Tranexamic acid, danazol, intravenous and subcutaneous nanofiltered purified plasma-derived human C1 esterase inhibitor concentrate, and lanadelumab can be used for long-term prophylaxis.New drugs are being investigated, mainly as long-term prophylaxis, and are aimed at blocking the kallikrein-kinin system by means of antiprekallikrein, antikallikrein, and anti-activated FXII action


El angioedema hereditario por déficit del inhibidor de la C1 esterasa (AEH-C1-INH) es una enfermedad rara hereditaria autosómica dominante.En la última década nuevos fármacos y nuevas indicaciones de antiguos fármacos han llegado al área del AEH-C1-INH. En esta revisión se valora el conjunto de fármacos disponibles para el AEH-C1-INH, junto con los fármacos en desarrollo. Los avances en el conocimiento de la fisiopatología del AEH-C1-INH han sido fundamentales para este desarrollo, con la inhibición del sistema de calicreína-cininas (calicreína plasmática, factor XII activado) como un punto caliente para el descubrimiento de nuevos fármacos, algunos de los cuales ya han llegado al mercado del AEH-C1-INH.El tratamiento farmacológico se basa en tres pilares: tratamiento de los ataques agudos de angioedema (tratamiento a demanda), profilaxis a corto plazo o preprocedimiento, profilaxis a largo plazo.Hay actualmente 4 fármacos disponibles para el tratamiento de los ataques agudos de angioedema (concentrado plasmático purificado nanofiltrado del inhibidor de la C1 esterasa humana, acetato de icatibant, ecalantida, inhibidor recombinante de la C1 esterasa humana), todos ellos autorizados para autoadministración, excepto la ecalantida.El concentrado plasmático del inhibidor de la C1 esterasa humana es el tratamiento de elección como profilaxis a corto plazo.Como profilaxis a largo plazo se puede utilizar ácido tranexámico, danazol, concentrado plasmático del inhibidor de la C1 esterasa humano intravenoso y subcutáneo y lanadelumab.Se están investigando nuevos fármacos, fundamentalmente como profilaxis a largo plazo, dirigidos a bloquear el sistema calicreína cininas con acción anti precalicreína, anti calicreína y anti FXII activado


Subject(s)
Humans , Angioedema/classification , Hereditary Angioedema Types I and II/drug therapy , Kallikreins/therapeutic use , Angioedemas, Hereditary/drug therapy , Complement C1 Inhibitor Protein/therapeutic use , Bradykinin/therapeutic use , Angioedemas, Hereditary/physiopathology , Antibiotic Prophylaxis , Antibodies, Monoclonal/therapeutic use
8.
Acta Pharmacol Sin ; 41(12): 1597-1608, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32300244

ABSTRACT

Tissue kallikrein has protective function against various types of injury. In this study, we investigated whether exogenous pancreatic kininogenase (PK) conferred renoprotection in a rat model of unilateral ureteral obstruction (UUO) and H2O2-treated HK-2 cells in vitro. SD rats were subjected to UUO surgery, then PK (7.2 U/g per day, ip) was administered for 7 or 14 days. After the treatment, rats were euthanized; the obstructed kidneys were harvested for further examination. We found that PK administration significantly attenuated interstitial inflammation and fibrosis, and downregulated the expression of proinflammatory (MCP-1, TLR-2, and OPN) and profibrotic (TGF-ß1 and CTGF) cytokines in obstructed kidney. UUO-induced oxidative stress, closely associated with excessive apoptotic cell death and autophagy via PI3K/AKT/FoxO1a signaling, which were abolished by PK administration. We further showed that PK administration increased the expression of bradykinin receptors 1 and 2 (B1R and B2R) mRNA and the production of NO and cAMP in kidney tissues. Coadministration with either B1R antagonist (des-Arg9-[Leu8]-bradykinin) or B2R antagonist (icatibant) abrogated the renoprotective effects of PK, and reduced the levels of NO and cAMP in obstructed kidney. In H2O2-treated HK-2 cells, addition of PK (6 pg/mL) significantly decreased ROS production, regulated the expression of oxidant and antioxidant enzymes, suppressed the expression of TGF-ß1 and MCP-1, and inhibited cell apoptosis. Our data demonstrate that PK treatment protects against the progression of renal fibrosis in obstructed kidneys.


Subject(s)
Fibrosis/prevention & control , Kallikreins/therapeutic use , Kidney/metabolism , Pancreas/enzymology , Protective Agents/therapeutic use , Ureteral Obstruction/complications , Animals , Cell Death/drug effects , Cell Line , Fibrosis/etiology , Fibrosis/pathology , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Kallikrein-Kinin System/drug effects , Kidney/pathology , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ureteral Obstruction/pathology
9.
Neurol Res ; 42(4): 286-290, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32138624

ABSTRACT

Objectives: Intravenous thrombolysis and thrombectomy are recommended for patients whose stroke onsets are within first 6 h, and very few options are available for patients whose stroke onset is more than 6 h, which includes most ischemic stroke patients. Human urinary kallidinogenase (HUK) showed potential clinical benefits in acute ischemic stroke patients. This study aims to investigate the safety and clinical benefits of HUK in ischemic stroke patients.Patients and methods: Patients were recruited for a multicenter double-blind, placebo-controlled phase II b and phase III trial. Neurophysiological outcomes were assessed by the European Stroke Scale (ESS) and the functional outcomes were assessed by the activity of daily living scale (ADL). Safety was monitored by recording adverse events.Results: The improvements in ESS scores and ADL scores in the HUK group were significantly greater than that in patients receiving placebo. Furthermore, HUK treatment was also associated with a lower rate of disable, according to ADL. HUK-related adverse events occurred at a low rate, in 1.73% of HUK-treated patients.Conclusion: HUK is safe and provides potential clinical benefits as a treatment for acute ischemic stroke. Further large post-marketing observational studies are needed.


Subject(s)
Brain Ischemia/diagnosis , Brain Ischemia/drug therapy , Data Analysis , Ischemic Stroke/diagnosis , Ischemic Stroke/drug therapy , Kallikreins/therapeutic use , Adolescent , Adult , Aged , Coagulants/therapeutic use , Coagulants/urine , Double-Blind Method , Female , Humans , Kallikreins/urine , Male , Middle Aged , Prospective Studies , Treatment Outcome , Young Adult
10.
Brain Behav ; 10(1): e01461, 2020 01.
Article in English | MEDLINE | ID: mdl-31793238

ABSTRACT

INTRODUCTION: To evaluate effectiveness of human urinary kallindinogenase (HUK) in patients with acute ischemic stroke (AIS) according to Chinese ischemic stroke subclassification (CISS) and analyzed risk factors of clinical efficacy. METHODS: In this retrospective study, 134 patients received conventional therapy were enrolled to control group, and 132 patients received HUK treatment were enrolled to HUK group. National Institute of Health Stroke Scale (NIHSS) score was used to evaluate the clinical efficacy. Multivariate analysis of risk factors was performed by using logistic regression. RESULTS: After treatment, NIHSS score of HUK group was significant lower than that of control group (p = .009). Effectiveness rate was 71.2% in HUK group, and 53.7% in control group, respectively (p = .003). The NIHSS of patients with large artery atherosclerosis (LAA) subtype in HUK group was significantly lower than that in control group (p = .005). The absence of HUK (OR = 2.75), homocysteine (OR = 0.15), and CS subtype (OR = 0.18) were risk factors for HUK clinical efficacy. CONCLUSIONS: Human urinary kallindinogenase is an effective therapeutic approach for treatment of patients with AIS, especially in patients with LAA subtype. The absence of HUK, elevated homocysteine, and cardiogenic stroke subtype were risk factor for clinical efficacy of HUK.


Subject(s)
Ischemic Stroke/drug therapy , Kallikreins/therapeutic use , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome
11.
Eur Rev Med Pharmacol Sci ; 23(15): 6665-6671, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31378909

ABSTRACT

OBJECTIVE: To investigate the influences of urinary kallidinogenase on neuronal apoptosis in rats with cerebral infarction through the nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) oxidative stress pathway. MATERIALS AND METHODS: A total of 30 male rats were divided into group A (model control group), group B (rat model of cerebral infarction) and group C (rat model of cerebral infarction + medical treatment with urinary kallidinogenase). The percentage of cerebral infarct volume and the apoptosis of brain cells in the three groups of rats were detected via 2,3,5-Triphenyltetrazolium chloride (TTC) staining, the pathological morphology of brain tissues in the three groups of rats was observed via hematoxylin and eosin (HE) staining, and the protein levels of Nrf2 and superoxide dismutase 1 (SOD1) in the brain tissues in the three groups of rats were measured using the Western blotting assay. RESULTS: The degree of neurological deficit in group B was remarkably higher than that in group A (p<0.05), and it was markedly decreased in group C compared to that in group B, displaying statistically significant differences (p<0.05). Compared to that in group A, the cell apoptosis was significantly aggravated in group B, while a remarkably alleviated cell apoptosis was observed in group C compared to that of group B, and the differences were statistically significant (p<0.05). The cerebral infarct volume accounted for 34.87% of the whole brain volume in group B, and a mild cerebral infarction was detected in group C, with a percentage of cerebral infarct volume of 21.14%. Group B showed a more evident increase in the cerebral infarct volume than in group C (p<0.05). Compared to those of group A, pyknotic nuclei and neuron staining of brain tissue cells were evidently increased, and the neuronal cell injury was aggravated in group B. Moreover, prominently decreased pyknotic nuclei and neuron staining (p<0.05) as well as mild neuronal cell injury (p<0.05) were detected in group C compared to those in group B. The levels of Nrf2 and SOD1 protein in the brain tissues in group B were remarkably lower than those of group C (p<0.05). CONCLUSIONS: Urinary kallidinogenase can inhibit the neuronal apoptosis in rats and protect the rats from cerebral infarction, whose mechanism is associated with the activation of the Nrf2/ARE oxidative stress pathway.


Subject(s)
Apoptosis/drug effects , Brain Infarction/drug therapy , Brain/pathology , Kallikreins/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidant Response Elements/genetics , Apoptosis/genetics , Brain/cytology , Brain/drug effects , Brain Infarction/pathology , Disease Models, Animal , Humans , Injections, Intraperitoneal , Kallikreins/therapeutic use , Male , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Superoxide Dismutase-1/metabolism
12.
Diabetologia ; 62(6): 1074-1086, 2019 06.
Article in English | MEDLINE | ID: mdl-30838453

ABSTRACT

AIMS/HYPOTHESIS: Many studies have shown that tissue kallikrein has effects on diabetic vascular complications such as nephropathy, cardiomyopathy and neuropathy, but its effects on diabetic retinopathy are not fully understood. Here, we investigated the retinoprotective role of exogenous pancreatic kallikrein and studied potential mechanisms of action. METHODS: We used KK Cg-Ay/J (KKAy) mice (a mouse model of spontaneous type 2 diabetes) and mice with high-fat diet/streptozotocin (STZ)-induced type 2 diabetes as our models. After the onset of diabetes, both types of mice were injected intraperitoneally with either pancreatic kallikrein (KKAy + pancreatic kallikrein and STZ + pancreatic kallikrein groups) or saline (KKAy + saline and STZ + saline groups) for 12 weeks. C57BL/6J mice were used as non-diabetic controls for both models. We analysed pathological changes in the retina; evaluated the effects of pancreatic kallikrein on retinal oxidative stress, inflammation and apoptosis; and measured the levels of bradykinin and B1 and B2 receptors in both models. RESULTS: In both models, pancreatic kallikrein improved pathological structural features of the retina, increasing the thickness of retinal layers, and attenuated retinal acellular capillary formation and vascular leakage (p < 0.05). Furthermore, pancreatic kallikrein ameliorated retinal oxidative stress, inflammation and apoptosis in both models (p < 0.05). We also found that the levels of bradykinin and B1 and B2 receptors were increased after pancreatic kallikrein in both models (p < 0.05). CONCLUSIONS/INTERPRETATION: Pancreatic kallikrein can protect against diabetic retinopathy by activating B1 and B2 receptors and inhibiting oxidative stress, inflammation and apoptosis. Thus, pancreatic kallikrein may represent a new therapeutic agent for diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Retinopathy/drug therapy , Diet, High-Fat/adverse effects , Kallikreins/therapeutic use , Streptozocin/toxicity , Animals , Apoptosis/drug effects , Diabetes Mellitus, Type 2/chemically induced , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Retina/drug effects , Retina/metabolism
13.
Int Ophthalmol ; 39(6): 1307-1313, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29752593

ABSTRACT

PURPOSE: To evaluate the effectiveness of the combination of vitrectomy with kallidinogenase for diabetic macular edema (DME). METHODS: This study was designed as a prospective, randomized, multicenter study comparing 19 eyes of 19 patients who received 150 units of kallidinogenase administered a day for 52 weeks from the day after vitrectomy (study group) with 20 eyes of 20 patients who received no kallidinogenase (control group). The main outcome measurements included logMAR visual acuity and central foveal thickness (CFT) before surgery and at 3, 6, 9, and 12 months after vitrectomy. RESULTS: During follow-up, 11 patients dropped out (six in the study group and five in the control group), leaving 28 eyes in 28 patients for analysis (13 in the study group and 15 in the control group). Visual acuity improved significantly at 12 months in both groups compared with before surgery. The degree of improvement did not differ significantly between the groups. At 12 months, the mean CFT decreased significantly in both groups, with no significant difference in the rate of change between the two groups. In the study group, the visual acuity and CFT significantly improved from 3 to 12 months and from 6 to 12 months, whereas these parameters did not continue to improve in the control group after 6 months (for visual acuity) or 3 months (for CFT). CONCLUSION: After vitrectomy for DME, visual acuity and CFT improved significantly in both groups, but only patients treated with kallidinogenase continued to have significant improvement throughout the study period.


Subject(s)
Coagulants/therapeutic use , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/surgery , Kallikreins/therapeutic use , Macular Edema/drug therapy , Macular Edema/surgery , Vitrectomy/methods , Aged , Diabetic Retinopathy/physiopathology , Female , Fovea Centralis/pathology , Humans , Macular Edema/physiopathology , Male , Middle Aged , Prospective Studies , Tomography, Optical Coherence , Visual Acuity/physiology
14.
Urology ; 122: 89-96, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30171922

ABSTRACT

OBJECTIVES: To investigate the effect of kallikrein-related peptidase KLK1 on azoospermic mice induced by busulfan and mouse spermatogonial stem cell. METHODS: Mice were treated with a single intraperitoneal injection of busulfan, and 4 weeks later, they received a daily intraperitoneal injection of KLK1 at different doses for another 4 weeks. Eight weeks after the busulfan treatment, all mice were sacrificed and their testes were collected for histological evaluation, immunostaining and protein extraction. In vitro, immortalized mouse spermatogonial stem cells, namely C18-4 cells, were treated with KLK1 for proliferation assays. RESULTS: Histological evaluation of testes, epididymis and epididymal fluid showed that KLK1-treated mice had better spermatogenesis than the control group. Immunostaining showed that tissue samples from testes of KLK1-treated mice had more PLZF- and SCP3-positive cells per seminiferous tubule as well as more PNA-positive cells in the seminiferous tubules. Western blots revealed higher expression levels of PCNA in KLK1-treated mice than in control mice. C18-4 cells treated with KLK1 had a higher proliferation rate and higher expression levels of PCNA, Cyclin A and Cyclin E, and the level of phosphorylated ERK2 were increased after KLK1 treatment. CONCLUSION: Collectively, KLK1 can improve spermatogenesis in azoospermic mice, and KLK1 can promote the proliferation of mouse spermatogonial stem cells via activating ERK1/2 and cell cycle proteins Cyclin A and Cyclin E. This study could offer novel approach and provide new targets for the treatment of azoospermia.


Subject(s)
Azoospermia/drug therapy , Cell Proliferation/drug effects , Kallikreins/pharmacology , Regeneration/drug effects , Animals , Azoospermia/chemically induced , Busulfan/toxicity , Cell Line , Disease Models, Animal , Humans , Injections, Intraperitoneal , Kallikreins/therapeutic use , Male , Mice , Mice, Inbred ICR , Spermatogenesis/drug effects , Spermatogonia/drug effects , Spermatogonia/physiology , Stem Cells/drug effects , Stem Cells/physiology , Testis/cytology , Testis/drug effects , Testis/metabolism , Treatment Outcome
15.
Brain Behav ; 8(10): e01060, 2018 10.
Article in English | MEDLINE | ID: mdl-30160055

ABSTRACT

OBJECTIVE: To evaluate the clinical efficacy of Human Urinary Kallidinogenase (HUK) on the outcome of patients with ruptured intracranial aneurysm. METHODS: This was a prospective, open-label study. At the Department of Neurosurgery in our hospital, 127 patients were treated and operated due to ruptured intracranial aneurysm in the period 2015-2016. After surgery, all the patients received basic treatment and 70 patients received additional HUK treatment (HUK group) according to their willing. In detail, 0.15 PNA unit of HUK injection plus 100 ml saline in intravenous infusion was performed, with once a day for 14 consecutive days. The modified Rankin Scale (mRS) scores and favorable mRS rates (mRS 0-1) were analyzed 3-month after the treatment. RESULTS: No difference was shown in the basic characteristics between the two groups (p > 0.05). Favorable mRS rate in the HUK group (71.43%) was significantly higher than that in control group (50.88%, p < 0.05). In addition, 3-month death rate was significantly lower in the HUK group. Delayed ischemic stroke rate was similar between the two groups. CONCLUSION: HUK can reduce morbidity and mortality of patients with ruptured intracranial aneurysm after surgery.


Subject(s)
Aneurysm, Ruptured/drug therapy , Intracranial Aneurysm/drug therapy , Kallikreins/therapeutic use , Adult , Aged , Female , Humans , Male , Middle Aged , Prospective Studies , Treatment Outcome
16.
Asian J Androl ; 20(5): 448-453, 2018.
Article in English | MEDLINE | ID: mdl-29676291

ABSTRACT

Erectile dysfunction (ED) associated with type 2 diabetes is a severe problem that requires effective treatment. Pancreatic kininogenase (PK) has the potential to improve the erectile function of ED patients. This study aims to investigate the effect of PK on erectile function in streptozotocin-induced type 2 diabetic ED rats. To achieve this goal, we divided male Sprague-Dawley rats into five groups. One group was not treated, and the other four groups were treated with saline, sildenafil, PK or sildenafil, and PK, respectively, for 4 weeks after the induction of type 2 diabetic ED. Then, intracavernous pressure under cavernous nerve stimulation was measured, and penile tissue was collected for further study. Endothelial nitric oxide synthase levels, smooth muscle content, endothelium content, cyclic guanosine monophosphate (cGMP) levels in the corpus cavernosum, and neuronal nitric oxide synthase levels in the dorsal penile nerve were measured. Improved erectile function and endothelium and smooth muscle content in the corpus cavernosum were observed in diabetic ED rats. When treating diabetic ED rats with PK and sildenafil at the same time, a better therapeutic effect was achieved. These data demonstrate that intraperitoneal injection of PK can improve erectile function in a rat model of type 2 diabetic ED. With further research on specific mechanisms of erectile function improvement, PK may become a novel treatment for diabetic ED.


Subject(s)
Diabetes Mellitus, Experimental/complications , Erectile Dysfunction/drug therapy , Kallikreins/therapeutic use , Penile Erection/drug effects , Penis/drug effects , Urological Agents/therapeutic use , Animals , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Erectile Dysfunction/etiology , Erectile Dysfunction/metabolism , Erectile Dysfunction/physiopathology , Kallikreins/pharmacology , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Penile Erection/physiology , Penis/metabolism , Rats , Rats, Sprague-Dawley , Sildenafil Citrate/pharmacology , Sildenafil Citrate/therapeutic use , Treatment Outcome , Urological Agents/pharmacology
17.
Brain Res ; 1687: 129-136, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29510144

ABSTRACT

An urgent need exists to develop intra-arterial treatment for acute ischemic stroke in animal study. This study aimed to explore the beneficial effects of intra-arterial administration of human urinary kallidinogenase (HUK) on brain injury after permanent middle cerebral artery occlusion (pMCAO) in a rat model, and the potential underlying molecular mechanisms. Brain injury induced by pMCAO was evaluated through measuring neurological deficit scores, neuropathological changes, and inflammatory factors. Neurological deficits were observed 24 h after pMCAO and were alleviated by intra-arterial HUK treatment obviously. Inhibition of PI3K by LY294002 blocked the beneficial effect of HUK on neurological functions. In contrast to the pMCAO group, the intra-arterial HUK treatment group showed relatively more regularly arranged neurons and fewer pyknosis. Neurodegeneration, necrosis, infarct area and markers for brain injury were all ameliorated by intra-arterial HUK treatment. Moreover, a lower expression of inflammatory factors including interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α, and a higher expression of IL-10 were observed in the intra-arterial HUK treatment group than that in the pMCAO group. Additionally, when comparing with pMCAO group, a lower level of caspase-3, bax, and apoptotic rate, and a higher level of bcl-2, p-PI3K, p-AKT and p-FoxO1were observed in the pMCAO + HUK group. These results suggest that intra-arterial administration of HUK is a promising therapeutic strategy against pMCAO induced brain injury, and PI3K/AKT/FoxO1 signaling pathway may be involved in this process.


Subject(s)
Brain Injuries/drug therapy , Kallikreins/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Animals , Brain Injuries/etiology , Caspase 3/metabolism , Chromones/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/drug effects , Humans , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/complications , Injections, Intra-Arterial/methods , Male , Morpholines/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacokinetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
18.
Asian Journal of Andrology ; (6): 448-453, 2018.
Article in English | WPRIM (Western Pacific) | ID: wpr-1009605

ABSTRACT

Erectile dysfunction (ED) associated with type 2 diabetes is a severe problem that requires effective treatment. Pancreatic kininogenase (PK) has the potential to improve the erectile function of ED patients. This study aims to investigate the effect of PK on erectile function in streptozotocin-induced type 2 diabetic ED rats. To achieve this goal, we divided male Sprague-Dawley rats into five groups. One group was not treated, and the other four groups were treated with saline, sildenafil, PK or sildenafil, and PK, respectively, for 4 weeks after the induction of type 2 diabetic ED. Then, intracavernous pressure under cavernous nerve stimulation was measured, and penile tissue was collected for further study. Endothelial nitric oxide synthase levels, smooth muscle content, endothelium content, cyclic guanosine monophosphate (cGMP) levels in the corpus cavernosum, and neuronal nitric oxide synthase levels in the dorsal penile nerve were measured. Improved erectile function and endothelium and smooth muscle content in the corpus cavernosum were observed in diabetic ED rats. When treating diabetic ED rats with PK and sildenafil at the same time, a better therapeutic effect was achieved. These data demonstrate that intraperitoneal injection of PK can improve erectile function in a rat model of type 2 diabetic ED. With further research on specific mechanisms of erectile function improvement, PK may become a novel treatment for diabetic ED.


Subject(s)
Animals , Male , Rats , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental/physiopathology , Erectile Dysfunction/physiopathology , Kallikreins/therapeutic use , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Penile Erection/physiology , Penis/metabolism , Rats, Sprague-Dawley , Sildenafil Citrate/therapeutic use , Treatment Outcome , Urological Agents/therapeutic use
19.
Brain Behav ; 7(8): e00752, 2017 08.
Article in English | MEDLINE | ID: mdl-28828213

ABSTRACT

AIM: To evaluate the clinical efficacy of Human Urinary kallidinogenase (HUK) in the treatment of acute ischemic stroke (AIS) patients with level 3 hypertension. METHODS: In this retrospective study, from January 2015 to June 2016, 150 consecutive AIS patients were registered in our database. Among them, 47 with level 3 hypertension received either HUK treatment (HUK group, 22 cases) or basic treatment (control group, 25 cases). Basic treatment was administrated on all patients. 0.15 PNA unit of HUK injection plus 100 ml saline in intravenous infusion was performed in the HUK group, with once a day for 14 consecutive days. The modified Rankin Scale (mRS) scores in two groups were analyzed 3 months after the treatment. RESULTS: No difference was found in the NIHSS scores, age, gender, and comorbidities between two groups before treatment (p > .05). While after treatment, 3-month mRS score was significantly lower in the HUK group (2.1 ± 1.4 vs. 3.1 ± 1.3, p = .012) and good recovery rate (3-month mRS score ≤2) in the HUK group was significantly higher than that in the control group (p < .05). CONCLUSION: HUK is able to promote long-term recovery for AIS patients with level 3 hypertension remarkably.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/drug therapy , Hypertension/complications , Kallikreins/therapeutic use , Stroke/complications , Stroke/drug therapy , Aged , Coagulants/therapeutic use , Female , Humans , Male , Retrospective Studies , Sodium Chloride/therapeutic use , Treatment Outcome
20.
J Pharmacol Sci ; 134(2): 86-92, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28619445

ABSTRACT

Kallidinogenase has been used to treat retinal vein occlusion (RVO) in patients, although there are no evidences on the effects of kallidinogenase on the retinal edema and the non-perfused areas in eyes with a RVO. We have established a murine RVO model with retinal edema and non-perfused areas. The purpose of this study was to evaluate the effects of kallidinogenase on the retinal edema and size of the non-perfused areas in the mouse RVO model. We evaluated the thickness of the retinal layers and size of the non-perfused areas, and the blood flow by laser speckle flowgraphy in RVO model. The effects of an intravenous injection of kallidinogenase on the retinal edema and size of the non-perfused areas were determined. In addition, the expressions of phosphorylated protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) were measured by Western blotting. Our results showed that kallidinogenase reduced the degree of retinal edema and size of the non-perfused areas by an increase in the blood flow in RVO model. Kallidinogenase also increased the levels of phosphorylated Akt and eNOS. These findings indicate that kallidinogenase acted through Akt/eNOS-dependent phosphorylation. Thus, kallidinogenase should be considered as a possible therapeutic agent for RVO patients.


Subject(s)
Kallikreins/therapeutic use , Papilledema/drug therapy , Retinal Vein Occlusion/drug therapy , Animals , Humans , Male , Mice , Nitric Oxide Synthase Type III/metabolism , Papilledema/metabolism , Papilledema/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Regional Blood Flow , Retinal Vein Occlusion/metabolism , Retinal Vein Occlusion/pathology
SELECTION OF CITATIONS
SEARCH DETAIL