Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557489

RESUMO

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Assuntos
Canais de Cálcio , Cálcio , Camundongos , Animais , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Pâncreas/metabolismo , Exocitose/fisiologia , Vesículas Secretórias/genética
2.
Int J Mol Sci ; 24(17)2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37686032

RESUMO

Chronic kidney disease (CKD) progression is associated with persisting oxidative stress, which impairs the NO-sGC-cGMP signaling cascade through the formation of oxidized and heme-free apo-sGC that cannot be activated by NO. Runcaciguat (BAY 1101042) is a novel, potent, and selective sGC activator that binds and activates oxidized and heme-free sGC and thereby restores NO-sGC-cGMP signaling under oxidative stress. Therefore, runcaciguat might represent a very effective treatment option for CKD/DKD. The potential kidney-protective effects of runcaciguat were investigated in ZSF1 rats as a model of CKD/DKD, characterized by hypertension, hyperglycemia, obesity, and insulin resistance. ZSF1 rats were treated daily orally for up to 12 weeks with runcaciguat (1, 3, 10 mg/kg/bid) or placebo. The study endpoints were proteinuria, kidney histopathology, plasma, urinary biomarkers of kidney damage, and gene expression profiling to gain information about relevant pathways affected by runcaciguat. Furthermore, oxidative stress was compared in the ZSF1 rat kidney with kidney samples from DKD patients. Within the duration of the 12-week treatment study, kidney function was significantly decreased in obese ZSF1 rats, indicated by a 20-fold increase in proteinuria, compared to lean ZSF1 rats. Runcaciguat dose-dependently and significantly attenuated the development of proteinuria in ZSF1 rats with reduced uPCR at the end of the study by -19%, -54%, and -70% at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo treatment. Additionally, average blood glucose levels measured as HbA1C, triglycerides, and cholesterol were increased by five times, twenty times, and four times, respectively, in obese ZSF1 compared to lean rats. In obese ZSF1 rats, runcaciguat reduced HbA1c levels by -8%, -34%, and -76%, triglycerides by -42%, -55%, and -71%, and cholesterol by -16%, -17%, and -34%, at 1, 3, and 10 mg/kg/bid, respectively, compared to placebo. Concomitantly, runcaciguat also reduced kidney weights, morphological kidney damage, and urinary and plasma biomarkers of kidney damage. Beneficial effects were accompanied by changes in gene expression that indicate reduced fibrosis and inflammation and suggest improved endothelial stabilization. In summary, the sGC activator runcaciguat significantly prevented a decline in kidney function in a DKD rat model that mimics common comorbidities and conditions of oxidative stress of CKD patients. Thus, runcaciguat represents a promising treatment option for CKD patients, which is in line with recent phase 2 clinical study data, where runcaciguat showed promising efficacy in CKD patients (NCT04507061).


Assuntos
Rim , Insuficiência Renal Crônica , Animais , Ratos , GMP Cíclico , Hemoglobinas Glicadas , Heme , Obesidade , Proteinúria , Insuficiência Renal Crônica/tratamento farmacológico , Ensaios Clínicos Fase II como Assunto
3.
Cell Rep Med ; 4(4): 100992, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37023747

RESUMO

Diabetic kidney disease (DKD) is the most common cause of renal failure. Therapeutics development is hampered by our incomplete understanding of animal models on a cellular level. We show that ZSF1 rats recapitulate human DKD on a phenotypic and transcriptomic level. Tensor decomposition prioritizes proximal tubule (PT) and stroma as phenotype-relevant cell types exhibiting a continuous lineage relationship. As DKD features endothelial dysfunction, oxidative stress, and nitric oxide depletion, soluble guanylate cyclase (sGC) is a promising DKD drug target. sGC expression is specifically enriched in PT and stroma. In ZSF1 rats, pharmacological sGC activation confers considerable benefits over stimulation and is mechanistically related to improved oxidative stress regulation, resulting in enhanced downstream cGMP effects. Finally, we define sGC gene co-expression modules, which allow stratification of human kidney samples by DKD prevalence and disease-relevant measures such as kidney function, proteinuria, and fibrosis, underscoring the relevance of the sGC pathway to patients.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Humanos , Ratos , Animais , Guanilil Ciclase Solúvel/metabolismo , Guanilil Ciclase Solúvel/farmacologia , Guanilil Ciclase Solúvel/uso terapêutico , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/patologia , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Guanilato Ciclase/farmacologia , Rim/metabolismo , Fibrose
4.
Toxicol Sci ; 187(1): 35-50, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35244176

RESUMO

Micro-RNAs (miRNAs) are regulators of gene expression and play an important role in physiological homeostasis and disease. In biofluids, miRNAs can be found in protein complexes or in extracellular vesicles (EVs). Altered urinary miRNAs are reported as potential biomarkers for chronic kidney disease (CKD). In this context, we compared established urinary protein biomarkers for kidney injury with urinary miRNA profiles in obese ZSF1 and hypertensive renin transgenic rats. Additionally, the benefit of urinary EV enrichment was investigated in vivo and the potential association of urinary miRNAs with renal fibrosis in vitro. Kidney damage in both rat models was confirmed by histopathology, proteinuria, and increased levels of urinary protein biomarkers. In total, 290 miRNAs were elevated in obese ZSF1 rats compared with lean controls, whereas 38 miRNAs were altered in obese ZSF1 rats during 14-26 weeks of age. These 38 miRNAs correlated better with disease progression than established urinary protein biomarkers. MiRNAs increased in obese ZSF1 rats were associated with renal inflammation, fibrosis, and glomerular injury. Eight miRNAs were also changed in urinary EVs of renin transgenic rats, including one which might play a role in endothelial dysfunction. EV enrichment increased the number and detection level of several miRNAs implicated in renal fibrosis in vitro and in vivo. Our results show the benefit of EV enrichment for miRNA detection and the potential of total urine and urinary EV-associated miRNAs as biomarkers of altered kidney physiology, renal fibrosis and glomerular injury, and disease progression in hypertension and obesity-induced CKD.


Assuntos
Vesículas Extracelulares , Hipertensão , MicroRNAs , Insuficiência Renal Crônica , Animais , Biomarcadores/metabolismo , Progressão da Doença , Vesículas Extracelulares/metabolismo , Feminino , Fibrose , Humanos , Hipertensão/metabolismo , Rim/metabolismo , Masculino , MicroRNAs/genética , Obesidade/metabolismo , Ratos , Renina/metabolismo
5.
Naunyn Schmiedebergs Arch Pharmacol ; 394(12): 2363-2379, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34550407

RESUMO

Chronic kidney diseaQueryse (CKD) is associated with oxidative stress which can interrupt the nitric oxide (NO)/soluble guanylyl cyclase (sGC) signaling and decrease cyclic guanosine monophosphate (cGMP) production. Low cGMP concentrations can cause kidney damage and progression of CKD. The novel sGC activator runcaciguat targets the oxidized and heme-free form of sGC, restoring cGMP production under oxidative stress. The purpose of this study is to investigate if runcaciguat could provide an effective treatment for CKD. Runcaciguat was used for the treatment not only in rat CKD models with different etiologies and comorbidities, namely of hypertensive rats, the renin transgenic (RenTG) rat, and angiotensin-supplemented (ANG-SD) rat, but also in rats with diabetic and metabolic CKD, the Zucker diabetic fatty (ZDF) rat. The treatment duration was 2 to 42 weeks and runcaciguat was applied orally in doses from 1 to 10 mg/kg/bid. In these different rat CKD models, runcaciguat significantly reduced proteinuria (urinary protein to creatinine ratio; uPCR). These effects were also significant at doses which did not or only moderately decrease systemic blood pressure. Moreover, runcaciguat significantly decreased kidney injury biomarkers and attenuated morphological kidney damages. In RenTG rats, runcaciguat improved survival rates and markers of heart injury. These data demonstrate that the sGC activator runcaciguat exhibits cardio-renal protection at doses which did not reduce blood pressure and was effective in hypertensive as well as diabetic and metabolic CKD models. These data, therefore, suggest that runcaciguat, with its specific mode of action, represents an efficient treatment approach for CKD and associated CV diseases.


Assuntos
Ciclopropanos , Diabetes Mellitus Experimental , Hipertensão , Insuficiência Renal Crônica , Animais , Masculino , Ratos , Pressão Sanguínea/efeitos dos fármacos , GMP Cíclico/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativadores de Enzimas/administração & dosagem , Ativadores de Enzimas/farmacologia , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Ratos Sprague-Dawley , Ratos Transgênicos , Ratos Zucker , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/prevenção & controle , Guanilil Ciclase Solúvel/efeitos dos fármacos , Guanilil Ciclase Solúvel/metabolismo , Fatores de Tempo , Ciclopropanos/farmacologia , Ciclopropanos/uso terapêutico
6.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34360780

RESUMO

Duchenne muscular dystrophy (DMD) is a severe and progressive muscle wasting disorder, affecting one in 3500 to 5000 boys worldwide. The NO-sGC-cGMP pathway plays an important role in skeletal muscle function, primarily by improving blood flow and oxygen supply to the muscles during exercise. In fact, PDE5 inhibitors have previously been investigated as a potential therapy for DMD, however, a large-scale Phase III clinical trial did not meet its primary endpoint. Since the efficacy of PDE5i is dependent on sufficient endogenous NO production, which might be impaired in DMD, we investigated if NO-independent sGC stimulators, could have therapeutic benefits in a mouse model of DMD. Male mdx/mTRG2 mice aged six weeks were given food supplemented with the sGC stimulator, BAY-747 (150 mg/kg of food) or food alone (untreated) ad libitum for 16 weeks. Untreated C57BL6/J mice were used as wild type (WT) controls. Assessments of the four-limb hang, grip strength, running wheel and serum creatine kinase (CK) levels showed that mdx/mTRG2 mice had significantly reduced skeletal muscle function and severe muscle damage compared to WT mice. Treatment with BAY-747 improved grip strength and running speed, and these mice also had reduced CK levels compared to untreated mdx/mTRG2 mice. We also observed increased inflammation and fibrosis in the skeletal muscle of mdx/mTRG2 mice compared to WT. While gene expression of pro-inflammatory cytokines and some pro-fibrotic markers in the skeletal muscle was reduced following BAY-747 treatment, there was no reduction in infiltration of myeloid immune cells nor collagen deposition. In conclusion, treatment with BAY-747 significantly improves several functional and pathological parameters of the skeletal muscle in mdx/mTRG2 mice. However, the effect size was moderate and therefore, more studies are needed to fully understand the potential treatment benefit of sGC stimulators in DMD.


Assuntos
Ativadores de Enzimas/farmacologia , Músculo Esquelético/enzimologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Guanilil Ciclase Solúvel/metabolismo , Animais , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia
7.
Am J Nephrol ; 52(8): 642-652, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34111864

RESUMO

INTRODUCTION: The nonsteroidal mineralocorticoid receptor (MR) antagonist finerenone and sodium-glucose cotransporter-2 (SGLT2) inhibitors have demonstrated clinical benefits in CKD patients with type 2 diabetes. Clinical data analyzing the potential value of a combination therapy are currently limited. We therefore investigated cardiorenal protection of respective mono- and combination therapy in a preclinical model of hypertension-induced end-organ damage. METHODS: Cardiovascular (CV) morbidity and mortality were studied in hypertensive, N(ω)-nitro-L-arginine methyl ester-treated, renin-transgenic (mRen2)27 rats. Rats (10- to 11-week-old females, n = 13-17/group) were treated once daily orally for up to 7 weeks with placebo, finerenone (1 and 3 mg/kg), empagliflozin (3 and 10 mg/kg), or a combination of the respective low doses. Key outcome parameters included mortality, proteinuria, plasma creatinine and uric acid, blood pressure, and cardiac and renal histology. RESULTS: Placebo-treated rats demonstrated a 50% survival rate over the course of 7 weeks. Drug treatment resulted in variable degrees of survival benefit, most prominently in the low-dose combination group with a survival benefit of 93%. Monotherapies of finerenone or empagliflozin dose-dependently reduced proteinuria, while low-dose combination revealed an early, sustained, and over-additive reduction in proteinuria. Empagliflozin induced a strong and dose-dependent increase in urinary glucose excretion which was not influenced by finerenone coadministration in the combination arm. Low-dose combination but not respective low-dose monotherapies significantly reduced plasma creatinine and plasma uric acid after 6 weeks. Treatment with finerenone and the low-dose combination significantly decreased systolic blood pressure after 5 weeks. There was a dose-dependent protection from cardiac and kidney fibrosis and vasculopathy with both agents, while low-dose combination therapy was more efficient than the respective monotherapy dosages on most cardiorenal histology parameters. DISCUSSION/CONCLUSIONS: Nonsteroidal MR antagonism by finerenone and SGLT2 inhibition by empagliflozin confer CV protection in preclinical hypertension-induced cardiorenal disease. Combination of these 2 independent modes of action at low dosages revealed efficacious reduction in important functional parameters such as proteinuria and blood pressure, plasma markers including creatinine and uric acid, cardiac and renal lesions as determined by histopathology, and mortality indicating a strong potential for combined clinical use in cardiorenal patient populations.


Assuntos
Compostos Benzidrílicos/administração & dosagem , Glucosídeos/administração & dosagem , Cardiopatias/prevenção & controle , Nefropatias/prevenção & controle , Naftiridinas/administração & dosagem , Inibidores do Transportador 2 de Sódio-Glicose/administração & dosagem , Animais , Modelos Animais de Doenças , Combinação de Medicamentos , Feminino , Cardiopatias/etiologia , Hipertensão/complicações , Nefropatias/etiologia , Ratos
8.
Cell Physiol Biochem ; 54(6): 1115-1131, 2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33166100

RESUMO

BACKGROUND/AIMS: The release of insulin in response to increased levels of glucose in the blood strongly depends on Ca2+ influx into pancreatic beta cells by the opening of voltage-gated Ca2+ channels. Transient Receptor Potential Melastatin 3 proteins build Ca2+ permeable, non-selective cation channels serving as pain sensors of noxious heat in the peripheral nervous system. TRPM3 channels are also strongly expressed in pancreatic beta cells that respond to the TRPM3 agonist pregnenolone sulfate with Ca2+ influx and increased insulin release. Therefore, we hypothesized that in beta cells TRPM3 channels may contribute to pregnenolone sulfate- as well as to glucose-induced insulin release. METHODS: We used INS-1 cells as a beta cell model in which we analysed the occurrence of TRPM3 isoformes by immunoprecipitation and western blotting and by cloning of RT-PCR amplified cDNA fragments. We applied pharmacological as well as CRISPR/Cas9-based strategies to analyse the interplay of TRPM3 and voltage-gated Ca2+ channels in imaging experiments (FMP, Fura-2) and electrophysiological recordings. In immunoassays, we examined the contribution of TRPM3 channels to pregnenolone sulfate- and glucose-induced insulin release. To confirm our findings, we generated beta cell-specific Trpm3-deficient mice and compared their glucose clearance with the wild type in glucose tolerance tests. RESULTS: TRPM3 channels triggered the activity of voltage-gated Ca2+ channels and both channels together contributed to insulin release after TRPM3 activation. Trpm3-deficient INS-1 cells lacked pregnenolone sulfate-induced Ca2+ signals just like the pregnenolone sulfate-induced insulin release. Both, glucose-induced Ca2+ signals and the glucose-induced insulin release were strongly reduced. Accordingly, Trpm3-deficient mice displayed an impaired decrease of the blood sugar concentration after intraperitoneal or oral administration of glucose. CONCLUSION: The present study suggests an important role for TRPM3 channels in the control of glucose-dependent insulin release.


Assuntos
Sinalização do Cálcio , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Linhagem Celular , Camundongos , Camundongos Mutantes , Ratos , Canais de Cátion TRPM/genética
9.
JCI Insight ; 4(19)2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31487266

RESUMO

Nitric oxide regulates BP by binding the reduced heme iron (Fe2+) in soluble guanylyl cyclase (sGC) and relaxing vascular smooth muscle cells (SMCs). We previously showed that sGC heme iron reduction (Fe3+ → Fe2+) is modulated by cytochrome b5 reductase 3 (CYB5R3). However, the in vivo role of SMC CYB5R3 in BP regulation remains elusive. Here, we generated conditional smooth muscle cell-specific Cyb5r3 KO mice (SMC CYB5R3-KO) to test if SMC CYB5R3 loss affects systemic BP in normotension and hypertension via regulation of the sGC redox state. SMC CYB5R3-KO mice exhibited a 5.84-mmHg increase in BP and impaired acetylcholine-induced vasodilation in mesenteric arteries compared with controls. To drive sGC oxidation and elevate BP, we infused mice with angiotensin II. We found that SMC CYB5R3-KO mice exhibited a 14.75-mmHg BP increase, and mesenteric arteries had diminished nitric oxide-dependent vasodilation but increased responsiveness to sGC heme-independent activator BAY 58-2667 over controls. Furthermore, acute injection of BAY 58-2667 in angiotensin II-treated SMC CYB5R3-KO mice showed greater BP reduction compared with controls. Together, these data provide the first in vivo evidence to our knowledge that SMC CYB5R3 is an sGC heme reductase in resistance arteries and provides resilience against systemic hypertension development.


Assuntos
Angiotensina II/metabolismo , Citocromo-B(5) Redutase/genética , Citocromo-B(5) Redutase/metabolismo , Heme/metabolismo , Hipertensão/metabolismo , Músculo Liso Vascular/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Animais , Benzoatos , Pressão Sanguínea , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Oxirredução , Transcriptoma , Vasodilatação
10.
Cell Calcium ; 78: 66-75, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30658323

RESUMO

Methylglyoxal (MG) is a by-product of glucose metabolism and its accumulation has been linked to the development of diabetic complications such as retinopathy and nephropathy by affecting multiple signalling pathways. However, its influence on the intracellular Ca2+ homeostasis and particularly Ca2+ entry, which has been reported to be mediated via TRPA1 channels in DRG neurons, has not been studied in much detail in other cell types. In this study, we report the consequences of acute and long-term MG application on intracellular Ca2+ levels in endothelial cells. We showed that acute MG application doesn't evoke any instantaneous changes in the intracellular Ca2+ concentration in immortalized mouse cardiac endothelial cells (MCECs) and murine microvascular endothelial cells (muMECs). In contrast, an MG-induced rise in intracellular Ca2+ level was observed in primary mouse mesangial cells within 30 s, indicating that the modulation of Ca2+ homeostasis by MG is strictly cell type specific. The formation of the MG-derived advanced glycation end product (AGE) MG-H1 was found to be time and concentration-dependent in MCECs. Likewise, MG pre-incubation for 6 h increased the angiotensin II-evoked Ca2+ entry in MCECs and muMECs which was abrogated by inhibition of Calcium release activated calcium (CRAC) channels with GSK-7975A, but unaffected by an inhibitor specific to TRPA1 channels. Quantitative PCR analysis revealed that MG pre-treatment did not affect expression of the genes encoding the angiotensin receptors AT1R (Agtr 1a & Agtr 1b), Trpa1 nor Orai1, Orai2, Orai3, Stim1, Stim2 and Saraf which operate as constituents or regulators of CRAC channels and store-operated Ca2+ entry (SOCE) in other cell types. Together, our results show that long-term MG stimulation leads to the formation of glycation end products, which facilitates the agonist-evoked Ca2+ entry in endothelial cells, and this could be a new pathway that might lead to MG-evoked vasoregression observed in diabetic vasculopathies.


Assuntos
Canais de Cálcio Ativados pela Liberação de Cálcio/metabolismo , Cálcio/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Aldeído Pirúvico/farmacologia , Animais , Células Cultivadas , Camundongos
11.
Oncotarget ; 9(51): 29634-29643, 2018 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-30038709

RESUMO

(-)-Englerin A (EA) is a natural product which has potent cytotoxic effects on renal cell carcinoma cells and other types of cancer cell but not non-cancer cells. Although selectively cytotoxic to cancer cells, adverse reaction in mice and rats has been suggested. EA is a remarkably potent activator of ion channels formed by Transient Receptor Potential Canonical 4 and 5 proteins (TRPC4 and TRPC5) and TRPC4 is essential for EA-mediated cancer cell cytotoxicity. Here we specifically investigated the relevance of TRPC4 and TRPC5 to the adverse reaction. Injection of EA (2 mg.kg-1 i.p.) adversely affected mice for about 1 hour, manifesting as a marked reduction in locomotor activity, after which they fully recovered. TRPC4 and TRPC5 single knockout mice were partially protected and double knockout mice fully protected. TRPC4/TRPC5 double knockout mice were also protected against intravenous injection of EA. Importance of TRPC4/TRPC5 channels was further suggested by pre-administration of Compound 31 (Pico145), a potent and selective small-molecule inhibitor of TRPC4/TRPC5 channels which did not cause adverse reaction itself but prevented adverse reaction to EA. EA was detected in the plasma but not the brain and so peripheral mechanisms were implicated but not identified. The data confirm the existence of adverse reaction to EA in mice and suggest that it depends on a combination of TRPC4 and TRPC5 which therefore overlaps partially with TRPC4-dependent cancer cell cytotoxicity. The underlying nature of the observed adverse reaction to EA, as a consequence of TRPC4/TRPC5 channel activation, remains unclear and warrants further investigation.

12.
Mol Metab ; 9: 156-167, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29373286

RESUMO

OBJECTIVE: Diabetic retinopathy (DR) is induced by an accumulation of reactive metabolites such as ROS, RNS, and RCS species, which were reported to modulate the activity of cation channels of the TRPC family. In this study, we use Trpc1/4/5/6-/- compound knockout mice to analyze the contribution of these TRPC proteins to diabetic retinopathy. METHODS: We used Nanostring- and qPCR-based analysis to determine mRNA levels of TRPC channels in control and diabetic retinae and retinal cell types. Chronic hyperglycemia was induced by Streptozotocin (STZ) treatment. To assess the development of diabetic retinopathy, vasoregression, pericyte loss, and thickness of individual retinal layers were analyzed. Plasma and cellular methylglyoxal (MG) levels, as well as Glyoxalase 1 (GLO1) enzyme activity and protein expression, were measured in WT and Trpc1/4/5/6-/- cells or tissues. MG-evoked toxicity in cells of both genotypes was compared by MTT assay. RESULTS: We find that Trpc1/4/5/6-/- mice are protected from hyperglycemia-evoked vasoregression determined by the formation of acellular capillaries and pericyte drop-out. In addition, Trpc1/4/5/6-/- mice are resistant to the STZ-induced reduction in retinal layer thickness. The RCS metabolite methylglyoxal, which represents a key mediator for the development of diabetic retinopathy, was significantly reduced in plasma and red blood cells (RBCs) of STZ-treated Trpc1/4/5/6-/- mice compared to controls. GLO1 is the major MG detoxifying enzyme, and its activity and protein expression were significantly elevated in Trpc1/4/5/6-deficient cells, which led to significantly increased resistance to MG toxicity. GLO1 activity was also increased in retinal extracts from Trpc1/4/5/6-/- mice. The TRPCs investigated here are expressed at different levels in endothelial and glial cells of the retina. CONCLUSION: The protective phenotype in diabetic retinopathy observed in Trpc1/4/5/6-/- mice is suggestive of a predominant action of TRPCs in Müller cells and microglia because of their central position in the retention of a proper homoeostasis of the neurovascular unit.


Assuntos
Retinopatia Diabética/metabolismo , Lactoilglutationa Liase/metabolismo , Aldeído Pirúvico/metabolismo , Canais de Cátion TRPC/genética , Animais , Células Cultivadas , Retinopatia Diabética/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Aldeído Pirúvico/sangue , Retina/metabolismo , Canais de Cátion TRPC/metabolismo
13.
Am J Respir Cell Mol Biol ; 58(5): 636-647, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29268036

RESUMO

Sickle cell disease (SCD) is associated with intravascular hemolysis and oxidative inhibition of nitric oxide (NO) signaling. BAY 54-6544 is a small-molecule activator of oxidized soluble guanylate cyclase (sGC), which, unlike endogenous NO and the sGC stimulator, BAY 41-8543, preferentially binds and activates heme-free, NO-insensitive sGC to restore enzymatic cGMP production. We tested orally delivered sGC activator, BAY 54-6544 (17 mg/kg/d), sGC stimulator, BAY 41-8543, sildenafil, and placebo for 4-12 weeks in the Berkeley transgenic mouse model of SCD (BERK-SCD) and their hemizygous (Hemi) littermate controls (BERK-Hemi). Right ventricular (RV) maximum systolic pressure (RVmaxSP) was measured using micro right-heart catheterization. RV hypertrophy (RVH) was determined using Fulton's index and RV corrected weight (ratio of RV to tibia). Pulmonary artery vasoreactivity was tested for endothelium-dependent and -independent vessel relaxation. Right-heart catheterization revealed higher RVmaxSP and RVH in BERK-SCD versus BERK-Hemi, which worsened with age. Treatment with the sGC activator more effectively lowered RVmaxSP and RVH, with 90-day treatment delivering superior results, when compared with other treatments and placebo groups. In myography experiments, acetylcholine-induced (endothelium-dependent) and sodium-nitroprusside-induced (endothelium-independent NO donor) relaxation of the pulmonary artery harvested from placebo-treated BERK-SCD was impaired relative to BERK-Hemi but improved after therapy with sGC activator. By contrast, no significant effect for sGC stimulator or sildenafil was observed in BERK-SCD. These findings suggest that sGC is oxidized in the pulmonary arteries of transgenic SCD mice, leading to blunted responses to NO, and that the sGC activator, BAY 54-6544, may represent a novel therapy for SCD-associated pulmonary arterial hypertension and cardiac remodeling.


Assuntos
Anemia Falciforme/complicações , Ativadores de Enzimas/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Esquerda/prevenção & controle , Artéria Pulmonar/efeitos dos fármacos , Guanilil Ciclase Solúvel/metabolismo , Disfunção Ventricular Direita/tratamento farmacológico , Função Ventricular Direita/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Anemia Falciforme/genética , Animais , Pressão Arterial/efeitos dos fármacos , Modelos Animais de Doenças , Ativação Enzimática , Ativadores de Enzimas/farmacocinética , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/fisiopatologia , Camundongos Transgênicos , Morfolinas/farmacologia , Óxido Nítrico/metabolismo , Artéria Pulmonar/enzimologia , Artéria Pulmonar/fisiopatologia , Pirimidinas/farmacologia , Citrato de Sildenafila/farmacologia , Vasodilatação/efeitos dos fármacos , Disfunção Ventricular Direita/enzimologia , Disfunção Ventricular Direita/genética , Disfunção Ventricular Direita/fisiopatologia , Pressão Ventricular/efeitos dos fármacos
14.
Eur J Appl Physiol ; 118(1): 195-203, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29159668

RESUMO

PURPOSE: Testing of investigational drugs in animal models is a critical step in drug development. Current models of pulmonary hypertension (PH) have limitations. The most relevant outcome parameters such as pulmonary artery pressure (PAP) are measured invasively which requires anesthesia of the animal. We developed a new canine PH model in which pulmonary vasodilators can be characterized in conscious dogs and lung selectivity can be assessed non-invasively. METHODS: Telemetry devices were implanted to measure relevant hemodynamic parameters in conscious dogs. A hypoxic chamber was constructed in which the animals were placed in a conscious state. By reducing the inspired oxygen fraction (FiO2) to 10%, a hypoxic pulmonary vasoconstriction was induced leading to PH. The PDE-5 inhibitor sildenafil, the current standard of care was compared to atrial natriuretic peptide (ANP). RESULTS: The new hypoxic chamber provided a stable hypoxic atmosphere during all experiments. The mean PAP under normoxic conditions was 15.8 ± 1.8 mmHg. Hypoxia caused a reliable increase in mean PAP (+ 12.2 ± 3.2 mmHg, p < 0.0001). Both, sildenafil (- 6.8 ± 4.4 mmHg) and ANP (- 6.4 ± 3.8 mmHg) significantly (p < 0.05) decreased PAP. Furthermore sildenafil and ANP showed similar effects on systemic hemodynamics. In subsequent studies, the in vitro effects and gene expression pattern of the two pathways were exemplified. CONCLUSIONS: By combining the hypoxic environment with the telemetric approach, we could successfully establish a new acute PH model. Sildenafil and ANP demonstrated equal effects regarding pulmonary selectivity. This non-invasive model could help to rapidly screen pulmonary vasodilators with decreased animal burden.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Hipertensão Pulmonar/tratamento farmacológico , Artéria Pulmonar/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Fator Natriurético Atrial/farmacologia , Fator Natriurético Atrial/uso terapêutico , Modelos Animais de Doenças , Cães , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , Artéria Pulmonar/fisiopatologia , Citrato de Sildenafila/farmacologia , Citrato de Sildenafila/uso terapêutico , Telemetria/métodos , Vasodilatadores/uso terapêutico , Vigília
15.
EMBO J ; 36(18): 2770-2789, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28790178

RESUMO

Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.


Assuntos
Hipocampo/fisiologia , Memória de Curto Prazo , Multimerização Proteica , Transmissão Sináptica , Canais de Cátion TRPC/metabolismo , Animais , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Knockout , Canais de Cátion TRPC/genética
16.
Sci Rep ; 6: 32981, 2016 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-27624684

RESUMO

TRPM4 proteins form Ca(2+)-activated non selective cation (CAN) channels that affect transmembrane Ca(2+)-influx by determining the membrane potential. Tight control of the intracellular Ca(2+) concentration is essential for mast cell responses. In this study, we analyzed the expression of TRPM4 in peritoneal mast cells (PCMC) as a model for connective tissue type mast cells with respect to FcεRI-evoked calcium changes and the subcellular localization of fluorescently labeled TRPM4 using two viral transduction systems before and following antigen stimulation. Our results show that TRPM4 is expressed in PCMCs, is an essential constituent of the endogenous CAN channels in PCMCs and regulates antigen-evoked increases in intracellular calcium that are significantly enhanced in TRPM4-deficient PCMCs. Compared to PCMCs analyzed before antigen stimulation, the cells depict a substantially increased localization of TRPM4 proteins towards the plasma membrane after FcεRI stimulation. Thus, TRPM4 functions as a limiting factor for antigen evoked calcium rise in connective tissue type mast cells and concurrent translocation of TRPM4 into the plasma membrane is part of this mechanism.


Assuntos
Cálcio/metabolismo , Células do Tecido Conjuntivo/metabolismo , Mastócitos/metabolismo , Receptores de IgE/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Potenciais da Membrana , Camundongos , Transporte Proteico
17.
J Vis Exp ; (103)2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26436250

RESUMO

Papillary muscle isolated from adult mouse hearts can be used to study cardiac contractility during different physiological/pathological conditions. The contractile characteristics can be evaluated independently of external influences such as vascular tonus or neurohumoral status. It depicts a scientific approach between single cell measurements with isolated cardiac myocytes and in vivo studies like echocardiography. Thus, papillary muscle preparations serve as an excellent model to study cardiac physiology/pathophysiology and can be used for investigations like the modulation by pharmacological agents or the exploration of transgenic animal models. Here, we describe a method of isolating the murine left anterior papillary muscle to investigate cardiac contractility in an organ bath setup. In contrast to a muscle strip preparation isolated from the ventricular wall, the papillary muscle can be prepared in toto without damaging the muscle tissue severely. The organ bath setup consists of several temperature-controlled, gassed and electrode-equipped organ bath chambers. The isolated papillary muscle is fixed in the organ bath chamber and electrically stimulated. The evoked twitch force is recorded using a pressure transducer and parameters such as twitch force amplitude and twitch kinetics are analyzed. Different experimental protocols can be performed to investigate the calcium- and frequency-dependent contractility as well as dose-response curves of contractile agents such as catecholamines or other pharmaceuticals. Additionally, pathologic conditions like acute ischemia can be simulated.


Assuntos
Preparação de Coração Isolado/métodos , Contração Miocárdica/fisiologia , Músculos Papilares/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Cálcio/metabolismo , Estimulação Elétrica , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/metabolismo , Músculos Papilares/cirurgia , Transdução de Sinais
18.
Eur Heart J ; 36(33): 2257-66, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26069213

RESUMO

AIMS: Pathological cardiac hypertrophy is a major predictor for the development of cardiac diseases. It is associated with chronic neurohumoral stimulation and with altered cardiac Ca(2+) signalling in cardiomyocytes. TRPC proteins form agonist-induced cation channels, but their functional role for Ca(2+) homeostasis in cardiomyocytes during fast cytosolic Ca(2+) cycling and neurohumoral stimulation leading to hypertrophy is unknown. METHODS AND RESULTS: In a systematic analysis of multiple knockout mice using fluorescence imaging of electrically paced adult ventricular cardiomyocytes and Mn(2+)-quench microfluorimetry, we identified a background Ca(2+) entry (BGCE) pathway that critically depends on TRPC1/C4 proteins but not others such as TRPC3/C6. Reduction of BGCE in TRPC1/C4-deficient cardiomyocytes lowers diastolic and systolic Ca(2+) concentrations both, under basal conditions and under neurohumoral stimulation without affecting cardiac contractility measured in isolated hearts and in vivo. Neurohumoral-induced cardiac hypertrophy as well as the expression of foetal genes (ANP, BNP) and genes regulated by Ca(2+)-dependent signalling (RCAN1-4, myomaxin) was reduced in TRPC1/C4 knockout (DKO), but not in TRPC1- or TRPC4-single knockout mice. Pressure overload-induced hypertrophy and interstitial fibrosis were both ameliorated in TRPC1/C4-DKO mice, whereas they did not show alterations in other cardiovascular parameters contributing to systemic neurohumoral-induced hypertrophy such as renin secretion and blood pressure. CONCLUSIONS: The constitutively active TRPC1/C4-dependent BGCE fine-tunes Ca(2+) cycling in beating adult cardiomyocytes. TRPC1/C4-gene inactivation protects against development of maladaptive cardiac remodelling without altering cardiac or extracardiac functions contributing to this pathogenesis.


Assuntos
Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Cardiomegalia/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Cátion TRPC/fisiologia , Angiotensina II/metabolismo , Angiotensinogênio/metabolismo , Animais , Cálcio/metabolismo , Cardiomegalia/fisiopatologia , Hemodinâmica/fisiologia , Homeostase/fisiologia , Camundongos Knockout , Remodelação Ventricular
19.
J Mol Cell Cardiol ; 74: 307-17, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24972051

RESUMO

TRPM4 and TRPM5 proteins belong to the Transient Receptor Potential (TRP) ion channel family and form Ca(2+)-activated nonselective cation channels. Recently we showed a significant increase of Isoprenaline-induced inotropy in TRPM4-deficient (Trpm4(-/-)) mice. This is caused by increased Ca(2+) entry via L-type calcium channels due to faster action potential repolarization in Trpm4(-/-) ventricular myocytes [Mathar et al., 2013]. Here, we investigated the contribution of various steps of the ß-adrenergic signalling cascade to the augmented positive inotropic response in the absence of TRPM4, and whether the closely related TRPM5 additively contributes to this process using TRPM4/TRPM5-double deficient (Trpm4/Trpm5((-/-)2)) mice. We performed contractility measurements on isolated papillary muscles from wild type, Trpm4(-/-) and Trpm4/Trpm5((-/-)2) mice. As shown in Trpm4(-/-) mice, Isoprenaline-induced inotropy in Trpm4/Trpm5((-/-)2) papillary muscles was significantly increased compared to wild type, whereas basal, frequency- and Ca(2+)-dependent contractility was unaltered. Equivalent to Isoprenaline, activation of adenylyl cyclase using Forskolin led to a significantly increased twitch force in Trpm4(-/-) heart preparations whereas the Isoprenaline-mediated increase in cAMP level was comparable to wild type mice. Notably, the positive inotropic response evoked by phosphodiesterase inhibition with 3-isobutyl-1-methylxanthine (IBMX) was unchanged between both genotypes. Furthermore, experiments performed with increasing concentrations of IBMX after prestimulation with Forskolin and vice versa did not provide evidence that the increased ß-adrenergic positive inotropic response in TRPM4-deficient papillary muscles is due to differences in accumulation of cAMP. Compared to inhibition of phosphodiesterase, the rise of intracellular cAMP by activating adenylyl cyclase is accompanied by ATP breakdown. To test the relevance of TRPM4 during forced ATP consumption we measured contractility under ischemic conditions. Here, Trpm4(-/-) papillary muscles showed improved contractile function in comparison to wild type. Our results are consistent with the hypothesis that TRPM4 has a limiting effect on cardiac contractility specifically in ATP depleting conditions. The increased positive inotropic response in Trpm4(-/-) papillary muscles evoked by stimulation of adenylyl cyclase activity is not observed without active enhancement of ATP hydrolysis. Furthermore, the contractility of Trpm4(-/-) papillary muscles was also increased during ischemic simulation. These data underscore the potential of TRPM4 inactivation as an approach to increase inotropy in specific conditions associated with increased catecholamine levels, such as heart failure and ischemia.


Assuntos
Adenilil Ciclases/genética , Cardiotônicos/farmacologia , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/genética , Canais de Cátion TRPM/genética , Trifosfato de Adenosina/metabolismo , Adenilil Ciclases/metabolismo , Animais , Cálcio/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Estimulação Elétrica , Regulação da Expressão Gênica , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Masculino , Camundongos , Camundongos Knockout , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/metabolismo , Músculos Papilares/patologia , Transdução de Sinais , Canais de Cátion TRPM/deficiência , Técnicas de Cultura de Tecidos
20.
Handb Exp Pharmacol ; 222: 461-87, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24756717

RESUMO

TRPM4 is a Ca(2+)-activated nonselective cation channel. The channel is activated by an increase of intracellular Ca(2+) and is regulated by several factors including temperature and Pi(4,5)P2. TRPM4 allows Na(+) entry into the cell upon activation, but is completely impermeable to Ca(2+). Unlike TRPM5, its closest relative in the transient receptor potential family, TRPM4 proteins are widely expressed in the body. Currents with properties that are reminiscent of TRPM4 have been described in a variety of tissues since the advent of the patch clamp technology, but their physiological role is only beginning to be clarified with the increasing characterization of knockout mouse models for TRPM4. Furthermore, mutations in the TRPM4 gene have been associated with cardiac conduction disorders in human patients. This review aims to overview the currently available data on the functional properties of TRPM4 and the current understanding of its physiological role in healthy and diseased tissue.


Assuntos
Canais de Cátion TRPM/metabolismo , Animais , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Potenciais da Membrana , Moduladores de Transporte de Membrana/farmacologia , Camundongos , Camundongos Knockout , Fenótipo , Conformação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Canais de Cátion TRPM/química , Canais de Cátion TRPM/deficiência , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...