Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Histochem Cell Biol ; 156(6): 539-553, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34545457

RESUMO

The olfactory receptor Olfr78 (prostate-specific G protein-coupled receptor PSGR) is a member of the G protein-coupled receptor family mediating olfactory chemosensation, but it is additionally expressed in other tissues. Olfr78 expressed in kidney participates in blood pressure regulation, and in prostate it plays a role in the development of cancer. We here screened many organs/tissues of transgenic mice co-expressing ß-galactosidase with Olfr78. X-gal-positive cells were detectable in smooth muscle cells of numerous arterioles of striated muscles (heart ventricles and skeletal muscles of various embryological origin). In addition, in most organs where we found expression of Olfr78 mRNA, X-gal staining was restricted to smooth muscle cells of small blood vessels. The dominant expression of Olfr78 in arteriolar smooth muscle cells supports the concept of an important role in blood pressure regulation and suggests a participation in the fine tuning of blood supply especially of striated muscles. This should be considered when targeting Olfr78 in other contexts such as prostate cancer.


Assuntos
Arteríolas/metabolismo , Músculo Esquelético/metabolismo , Músculo Liso/metabolismo , Miocárdio/metabolismo , Receptores Odorantes/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Odorantes/metabolismo
2.
Pulm Circ ; 8(4): 2045894018808240, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30284509

RESUMO

We show that genetic deficiency of the reactive oxygen species generating enzyme NADPH oxidase 4 (NOX4) impairs hypoxic pulmonary vasoconstriction in small (25-40 µm) intra-acinar, but not pre-acinar, arteries in murine precision cut lung slices. These data suggest an involvement of NOX4 in ventilation-perfusion matching at the acinar level.

3.
Front Physiol ; 9: 918, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30057556

RESUMO

Substance P (SP) is a neuropeptide engaged in the signal transmission of neural C fibers afferents in the myocardium. The actions of SP in the heart are extensive and they are mediated by the neurokinin 1 receptor (NK1R), a member of the tachykinin subfamily of G-protein coupled receptors. The receptors have been found in the heart, but to our knowledge, their exact localization in the heart has not been described yet. Here, we investigated the presence of NK1R protein in separate rat heart compartments by means of western blot and its tissue distribution by means of immunofluorescence. Specificity of NK1R immunolabeling was controlled by preabsorption of the antiserum with its corresponding peptide. Additionally, we investigated abundance of gene for NK1R in separated heart chambers by means of quantitative real-time PCR (RT-PCR). Relative abundance of NK1R mRNA was expressed as a ratio of target gene Cq value to Cq value of control gene - beta-actin. Finally, we studied abundance of NK1R mRNA in different cell types of heart isolated by laser capture microdissection. Immunofluorescence showed NK1R immunoreactivity on the surface of some intracardiac neurons and smooth muscle cells of coronary vessels. The results of quantitative RT-PCR indicate abundance of mRNA for NK1R in all heart chambers with highest level in the left atrium. The presence of NK1R mRNA was detected in some samples of dissected intracardiac neurons, but not in cardiomyocytes or smooth muscle cells of coronary vessels. In the course of long-term diabetes, a significant downregulation of the NK1R mRNA was seen in the right atrium and upregulation in the right ventricle 53 weeks after the induction of diabetes. Our results indicate localization of NK1R in some intracardiac neurons and smooth muscle cells. Impaired transcription of the NK1R gene in the diabetic heart may be induced by unidentified genes or factors involved in the development of diabetic cardiomyopathy.

4.
PLoS One ; 12(3): e0174071, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28301582

RESUMO

The two-pore domain potassium channel KCNK3 (TASK-1) is expressed in rat and human pulmonary artery smooth muscle cells. There, it is associated with hypoxia-induced signalling, and its dysfunction is linked to pathogenesis of human pulmonary hypertension. We here aimed to determine its role in hypoxic pulmonary vasoconstriction (HPV) in the mouse, and hence the suitability of this model for further mechanistic investigations, using appropriate inhibitors and TASK-1 knockout (KO) mice. RT-PCR revealed expression of TASK-1 mRNA in murine lungs and pre-acinar pulmonary arteries. Protein localization by immunohistochemistry and western blot was unreliable since all antibodies produced labelling also in TASK-1 KO organs/tissues. HPV was investigated by videomorphometric analysis of intra- (inner diameter: 25-40 µm) and pre-acinar pulmonary arteries (inner diameter: 41-60 µm). HPV persisted in TASK-1 KO intra-acinar arteries. Pre-acinar arteries developed initial HPV, but the response faded earlier (after 30 min) in KO vessels. This HPV pattern was grossly mimicked by the TASK-1 inhibitor anandamide in wild-type vessels. Hypoxia-provoked rise in pulmonary arterial pressure (PAP) in isolated ventilated lungs was affected neither by TASK-1 gene deficiency nor by the TASK-1 inhibitor A293. TASK-1 is dispensable for initiating HPV of murine intra-pulmonary arteries, but participates in sustained HPV specifically in pre-acinar arteries. This does not translate into abnormal rise in PAP. While there is compelling evidence that TASK-1 is involved in the pathogenesis of pulmonary arterial hypertension in humans, the mouse does not appear to serve as a suitable model to study the underlying molecular mechanisms.


Assuntos
Hipóxia/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição/fisiologia , Animais , Ácidos Araquidônicos/farmacologia , Endocanabinoides/farmacologia , Feminino , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Alcamidas Poli-Insaturadas/farmacologia , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/efeitos dos fármacos , RNA Mensageiro/genética
5.
Cell Tissue Res ; 364(2): 245-62, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26590824

RESUMO

Energy substrates and metabolic intermediates are proven ligands of a growing number of G-protein coupled receptors. In 2004, GPR91 and GPR99 were identified as receptors for the citric acid cycle intermediates, succinate and α-ketoglutarate, respectively. GPR91 seems to act as a first responder to local stress and GPR99 participates in the regulation of the acid-base balance through an intrarenal paracrine mechanism. However, a systematic analysis of the distribution of both receptors in mouse organs is still missing. The aim of this study was to examine the expression of GPR91 and GPR99 in a large number of different murine organs both at mRNA and protein level. Whereas GPR91 mRNA was detectable in almost all organs, GPR99 mRNA was mainly expressed in neuronal tissues. Widespread expression of GPR91 was also detected at the protein level by western blotting and immunohistochemistry. In addition to neuronal cells, GPR99 protein was found in renal intercalated cells and epididymal narrow cells. Double-labeling immunohistochemistry demonstrated the colocalization of GPR99 with the B1 subunit isoform of vacuolar H(+)-ATPases which is expressed only by a very limited number of cell types. In summary, our detailed expression analysis of GPR91 and GPR99 in murine tissues will allow a more directed search for additional functions of both receptors.


Assuntos
Glândulas Suprarrenais/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Rim/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos P2/metabolismo , Glândula Submandibular/metabolismo , Animais , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/genética , Receptores Purinérgicos P2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico/fisiologia
6.
J Histochem Cytochem ; 63(5): 329-39, 2015 05.
Artigo em Inglês | MEDLINE | ID: mdl-25673288

RESUMO

Recent evidence reveals a crucial role for acetylcholine and its receptors in the regulation of inflammation, particularly of nicotinic acetylcholine receptor α7 (Chrna7) and muscarinic acetylcholine receptor 3 (Chrm3). Immunohistochemistry is a key tool for their cellular localization in functional tissues. We evaluated nine different commercially available antibodies on back skin tissue from wild-type (Wt) and gene-deficient (KO) mice. In the immunohistochemical analysis, we focused on key AChR-ligand sensitive skin cells (mast cells, nerve fibers and keratinocytes). All five antibodies tested for Chrm3 and the first three Chrna7 antibodies stained positive in both Wt and respective KO skin. With the 4th antibody (ab23832) nerve fibers were unlabeled in the KO mice. By western blot analysis, this antibody detected bands in both Wt and Chrna7 KO skin and brain. qRT-PCR revealed mRNA amplification with a primer set for the undeleted region in both Wt and KO mice, but none with a primer set for the deleted region in KO mice. By 2D electrophoresis, we found ß-actin and ß-enolase cross reactivity, which was confirmed by double immunolabeling. In view of the present results, the tested antibodies are not suitable for immunolocalization in skin and suggest thorough control of antibody specificity is required if histomorphometry is intended.


Assuntos
Anticorpos/imunologia , Receptor Muscarínico M3/metabolismo , Pele/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Actinas/metabolismo , Animais , Especificidade de Anticorpos , Immunoblotting , Imuno-Histoquímica , Mastócitos/metabolismo , Camundongos Knockout , Fosfopiruvato Hidratase/metabolismo , RNA Mensageiro/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/imunologia , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/imunologia
7.
Brain Struct Funct ; 220(4): 1885-99, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24706047

RESUMO

In recent pain studies on animal models, α7 nicotinic acetylcholine receptor (nAChR) agonists demonstrated analgesic, anti-hyperalgesic and anti-inflammatory effects, apparently acting through some peripheral receptors. Assuming possible involvement of α7 nAChRs on nociceptive sensory neurons, we investigated the morphological and neurochemical features of the α7 nAChR-expressing subpopulation of dorsal root ganglion (DRG) neurons and their ability to transport α7 nAChR axonally. In addition, α7 receptor activity and its putative role in pain signal neurotransmitter release were studied. Medium-sized α7 nAChR-expressing neurons prevailed, although the range covered all cell sizes. These cells accounted for one-fifth of total medium and large DRG neurons and <5% of small ones. 83.2% of α7 nAChR-expressing DRG neurons were peptidergic nociceptors (CGRP-immunopositive), one half of which had non-myelinated C-fibers and the other half had myelinated Aδ- and likely Aα/ß-fibers, whereas 15.2% were non-peptidergic C-fiber nociceptors binding isolectin B4. All non-peptidergic and a third of peptidergic α7 nAChR-bearing nociceptors expressed TRPV1, a capsaicin-sensitive noxious stimulus transducer. Nerve crush experiments demonstrated that CGRPergic DRG nociceptors axonally transported α7 nAChRs both to the spinal cord and periphery. α7 nAChRs in DRG neurons were functional as their specific agonist PNU282987 evoked calcium rise enhanced by α7-selective positive allosteric modulator PNU120596. However, α7 nAChRs do not modulate neurotransmitter CGRP and glutamate release from DRG neurons since nicotinic ligands affected neither their basal nor provoked levels, showing the necessity of further studies to elucidate the true role of α7 nAChRs in those neurons.


Assuntos
Transporte Axonal/fisiologia , Gânglios Espinais/patologia , Nociceptores/metabolismo , Neuropatia Ciática/patologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Transporte Axonal/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Colinérgicos/farmacologia , Modelos Animais de Doenças , Feminino , Lectinas/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo , Agonistas Nicotínicos/farmacologia , Nociceptores/classificação , Nociceptores/efeitos dos fármacos , Nociceptores/patologia , Ratos , Ratos Wistar , Canais de Cátion TRPV/metabolismo
8.
Crit Care ; 18(2): R73, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24731244

RESUMO

INTRODUCTION: Ventilator-induced lung injury (VILI) contributes to morbidity and mortality in acute respiratory distress syndrome (ARDS). Particularly pre-injured lungs are susceptible to VILI despite protective ventilation. In a previous study, the endogenous peptide adrenomedullin (AM) protected murine lungs from VILI. We hypothesized that mechanical ventilation (MV) contributes to lung injury and sepsis in pneumonia, and that AM may reduce lung injury and multiple organ failure in ventilated mice with pneumococcal pneumonia. METHODS: We analyzed in mice the impact of MV in established pneumonia on lung injury, inflammation, bacterial burden, hemodynamics and extrapulmonary organ injury, and assessed the therapeutic potential of AM by starting treatment at intubation. RESULTS: In pneumococcal pneumonia, MV increased lung permeability, and worsened lung mechanics and oxygenation failure. MV dramatically increased lung and blood cytokines but not lung leukocyte counts in pneumonia. MV induced systemic leukocytopenia and liver, gut and kidney injury in mice with pneumonia. Lung and blood bacterial burden was not affected by MV pneumonia and MV increased lung AM expression, whereas receptor activity modifying protein (RAMP) 1-3 expression was increased in pneumonia and reduced by MV. Infusion of AM protected against MV-induced lung injury (66% reduction of pulmonary permeability p < 0.01; prevention of pulmonary restriction) and against VILI-induced liver and gut injury in pneumonia (91% reduction of AST levels p < 0.05, 96% reduction of alanine aminotransaminase (ALT) levels p < 0.05, abrogation of histopathological changes and parenchymal apoptosis in liver and gut). CONCLUSIONS: MV paved the way for the progression of pneumonia towards ARDS and sepsis by aggravating lung injury and systemic hyperinflammation leading to liver, kidney and gut injury. AM may be a promising therapeutic option to protect against development of lung injury, sepsis and extrapulmonary organ injury in mechanically ventilated individuals with severe pneumonia.


Assuntos
Adrenomedulina/uso terapêutico , Broncodilatadores/uso terapêutico , Pneumonia Pneumocócica/prevenção & controle , Respiração Artificial/efeitos adversos , Sepse/prevenção & controle , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/patologia , Sepse/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
9.
J Physiol ; 592(8): 1745-56, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24535440

RESUMO

Dopamine not only is a precursor of the catecholamines noradrenaline and adrenaline but also serves as an independent neurotransmitter and paracrine hormone. It plays an important role in the pathogenesis of hypertension and is a potent vasodilator in many mammalian systemic arteries, strongly suggesting an endogenous source of dopamine in the vascular wall. Here we demonstrated dopamine, noradrenaline and adrenaline in rat aorta and superior mesenteric arteries (SMA) by radioimmunoassay. Chemical sympathectomy with 6-hydroxydopamine showed a significant reduction of noradrenaline and adrenaline, while dopamine levels remained unaffected. Isolated endothelial cells were able to synthesize and release dopamine upon cAMP stimulation. Consistent with these data, mRNAs coding for catecholamine synthesizing enzymes, i.e. tyrosine hydroxylase (TH), aromatic l-amino acid decarboxylase, and dopamine-ß-hydroxylase were detected by RT-PCR in cultured endothelial cells from SMA. TH protein was detected by immunohistochemisty and Western blot. Exposure of endothelial cells to hypoxia (1% O2) increased TH mRNA. Vascular smooth muscle cells partially expressed catecholaminergic traits. A physiological role of endogenous vascular dopamine was shown in SMA, where D1 dopamine receptor blockade abrogated hypoxic vasodilatation. Experiments on SMA with endothelial denudation revealed a significant contribution of the endothelium, although subendothelial dopamine release dominated. From these results we conclude that endothelial cells and cells of the underlying vascular wall synthesize and release dopamine in an oxygen-regulated manner. In the splanchnic vasculature, this intrinsic non-neuronal dopamine is the dominating vasodilator released upon lowering of oxygen tension.


Assuntos
Aorta/fisiologia , Hipóxia Celular , Dopamina/metabolismo , Artérias Mesentéricas/fisiologia , Vasodilatação , Animais , Aorta/citologia , Aorta/metabolismo , Descarboxilases de Aminoácido-L-Aromático/genética , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Células Cultivadas , AMP Cíclico/farmacologia , Antagonistas de Dopamina/farmacologia , Dopamina beta-Hidroxilase/genética , Dopamina beta-Hidroxilase/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Artérias Mesentéricas/citologia , Artérias Mesentéricas/metabolismo , Norepinefrina/metabolismo , Ratos , Ratos Wistar , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
10.
J Vis Exp ; (83): e50970, 2014 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-24458260

RESUMO

Acute alveolar hypoxia causes pulmonary vasoconstriction (HPV) - also known as von Euler-Liljestrand mechanism - which serves to match lung perfusion to ventilation. Up to now, the underlying mechanisms are not fully understood. The major vascular segment contributing to HPV is the intra-acinar artery. This vessel section is responsible for the blood supply of an individual acinus, which is defined as the portion of lung distal to a terminal bronchiole. Intra-acinar arteries are mostly located in that part of the lung that cannot be selectively reached by a number of commonly used techniques such as measurement of the pulmonary artery pressure in isolated perfused lungs or force recordings from dissected proximal pulmonary artery segments(1,2). The analysis of subpleural vessels by real-time confocal laser scanning luminescence microscopy is limited to vessels with up to 50 µm in diameter(3). We provide a technique to study HPV of murine intra-pulmonary arteries in the range of 20-100 µm inner diameters. It is based on the videomorphometric analysis of cross-sectioned arteries in precision cut lung slices (PCLS). This method allows the quantitative measurement of vasoreactivity of small intra-acinar arteries with inner diameter between 20-40 µm which are located at gussets of alveolar septa next to alveolar ducts and of larger pre-acinar arteries with inner diameters between 40-100 µm which run adjacent to bronchi and bronchioles. In contrast to real-time imaging of subpleural vessels in anesthetized and ventilated mice, videomorphometric analysis of PCLS occurs under conditions free of shear stress. In our experimental model both arterial segments exhibit a monophasic HPV when exposed to medium gassed with 1% O2 and the response fades after 30-40 min at hypoxia.


Assuntos
Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Microscopia Confocal/métodos , Gravação em Vídeo/métodos , Animais , Pulmão/anatomia & histologia , Camundongos , Alvéolos Pulmonares/fisiopatologia , Artéria Pulmonar/anatomia & histologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição/fisiologia
11.
PLoS One ; 7(5): e35832, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563471

RESUMO

BACKGROUND: Even protective ventilation may aggravate or induce lung failure, particularly in preinjured lungs. Thus, new adjuvant pharmacologic strategies are needed to minimize ventilator-induced lung injury (VILI). Intermedin/Adrenomedullin-2 (IMD) stabilized pulmonary endothelial barrier function in vitro. We hypothesized that IMD may attenuate VILI-associated lung permeability in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Human pulmonary microvascular endothelial cell (HPMVEC) monolayers were incubated with IMD, and transcellular electrical resistance was measured to quantify endothelial barrier function. Expression and localization of endogenous pulmonary IMD, and its receptor complexes composed of calcitonin receptor-like receptor (CRLR) and receptor activity-modifying proteins (RAMPs) 1-3 were analyzed by qRT-PCR and immunofluorescence in non ventilated mouse lungs and in lungs ventilated for 6 h. In untreated and IMD treated mice, lung permeability, pulmonary leukocyte recruitment and cytokine levels were assessed after mechanical ventilation. Further, the impact of IMD on pulmonary vasoconstriction was investigated in precision cut lung slices (PCLS) and in isolated perfused and ventilated mouse lungs. IMD stabilized endothelial barrier function in HPMVECs. Mechanical ventilation reduced the expression of RAMP3, but not of IMD, CRLR, and RAMP1 and 2. Mechanical ventilation induced lung hyperpermeability, which was ameliorated by IMD treatment. Oxygenation was not improved by IMD, which may be attributed to impaired hypoxic vasoconstriction due to IMD treatment. IMD had minor impact on pulmonary leukocyte recruitment and did not reduce cytokine levels in VILI. CONCLUSIONS/SIGNIFICANCE: IMD may possibly provide a new approach to attenuate VILI.


Assuntos
Células Endoteliais/efeitos dos fármacos , Lesão Pulmonar/prevenção & controle , Pulmão/efeitos dos fármacos , Hormônios Peptídicos/farmacologia , Animais , Proteína Semelhante a Receptor de Calcitonina/genética , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Imunofluorescência , Expressão Gênica/efeitos dos fármacos , Humanos , Hipóxia , Técnicas In Vitro , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Lesão Pulmonar/etiologia , Lesão Pulmonar/genética , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Microvasos/efeitos dos fármacos , Microvasos/fisiopatologia , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Proteína 1 Modificadora da Atividade de Receptores/genética , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Proteína 2 Modificadora da Atividade de Receptores/genética , Proteína 2 Modificadora da Atividade de Receptores/metabolismo , Proteína 3 Modificadora da Atividade de Receptores/genética , Proteína 3 Modificadora da Atividade de Receptores/metabolismo , Respiração Artificial/efeitos adversos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vasoconstrição/efeitos dos fármacos
12.
Cardiovasc Res ; 93(4): 702-10, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22215723

RESUMO

AIMS: Alveolar hypoxia acutely elicits contraction of pulmonary arteries, leading to a rise in pulmonary arterial pressure (PAP) and shifting blood to better ventilated areas of the lung. The molecular mechanisms underlying this hypoxic pulmonary vasoconstriction (HPV) are still incompletely understood. Here, we investigated the role of succinate dehydrogenase (SDH; synonymous to mitochondrial complex II) in HPV, with particular emphasis on regional differences along the vascular bed and consequences for PAP and perfusion-to-ventilation matching, using mutant mice heterozygous for the SDHD subunit of complex II (SDHD(+/-)). METHODS AND RESULTS: Western blots revealed reduced protein content of complex II subunits SDHA, SDHB, and SDHC in lungs of SDHD(+/-) mice, despite unaffected mRNA content as determined by real-time PCR. Hypoxic pulmonary vasoconstriction of small (20-50 µm) intra-acinar and larger (51-100 µm) pre-acinar arteries was evaluated by videomorphometric analysis of precision-cut lung slices. The hypoxic response was detectable in pre-acinar arteries but absent from intra-acinar arteries of SDHD(+/-) mice. In isolated perfused lungs, basal PAP and its hypoxia-induced increase were indistinguishable between both mouse strains. Arterial oxygenation was measured after provocation of regional ventilatory failure by tracheal fluid instillation in anaesthetized mice, and it declined more in SDHD(+/-) than in wild-type mice. CONCLUSION: SDHD is required for the formation of a stable mitochondrial complex II and it is selectively important for HPV of intra-acinar vessels. This specialized vascular segment participates in perfusion-to-ventilation matching but does not significantly contribute to the acute hypoxic rise in PAP that results from more proximal vasoconstriction.


Assuntos
Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Artéria Pulmonar/fisiopatologia , Succinato Desidrogenase/fisiologia , Vasoconstrição/fisiologia , Animais , Pressão Sanguínea/fisiologia , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/fisiologia , Heterozigoto , Pulmão/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Mutantes , Modelos Animais , RNA Mensageiro/metabolismo , Succinato Desidrogenase/genética
13.
J Surg Res ; 166(2): 306-13, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19766239

RESUMO

Data on a protective role of fumarate in acute ischemia of the rat heart led to the obvious hypothesis that addition of fumarate to the preservation solution for kidney transplantation may have beneficial value. This study was designed to test this hypothesis. Kidneys of Lewis or Fischer 344 rats were flushed with University of Wisconsin (UW) solution or UW solution containing 5 mM fumarate. Grafts were immediately transplanted to Lewis recipients or stored at 4 °C for 5 h before transplantation. Renal function was assessed on d 10 and monthly for 6 mo. One group of isografts was removed on d 10 post-transplantation, the other groups of isografts and allografts after 6 mo. We detected a modest protective effect regarding proteinuria 10 d after isogeneic transplantation, and exclude the possibility that fumarate exerts acute nephrotoxicity. Surprisingly, fumarate strongly promoted intimal hyperplasia of allograft arteries, thickening of the arterial media of isografts and allografts, tubulo-interstitial allograft damage, and allograft infiltration by macrophages on the long run. To date, we do not know the mechanism resulting in fumarate-induced chronic graft damage. We suggest, however, that addition of fumarate to the conservation fluid does not improve graft outcome.


Assuntos
Fumaratos/toxicidade , Transplante de Rim , Soluções para Preservação de Órgãos/toxicidade , Disfunção Primária do Enxerto/induzido quimicamente , Disfunção Primária do Enxerto/patologia , Doença Aguda , Animais , Doença Crônica , Hiperplasia , Rim/metabolismo , Rim/patologia , Lactatos/metabolismo , Leucócitos/patologia , Masculino , Disfunção Primária do Enxerto/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Artéria Renal/metabolismo , Artéria Renal/patologia , Transplante Homólogo
14.
J Mol Cell Cardiol ; 49(6): 950-61, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20920510

RESUMO

α-Keto acids (α-KAs) are not just metabolic intermediates but are also powerful modulators of different cellular pathways. Here, we tested the hypothesis that α-KA concentrations are regulated by complex II (succinate dehydrogenase=SDH), which represents an intersection between the mitochondrial respiratory chain for which an important function in cardiopulmonary oxygen sensing has been demonstrated, and the Krebs cycle, a central element of α-KA metabolism. SDH subunit D heterozygous (SDHD(+/-)) and wild-type (WT) mice were housed at normoxia or hypoxia (10% O(2)) for 4 days or 3 weeks, and right ventricular pressure, right ventricle/(left ventricle+septum) ratio, cardiomyocyte ultrastructure, pulmonary vascular remodelling, ventricular complex II subunit expression, SDH activity and α-KA concentrations were analysed. In both strains, hypoxia induced increases in right ventricular pressure and enhanced muscularization of distal pulmonary arteries. Right ventricular hypertrophy was less severe in SDHD(+/-) mice although the cardiomyocyte ultrastructure and mitochondrial morphometric parameters were unchanged. Protein amounts of SDHA, SDHB and SDHC, and SDH activity were distinctly reduced in SDHD(+/-) mice. In normoxic SDHD(+/-) mice, α-ketoisocaproate concentration was lowered to 50% as compared to WT animals. Right/left ventricular concentration differences and the hypoxia-induced decline in individual α-KAs were less pronounced in SDHD(+/-) animals indicating that mitochondrial complex II participates in the adjustment of cardiac α-KA concentrations both under normoxic and hypoxic conditions. These characteristics are not related to the hemodynamic consequences of hypoxia-induced pulmonary vascular remodelling, since its extent and right ventricular pressure were not affected in SDHD(+/-) mice albeit right ventricular hypertrophy was attenuated.


Assuntos
Complexo II de Transporte de Elétrons/metabolismo , Hipóxia/enzimologia , Cetoácidos/metabolismo , Mitocôndrias/enzimologia , Miocárdio/enzimologia , Miocárdio/patologia , Animais , Pressão Sanguínea/fisiologia , Cardiomegalia/complicações , Cardiomegalia/enzimologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Regulação para Baixo , Ventrículos do Coração/enzimologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Heterozigoto , Hipóxia/complicações , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Camundongos , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Mutação/genética , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/ultraestrutura , Especificidade de Órgãos , Estabilidade Proteica , Subunidades Proteicas , Succinato Desidrogenase/metabolismo
15.
Cell Biochem Funct ; 27(8): 516-25, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19821261

RESUMO

Hypoxia-inducible factor-1alpha (HIF-1alpha) is a key transcription factor orchestrating hypoxic and inflammatory reactions. Here, we determined the impact of organ preservation solutions (Celsior; histidine-tryptophan-ketoglutarate solution, HTK; University of Wisconsin solution; UW), oxygen supply, and temperature on HIF-1alpha accumulation, recorded by Western blotting and immunocytochemistry, in the human hepatoma cell line HepG2. Generation of reactive oxygen species (ROS), NO, and cell viability were concomitantly assessed. At 4 degrees C, HIF-1alpha accumulation was not detectable. In normothermic (37 degrees C) cell culture medium (Dulbecco's Modified Eagle's Medium, DMEM), HepG2 cells accumulated HIF-1alpha even in normoxia (21% O(2)) which was not observed in either of the preservation solutions. This correlated to high generation of NO, a normoxic stabilizer of HIF-1alpha, and L-arginine content (substrate for NO synthesis) in DMEM, and low NO production and absence of L-arginine in preservation solutions. In normothermic hypoxia up to 24 h, intracellular HIF-1alpha accumulated in all conditions, but less in preservation solutions compared to DMEM. The inhibitory effect on accumulation and nuclear translocation was most prominent for HTK, the only solution containing the activator of HIF-1alpha degradation, alpha-ketoglutarate. Addition of other intermediates of the tricarbon acid cycle-succinate, fumarate, malate-did not alter HIF-1alpha accumulation, although succinate exhibited a beneficial effect on cell viability in cold storage. In conclusion, preservation solutions attenuate accumulation and nuclear translocation of the transcription factor HIF-1alpha, and this property is seemingly related to their chemical composition (L-arginine, alpha-ketoglutarate). Thus, it appears feasible to design preservation solution specifically to modify HIF-1alpha accumulation and nuclear translocation.


Assuntos
Núcleo Celular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Soluções para Preservação de Órgãos/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/fisiopatologia , Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Hipóxia/fisiopatologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/fisiopatologia , Preservação de Órgãos
16.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L837-45, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19684198

RESUMO

Accumulating evidence suggests a pivotal role of the calcitonin receptor-like receptor (CRLR) signaling pathway in preventing damage of the lung by stabilizing pulmonary barrier function. Intermedin (IMD), also termed adrenomedullin-2, is the most recently identified peptide targeting this receptor. Here we investigated the effect of hypoxia on the expression of IMD in the murine lung and cultured murine pulmonary microvascular endothelial cells (PMEC) as well as the role of IMD in regulating vascular permeability. Monoclonal IMD antibodies were generated, and transcript levels were assayed by quantitative RT-PCR. The promoter region of IMD gene was analyzed, and the effect of hypoxia-inducible factor (HIF)-1alpha on IMD expression was investigated in HEK293T cells. Isolated murine lungs and a human lung microvascular endothelial cell monolayer model were used to study the effect of IMD on vascular permeability. IMD was identified as a pulmonary endothelial peptide by immunohistochemistry and RT-PCR. Hypoxia caused an upregulation of IMD mRNA in the murine lung and PMEC. As shown by these results, HIF-1alpha enhances IMD promoter activity. Our functional studies showed that IMD abolished the increase in pressure-induced endothelial permeability. Moreover, IMD decreased basal and thrombin-induced hyperpermeability of an endothelial cell monolayer in a receptor-dependent manner and activated PKA in these cells. In conclusion, IMD is a novel hypoxia-induced gene and a potential interventional agent for the improvement of endothelial barrier function in systemic inflammatory responses and hypoxia-induced vascular leakage.


Assuntos
Permeabilidade Capilar , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Neuropeptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Peptídeos/metabolismo , Adrenomedulina/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Humanos , Técnicas In Vitro , Pulmão/irrigação sanguínea , Pulmão/citologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Células NIH 3T3 , Neuropeptídeos/genética , Hormônios Peptídicos/genética , Peptídeos/genética , Fosfoproteínas/metabolismo , Fosfosserina/metabolismo , Pressão , Regiões Promotoras Genéticas/genética , Frações Subcelulares/metabolismo , Ativação Transcricional/genética , Regulação para Cima
17.
Circulation ; 115(17): 2331-9, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17438150

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) is a life-threatening disease, characterized by vascular smooth muscle cell hyperproliferation. The calcium/calmodulin-dependent phosphodiesterase 1 (PDE1) may play a major role in vascular smooth muscle cell proliferation. METHODS AND RESULTS: We investigated the expression of PDE1 in explanted lungs from idiopathic PAH patients and animal models of PAH and undertook therapeutic intervention studies in the animal models. Strong upregulation of PDE1C in pulmonary arterial vessels in the idiopathic PAH lungs compared with healthy donor lungs was noted on the mRNA level by laser-assisted vessel microdissection and on the protein level by immunohistochemistry. In chronically hypoxic mouse lungs and lungs from monocrotaline-injected rats, PDE1A upregulation was detected in the structurally remodeled arterial muscular layer. Long-term infusion of the PDE1 inhibitor 8-methoxymethyl 3-isobutyl-1-methylxanthine in hypoxic mice and monocrotaline-injected rats with fully established pulmonary hypertension reversed the pulmonary artery pressure elevation, structural remodeling of the lung vasculature (nonmuscularized versus partially muscularized versus fully muscularized small pulmonary arteries), and right heart hypertrophy. CONCLUSIONS: Strong upregulation of the PDE1 family in pulmonary artery smooth muscle cells is noted in human idiopathic PAH lungs and lungs from animal models of PAH. Inhibition of PDE1 reverses structural lung vascular remodeling and right heart hypertrophy in 2 animal models. The PDE1 family may thus offer a new target for therapeutic intervention in pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/terapia , Diester Fosfórico Hidrolases/metabolismo , Artéria Pulmonar/enzimologia , 1-Metil-3-Isobutilxantina/farmacologia , 3',5'-GMP Cíclico Fosfodiesterases/metabolismo , Animais , Divisão Celular , Doença Crônica , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , DNA/biossíntese , Modelos Animais de Doenças , Humanos , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/terapia , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Inibidores de Fosfodiesterase/farmacologia , Artéria Pulmonar/citologia , Ratos , Regulação para Cima/fisiologia
18.
Respir Res ; 8: 15, 2007 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-17319968

RESUMO

BACKGROUND: Chronic hypoxia induces pulmonary arterial hypertension (PAH). Smooth muscle cell (SMC) proliferation and hypertrophy are important contributors to the remodeling that occurs in chronic hypoxic pulmonary vasculature. We hypothesized that rapamycin (RAPA), a potent cell cycle inhibitor, prevents pulmonary hypertension in chronic hypoxic mice. METHODS: Mice were held either at normoxia (N; 21% O2) or at hypobaric hypoxia (H; 0.5 atm; ~10% O2). RAPA-treated animals (3 mg/kg*d, i.p.) were compared to animals injected with vehicle alone. Proliferative activity within the pulmonary arteries was quantified by staining for Ki67 (positive nuclei/vessel) and media area was quantified by computer-aided planimetry after immune-labeling for alpha-smooth muscle actin (pixel/vessel). The ratio of right ventricle to left ventricle plus septum (RV/[LV+S]) was used to determine right ventricular hypertrophy. RESULTS: Proliferative activity increased by 34% at day 4 in mice held under H (median: 0.38) compared to N (median: 0.28, p = 0.028) which was completely blocked by RAPA (median HO+RAPA: 0.23, p = 0.003). H-induced proliferation had leveled off within 3 weeks. At this time point media area had, however, increased by 53% from 91 (N) to 139 (H, p < 0.001) which was prevented by RAPA (H+RAPA: 102; p < 0.001). RV/[LV+S] ratio which had risen from 0.17 (N) to 0.26 (H, p < 0.001) was attenuated in the H+RAPA group (0.22, p = 0.041). For a therapeutic approach animals were exposed to H for 21 days followed by 21 days in H +/- RAPA. Forty two days of H resulted in a media area of 129 (N: 83) which was significantly attenuated in RAPA-treated mice (H+RAPA: 92). RV/[LV+S] ratios supported prevention of PH (N 0.13; H 0.27; H+RAPA 0.17). RAPA treatment of N mice did not influence any parameter examined. CONCLUSION: Therapy with rapamycin may represent a new strategy for the treatment of pulmonary hypertension.


Assuntos
Hipertrofia Ventricular Direita/prevenção & controle , Hipóxia/tratamento farmacológico , Pulmão/irrigação sanguínea , Sirolimo/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/patologia , Hipóxia/complicações , Hipóxia/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Camundongos , Sirolimo/farmacologia
19.
Respir Res ; 7: 93, 2006 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-16808843

RESUMO

BACKGROUND: Acute alveolar hypoxia causes pulmonary vasoconstriction (HPV) which serves to match lung perfusion to ventilation. The underlying mechanisms are not fully resolved yet. The major vascular segment contributing to HPV, the intra-acinar artery, is mostly located in that part of the lung that cannot be selectively reached by the presently available techniques, e.g. hemodynamic studies of isolated perfused lungs, recordings from dissected proximal arterial segments or analysis of subpleural vessels. The aim of the present study was to establish a model which allows the investigation of HPV and its underlying mechanisms in small intra-acinar arteries. METHODS: Intra-acinar arteries of the mouse lung were studied in 200 mum thick precision-cut lung slices (PCLS). The organisation of the muscle coat of these vessels was characterized by alpha-smooth muscle actin immunohistochemistry. Basic features of intra-acinar HPV were characterized, and then the impact of reactive oxygen species (ROS) scavengers, inhibitors of the respiratory chain and Krebs cycle metabolites was analysed. RESULTS: Intra-acinar arteries are equipped with a discontinuous spiral of alpha-smooth muscle actin-immunoreactive cells. They exhibit a monophasic HPV (medium gassed with 1% O2) that started to fade after 40 min and was lost after 80 min. This HPV, but not vasoconstriction induced by the thromboxane analogue U46619, was effectively blocked by nitro blue tetrazolium and diphenyleniodonium, indicating the involvement of ROS and flavoproteins. Inhibition of mitochondrial complexes II (3-nitropropionic acid, thenoyltrifluoroacetone) and III (antimycin A) specifically interfered with HPV, whereas blockade of complex IV (sodium azide) unspecifically inhibited both HPV and U46619-induced constriction. Succinate blocked HPV whereas fumarate had minor effects on vasoconstriction. CONCLUSION: This study establishes the first model for investigation of basic characteristics of HPV directly in intra-acinar murine pulmonary vessels. The data are consistent with a critical involvement of ROS, flavoproteins, and of mitochondrial complexes II and III in intra-acinar HPV. In view of the lack of specificity of any of the classical inhibitors used in such types of experiments, validation awaits the use of appropriate knockout strains and siRNA interference, for which the present model represents a well-suited approach.


Assuntos
Pulmão/irrigação sanguínea , Músculo Liso Vascular/fisiopatologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animais , Antimicina A/farmacologia , Hipóxia Celular , Complexo II de Transporte de Elétrons/antagonistas & inibidores , Complexo II de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Técnicas In Vitro , Camundongos , Modelos Animais , Músculo Liso Vascular/metabolismo , Doadores de Óxido Nítrico/farmacologia , Nitrocompostos/farmacologia , Nitroazul de Tetrazólio/farmacologia , Propionatos/farmacologia , Artéria Pulmonar/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasoconstritores , Vasodilatação , Vasodilatadores/farmacologia
20.
Biochem Biophys Res Commun ; 342(3): 935-9, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16598846

RESUMO

Alpha-keto acids have recently been identified as potent regulators of cellular adaptations to hypoxia. Their actual intracellular concentrations under such conditions are unknown. Here, we determined concentrations of alpha-ketobutyrate, alpha-ketoglutarate, alpha-ketoisocaproate, alpha-ketoisovalerate, alpha-keto-beta-methylvalerate, phenylpyruvate, and pyruvate by a recently developed ultra-sensitive fluorescence HPLC method in ventricular myocardium of mice exposed to hypobaric hypoxia for up to 3 weeks. We observed characteristic alterations of cardiac alpha-keto acid concentrations that are specific for individual alpha-keto acids, show significant side differences (right versus left ventricles), and are suited to trigger some of the cardiac metabolic and structural adaptations to chronic hypoxia.


Assuntos
Hipóxia Celular/fisiologia , Ventrículos do Coração/metabolismo , Cetoácidos/metabolismo , Animais , Peso Corporal , Feminino , Masculino , Mercaptoetanol , Camundongos , Fenilenodiaminas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...