Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
J Renin Angiotensin Aldosterone Syst ; 21(2): 1470320320919607, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32370637

RESUMO

INTRODUCTION: The aims of this study were to assess the renal expression of angiotensin II type 1 receptor (AT1R), angiotensin II type 2 receptor (AT2R), and MAS receptor in human type 2 diabetic nephropathy (DN). MATERIALS AND METHODS: In total, 115 patients diagnosed with DN by renal biopsy were enrolled in this study. The protein expression levels of the AT1R, AT2R, and MAS receptors were assessed by immunohistochemistry. RESULTS: The protein expression levels of AT1R, AT2R, and MAS receptor in the renal biopsy tissue were correlated with the pathologic classification of DN. Tubulointerstitial AT1R expression in patients of class IIb was significantly stronger than control samples (p < 0.05). Expression of AT2R and MAS receptors were highest with class IIb DN patients. When DN patients were treated with AT1R blocker (ARB), the expression of AT1R was downregulated (p < 0.05), and the MAS receptor was upregulated in tubular interstitial (p < 0.05). CONCLUSIONS: Our results directly observed that renal expression levels of AT1R increase during the early stages of DN, ARB reducing AT1R while increasing MAS receptor. Therefore, ARB should be used as soon as possible in patients with DN.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Angiotensina II/biossíntese , Neuropatias Diabéticas/tratamento farmacológico , Neuropatias Diabéticas/metabolismo , Rim/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Adolescente , Adulto , Idoso , Biópsia , Neuropatias Diabéticas/patologia , Feminino , Humanos , Rim/patologia , Túbulos Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Receptor Tipo 1 de Angiotensina/biossíntese , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/genética , Adulto Jovem
2.
Hypertension ; 75(2): 500-509, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31813348

RESUMO

Bone marrow (BM) Ang II (angiotensin II) is a major participant in the regulation of hematopoiesis and immunity. The novel tissue substrate Ang-(1-12) [angiotensin-(1-12)] and its cleaving enzyme chymase are an essential source of Ang II production in cardiac tissue. We hypothesized this noncanonical chymase-mediated Ang II-producing mechanism exists in the BM tissue. Immunohistostaining and flow cytometry confirmed the presence of Ang-(1-12) immunoreaction in the BM of SD (Sprague Dawley) rats. Chymase-mediated Ang II-producing activity in BM was ≈1000-fold higher than ACE (angiotensin-converting enzyme)-mediated Ang II-producing activity (4531±137 and 4.2±0.3 fmol/min per mg, respectively; n=6; P<0.001) and 280-fold higher than chymase activity in the left ventricle of 16.3±1.7 fmol/min per mg (P<0.001). Adding a selective chymase inhibitor, TEI-F00806, eliminated almost all 125I-Ang II production. Flow cytometry demonstrated that delta median fluorescence intensity of chymase in cluster of differentiation 68 positive cells was significantly higher than that in cluster of differentiation 68 negative cells (1546±157 and 222±48 arbitrary units, respectively; P=0.0021). Cluster of differentiation 68 positive and side scatter low subsets, considered to be myeloid progenitors, express the highest chymase fluorescence intensity in rat BM. Chymase activity and cellular expression was similar in both male and female rats. In conclusion, myeloid lineage cells, especially myeloid progenitors, have an extraordinary Ang II-producing activity by chymase in the BM.


Assuntos
Angiotensina II/biossíntese , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Medula Óssea/metabolismo , Células Mieloides/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Feminino , Citometria de Fluxo , Imunidade Celular , Masculino , Microscopia Confocal , Modelos Animais , Células Mieloides/citologia , Ratos , Ratos Sprague-Dawley
3.
J Plast Reconstr Aesthet Surg ; 72(9): 1484-1493, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30528285

RESUMO

PURPOSE: To investigate the expression of components of the renin-angiotensin system (RAS): pro-renin receptor (PRR), angiotensin converting enzyme (ACE), angiotensin II receptor 1 (ATIIR1) and angiotensin II receptor 2 (ATIIR2) by the cancer stem cell (CSC) subpopulations in moderately differentiated head and neck cutaneous squamous cell carcinoma (MDHNCSCC). METHODOLOGY: 3,3-Diaminobenzidine (DAB) immunohistochemical (IHC) staining for PRR, ACE, ATIIR1 and ATIIR2 was performed on formalin-fixed paraffin-embedded sections of ten MDHNCSCC tissue samples. Immunofluorescence (IF) IHC staining was used to localise components of the RAS. Western blotting (WB) and RT-qPCR were performed on snap-frozen MDHNCSCC tissue samples and MDHNCSCC-derived primary cell lines to investigate protein transcription expression of these proteins, respectively. RESULTS: DAB IHC staining demonstrated the presence of PRR, ACE, ATIIR1 and ATIIR2 in all ten MDHNCSCC tissue samples. IF IHC staining showed expression of PRR and ATIIR2 by the OCT4+ cells, and ACE and ATIIR1 by the SOX2+ cells, within the tumour nests (TNs) and the peritumoural stroma (PTS). PRR, ACE, ATIIR1 and ATIIR2 were expressed by the endothelium of the microvessels within the PTS. WB confirmed protein expression for PRR, ACE and ATIIR1 in MDHNCSCC tissue samples and MDHNCSCC-derived primary cell lines. RT-qPCR showed transcriptional activation of PRR, ACE, ATIIR1 and ATIIR2 in MDHNCSCC tissue samples; and PRR, ACE, ATIIR1 but not ATIIR2, in MDHNCSCC-derived primary cell lines. CONCLUSION: PRR, ACE, ATIIR1 and ATIIR2 are expressed by the CSC subpopulations within the TNs, the PTS, and the endothelium of the microvessels within the PTS, in MDHNCSCC.


Assuntos
Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Células-Tronco Neoplásicas/patologia , Sistema Renina-Angiotensina/genética , Neoplasias Cutâneas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Idoso , Idoso de 80 Anos ou mais , Angiotensina II/biossíntese , Angiotensina II/genética , Biomarcadores Tumorais/biossíntese , Western Blotting , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Peptidil Dipeptidase A/biossíntese , Peptidil Dipeptidase A/genética , RNA Neoplásico/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo
4.
Crit Rev Eukaryot Gene Expr ; 28(2): 115-124, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30055537

RESUMO

The proliferative cell process that causes prostate enlargement, obstruction of the bladder outlet, and lower urinary tract symptoms (LUTS) is known as benign prostatic hyperplasia (BPH). The prevalence of BPH worldwide is approximately 10%, 20%, 50%, and 80% for men in their 30s, 40s, 60s, and 70s, respectively. Recent data have revealed that overactivation of the renin angiotensin aldosterone system increases the level of bioactive peptide hormone angiotensin II, which downregulates the ACE2-angiotensin 1-7/Mas receptor axis path and upregulates angiotensin receptor type 1-mediated signaling, which finally leads to a proliferation of cellular elements in the prostate. We have hypothesized the mechanism that reverses the downregulation of the ACE2-angiotensin 1-7/Mas receptor axis path and the upregulation of angiotensin receptor type 1-mediated signaling. Thus, we posit that ACE2, Ang-(1-7), and the Mas receptor could be novel therapeutic targets for treating BPH/LUTS.


Assuntos
Terapia de Alvo Molecular , Peptidil Dipeptidase A/genética , Hiperplasia Prostática/genética , Proteínas Proto-Oncogênicas/genética , Receptores Acoplados a Proteínas G/genética , Angiotensina II/biossíntese , Angiotensina II/genética , Enzima de Conversão de Angiotensina 2 , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/patologia , Proto-Oncogene Mas , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética , Transdução de Sinais/genética
5.
Am J Hypertens ; 30(12): 1211-1219, 2017 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-28992100

RESUMO

BACKGROUND: Prenatal lipopolysaccharide (LPS) exposure causes hypertension in rat offspring through an unknown mechanism. Here, we investigated the role of the intrarenal renin-angiotensin system (RAS) in hypertension induced by prenatal LPS exposure and also explored whether adipose tissue-derived mesenchymal stem cells (ADSCs) can ameliorate the effects of prenatal LPS exposure in rat offspring. METHODS: Sixty-four pregnant rats were randomly divided into 4 groups (n = 16 in each), namely, a control group and an LPS group, which were intraperitoneally injected with vehicle and 0.79 mg/kg LPS, respectively, on the 8th, 10th, and 12th days of gestation; an ADSCs group, which was intravenously injected with 1.8 × 107 ADSCs on the 8th, 10th, and 12th days of gestation; and an LPS + ADSCs group, which received a combination of the treatments administered to the LPS and ADSCs groups. RESULTS: Prenatal LPS exposure increased blood pressure, Ang II expression, Ang II-positive, monocyte and lymphocyte, apoptotic cells in the kidney, and induced renal histological changes in offspring; however, the LPS and control groups did not differ significantly with respect to plasma renin activity levels, Ang II levels, or renal function. ADSCs treatment attenuated the blood pressure and also ameliorated the other effects of LPS-treated adult offspring. CONCLUSIONS: Prenatal exposure to LPS activates the intrarenal RAS but not the circulating RAS and thus induces increases in blood pressure in adult offspring; however, ADSCs treatment attenuates the blood pressure increases resulting from LPS exposure and also ameliorates the other phenotypic changes induced by LPS treatment by inhibiting intrarenal RAS activation.


Assuntos
Tecido Adiposo/química , Rim/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Transplante de Células-Tronco Mesenquimais , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Sistema Renina-Angiotensina/efeitos dos fármacos , Angiotensina II/biossíntese , Angiotensina II/sangue , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea , Feminino , Rim/patologia , Testes de Função Renal , Células-Tronco Mesenquimais , Miocárdio/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Sprague-Dawley
6.
Kaohsiung J Med Sci ; 32(9): 439-45, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27638402

RESUMO

Acute pancreatitis (AP) usually causes acute lung injury, which is also known as acute pancreatitis associated lung injury (APALI). This study aimed to investigate whether captopril pretreatment was able to protect lung against APALI via inhibiting angiotensin II (Ang II) production and suppressing Rho/ROCK (Rho kinase) pathway in rats. Severe AP (SAP) was introduced to rats by bile-pancreatic duct retrograde injection of 5% sodium taurocholate. Rats were randomly divided into three groups. In the sham group, sham operation was performed; in the SAP group, SAP was introduced; in the pre-cpl + SAP group, rats were intragastrically injected with 5 mg/kg captopril 1 hour prior to SAP induction. Pathological examination of the lung and pancreas, evaluation of pulmonary vascular permeability by wet/dry ratio and Evans Blue staining, detection of serum amylase, Western blot assay for Ang II receptor type 1 (AT1), RhoA, ROCK (Rho kinase), and MLCK (myosin light chain kinase) were performed after the animals were sacrificed at 24 hours. After the surgery, characteristic findings of pancreatitis were observed, accompanied by lung injury. The serum amylase, Ang II, and lung expression of AT1, RhoA, ROCK, and MLCK increased dramatically in SAP rats. However, captopril pretreatment improved the histological changes, reduced the pathological score of the pancreas and lung, inhibited serum amylase and Ang II production, and decreased expression of AT1, RhoA, ROCK, and MLCK in the lung. These findings suggest that captopril pretreatment is able to protect the lung against APALI, which is, at least partially, related to the inhibition of Ang II production and the suppression of the Rho/ROCK pathway.


Assuntos
Angiotensina II/biossíntese , Captopril/uso terapêutico , Pulmão/patologia , Pancreatite/tratamento farmacológico , Pancreatite/prevenção & controle , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Doença Aguda , Amilases/sangue , Angiotensina II/sangue , Animais , Permeabilidade Capilar/efeitos dos fármacos , Captopril/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Lesão Pulmonar/patologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Pancreatite/sangue , Pancreatite/fisiopatologia , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Food Funct ; 7(5): 2409-19, 2016 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-27156533

RESUMO

This study aimed to determine the ability of peptides present in the non-digestible fraction (NDF) of common beans to decrease angiotensin II (AngII) through the blockade of RAS and its effect on the proliferation of HCT116 human colorectal cancer cells. Pure synthesized peptides GLTSK and GEGSGA and the peptide fractions (PF) of cultivars Azufrado Higuera and Bayo Madero were used. The cells were pretreated with pure peptides, PF or AGT at their IC50 or IC25 values, in comparison with the simultaneous treatment of peptides and AGT. For western blot and microscopy analysis, 100 µM and 0.5 mg mL(-1) were used for pure peptides and PF treatments, respectively. According to the ELISA tests, GLTSK and GEGSGA decreased (p < 0.05) the conversion rate of AGT to angiotensin I (AngI) by 38 and 28%, respectively. All the peptides tested reduced (p < 0.05) the conversion rate of AngI to AngII from 38 to 50%. When the cells were pretreated with both pure peptides and PF before exposure to AGT, the effectiveness inhibiting cell proliferation was higher than the simultaneous treatment suggesting their preventive effects. GLTSK and GEGSGA interacted with the catalytic site of renin, the angiotensin-I converting enzyme, and the AngII receptor, mainly through hydrogen bonds, polar, hydrophobic and cation-π interactions according to molecular docking. Through confocal microscopy, it was determined that GLTSK and GEGSGA caused the decrease (p < 0.05) of AngII-dependent STAT3 nuclear activation in HCT116 cells by 66 and 23%, respectively. The results suggest that peptides present in the common bean NDF could potentially ameliorate the effects of RAS overexpression in colorectal cancer.


Assuntos
Angiotensina II/biossíntese , Angiotensina II/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células HCT116/efeitos dos fármacos , Peptídeos/antagonistas & inibidores , Phaseolus/química , Extratos Vegetais/antagonistas & inibidores , Sistema Renina-Angiotensina/efeitos dos fármacos , Angiotensina I/análise , Angiotensina I/biossíntese , Angiotensina I/efeitos dos fármacos , Angiotensina II/análise , Domínio Catalítico/efeitos dos fármacos , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Digestão , Humanos , Concentração Inibidora 50 , Modelos Biológicos , Simulação de Acoplamento Molecular , Peptídeos/química , Peptidil Dipeptidase A/efeitos dos fármacos , Extratos Vegetais/química , Renina/efeitos dos fármacos
8.
Physiol Rep ; 4(8)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27122048

RESUMO

Short-term maternal corticosterone (Cort) administration at mid-gestation in the mouse reduces nephron number in both sexes while programming renal and cardiovascular dysfunction in 12-month male but not female offspring. The renal renin-angiotensin-aldosterone system (RAAS), functions in a sexually dimorphic manner to regulate both renal and cardiovascular physiology. This study aimed to identify if there are sex-specific differences in basal levels of the intrarenal RAAS and to determine the impact of maternal Cort exposure on the RAAS in male and female offspring at 6 months of age. While intrarenal renin concentrations were higher in untreated females compared to untreated males, renal angiotensin II concentrations were higher in males than females. Furthermore, basal plasma aldosterone concentrations were greater in females than males. Cort exposed male but not female offspring had reduced water intake and urine excretion. Cort exposure increased renal renin concentrations and elevated mRNA expression of Ren1, Ace2, and Mas1 in male but not female offspring. In addition, male Cort exposed offspring had increased expression of the aldosterone receptor, Nr3c2 and renal sodium transporters. In contrast, Cort exposure increased Agtr1a mRNA levels in female offspring only. This study demonstrates that maternal Cort exposure alters key regulators of renal function in a sex-specific manner at 6 months of life. These finding likely contribute to the disease outcomes in male but not female offspring in later life and highlights the importance of renal factors other than nephron number in the programming of renal and cardiovascular disease.


Assuntos
Corticosterona/toxicidade , Rim/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Caracteres Sexuais , Angiotensina II/biossíntese , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Proto-Oncogene Mas , RNA Mensageiro/análise , Renina/biossíntese , Sistema Renina-Angiotensina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Invest Ophthalmol Vis Sci ; 56(2): 855-64, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25564450

RESUMO

PURPOSE: To examine the expression of Angiotensin II (Ang II) and its type I, and type II receptors (AT1R, AT2R) in rabbit Tenon's capsule fibroblasts after trabeculectomy, and to investigate the effects of Ang II on cultured human Tenon's capsule fibroblasts (HTFs) proliferation, migration, phenotype transition, and extracellular matrix (ECM) synthesis. METHODS: In the rabbit, expression of Ang II, AT1R, and AT2R in Tenon's capsule fibroblasts of eyes after trabeculectomy was evaluated by immunohistochemistry. Ang II levels in aqueous humor and plasma were assessed by ELISA. Cultured HTFs, obtained from patients undergoing cataract surgery, were treated with Ang II, TGF-ß1, or vehicle control. Cell proliferation and migration were evaluated by Cell Counting Kit-8 and Transwell assay, and wound scratch assay, respectively. Protein expressions of α-smooth muscle actin (α-SMA) and fibronectin (FN) were measured by Western blot and immunofluorescence. Messenger RNA expressions of α-SMA and FN were measured by real-time PCR. RESULTS: In the rabbit, the expression of Ang II and AT1R increased from 1 day after surgery while AT2R increased from 7 days. In cultured HTFs, Ang II promoted cell proliferation and migration significantly (P < 0.05). Interestingly, the effect of 10(-7) M Ang II was more prominent than higher concentrations (10(-5) M; P < 0.05). Ang II also markedly induced the expression of α-SMA and FN, suggesting a phenotypic transition to myofibroblasts. CONCLUSIONS: Our results show that trabeculectomy alter the levels of Ang II and its receptors in Tenon's capsule fibroblasts, and that Ang II increase HTFs proliferation, migration, and phenotype transition, suggesting that Ang II may play a role in wound healing after trabeculectomy.


Assuntos
Angiotensina II/genética , Regulação da Expressão Gênica , Glaucoma/genética , RNA Mensageiro/genética , Cápsula de Tenon/metabolismo , Cicatrização , Angiotensina II/biossíntese , Animais , Humor Aquoso/metabolismo , Western Blotting , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibroblastos/metabolismo , Fibroblastos/patologia , Glaucoma/metabolismo , Glaucoma/cirurgia , Imuno-Histoquímica , Coelhos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Cápsula de Tenon/patologia , Trabeculectomia
10.
Genet Mol Res ; 13(4): 10562-73, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25511041

RESUMO

The renin-angiotensin system (RAS) influences cancer biology and is frequently dysregulated in malignancy. However, regulation of tumor local RAS remains poorly understood. Hypoxia is a hallmark of solid tumors and affects nearly every major aspect of cancer biology. Previous studies have shown that hypoxia can regulate RAS expression in somatic tissues and cells. The aim of this study was to investigate the influence of hypoxia on local RAS expression in mouse Lewis lung carcinoma (LLC) cells. For hypoxia treatment, LLC cells were cultured in a hypoxia incubator or treated with hypoxia-mimetic cobalt chloride. Hypoxia up-regulated angiotensin II, angiotensin-converting enzyme (ACE), and angiotensin II type 1 receptor (AT1R), and down-regulated ACE2 and angiotensin II type 2 receptor in LLC cells. Captopril, an ACE inhibitor, and losartan, an AT1R blocker, decreased expression of ACE and AT1R, but increased expression of ACE2 and angiotensin II type 2 receptor in LLC cells under hypoxia. Captopril and losartan also suppressed vascular endothelial growth factor-A expression in LLC cells under hypoxia. These findings suggest that hypoxia induces dysregulation of local RAS in LLC cells. The pathophysiological importance of hypoxia-induced RAS dysregulation and potentially therapeutic effects of RAS inhibitors on hypoxic tumor cells should be further examined.


Assuntos
Carcinoma Pulmonar de Lewis/genética , Hipóxia Celular , Sistema Renina-Angiotensina/genética , Angiotensina II/biossíntese , Enzima de Conversão de Angiotensina 2 , Animais , Carcinoma Pulmonar de Lewis/fisiopatologia , Carcinoma Pulmonar de Lewis/terapia , Regulação Neoplásica da Expressão Gênica , Humanos , Peptidil Dipeptidase A/biossíntese , Receptor Tipo 2 de Angiotensina/biossíntese
11.
Physiol Res ; 63(6): 723-32, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25157653

RESUMO

The relationship between obesity and renal lesions, especially in low estrogen levels, has been less documented. The aim of this study was to assess the renal changes in diet-induced obesity in ovariectomized rats. Wistar rats were ovariectomized or sham-operated and divided into four groups: sham-operated rats fed a standard diet (SSD); ovariectomized rats fed a standard diet (OSD); sham-operated rats fed a high-fat diet (SHFD); ovariectomized rats fed a high-fat diet (OHFD). Body weight and blood pressure were measured weekly. The rats were killed 24 weeks after initiation of standard or high-fat diet treatment, the kidneys were removed for immunohistochemical and histological studies. Blood and urine samples were collected to quantify sodium, potassium and creatinine. OHFD rats presented increases in visceral adipose tissue, serum insulin levels, blood pressure and proteinuria, and a decrease in fractional excretion of sodium as well. Histological and morphometric studies showed focal alterations in the renal cortex. Expression of macrophages, lymphocytes, nuclear factor-kappa B (NF-kappaB), Proliferating Cell Nuclear Antigen (PCNA), angiotensin II (ANG II) and vimentin was greater in OHFD rats than in control rats. Thus, these results demonstrate that the high-fat diet in ovariectomized rats promoted renal function and structure changes, renal interstitial infiltration of mononuclear cells and increased expression of ANG II and NF-kappaB.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Rim/patologia , Obesidade/patologia , Ovariectomia , Angiotensina II/biossíntese , Animais , Pressão Sanguínea/fisiologia , Peso Corporal , Ingestão de Energia , Feminino , Insulina/sangue , Macrófagos , NF-kappa B/biossíntese , Infiltração de Neutrófilos , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/sangue
12.
Ren Fail ; 36(4): 606-12, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24502693

RESUMO

Adriamycin (ADR) is commonly used for many solid tumor treatments. Its clinical utility is, however, largely limited by the adverse reactions, are known to be nephrotoxic. The mechanism by which it induces kidney damage is still not completely understood, but its nephrotoxicity might relate to increase reactive oxidant status (ROS), mitochondrial dysfunction. Until now, neurohormonal activation of it is unclear. ADR might activate the renin angiotensin system. Angiotensin-II also induced ROS and mitochondrial dysfunction. The aim of this study was to investigate whether angiotensin-II production inhibition has the protective effect on attenuation of mitochondrial function in rats with acute ADR-nephrotoxicity or not. Rats were divided into five groups as a control, ADR, co-treated ADR with captopril (CAP), co-treated ADR with Aliskren, co-treated ADR with both CAP and Aliskren groups. Creatinine kinase (CK) levels were measured at the end of treatment period. The kidneys were homogenized and biochemical measurements were made in mitochondria, cytosol. Mitochondria membrane potential (MMP) and ATP levels were determined. ADR increased CK levels and oxidative stress in mitochondria too (p<0.05). ADR significantly decreased MMP and ATP level in kidney mitochondria (p<0.05). Co-administration with ADR and Aliskren and CAP improved the dissipation of MMP (p<0.05). The decrease in ATP level was restored by treatment with inhibitors of ACE and renin. We concluded that inhibitors of angiotensin-II are effective against acute ADR induced nephrotoxicity via the restoration of MMP and ATP production and prevention of mitochondrial damage in vivo.


Assuntos
Angiotensina II/biossíntese , Doxorrubicina/toxicidade , Nefropatias/fisiopatologia , Rim/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Amidas/farmacologia , Animais , Captopril/farmacologia , Creatina Quinase/sangue , Fumaratos/farmacologia , Rim/metabolismo , Nefropatias/induzido quimicamente , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Renina/antagonistas & inibidores , Sistema Renina-Angiotensina/efeitos dos fármacos
13.
Mol Biol Rep ; 41(2): 1141-50, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24390237

RESUMO

Abnormal proliferation of human mesangial cells was the earliest pathological character in chronic kidney disease and linked to the accumulation of extracellular matrix and glomerular sclerosis. Multifunctional Angiotensin (AngII) had been emerged as a key player in initiation and progression of fibrogenic processes in kidney. In mesangial cells, treatment with the proliferation stimulus AngII triggered the escalated cyclinD1 expression, where its association with HuR increased dramatically. In our study, it was demonstrated that both in vivo and in vitro HuR redistribution in dysregulated mesangial cell proliferation accompanied by an abundant cyclinD1 expression following the AngII treatment. ActinomycinD experiments revealed that AngII stabilized cyclinD1 mRNA in human mesangial cells via HuR. Furthermore, employing the RIP-Chip assay yielded cyclinD1 mRNA with a higher affinity to HuR in mesangial cells induced by AngII compared with the normal ones in vitro study. Analysis of a cyclinD1 mRNA directly implicated HuR in regulating cyclinD1 production: cyclinD1 translation increased in HuR-shuttling cells induced by AngII and declined in cells in which HuR levels were lowered by RNA interference. We proposed that the release of HuR-bound mRNAs via an AngII-cyclinD1-HuR regulatory axis was implicated in the evolution of proliferative kidney diseases, providing us a novel therapeutic strategy to treat glomerular disease.


Assuntos
Angiotensina II/metabolismo , Ciclina D1/metabolismo , Proteínas ELAV/metabolismo , Células Mesangiais/metabolismo , Angiotensina II/biossíntese , Células Cultivadas , Ciclina D1/genética , Proteínas ELAV/genética , Regulação da Expressão Gênica , Humanos , RNA Mensageiro/biossíntese , Proteínas de Ligação a RNA/genética
14.
Mol Cell Biochem ; 388(1-2): 75-84, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24287563

RESUMO

Vascular remodeling is characterized by the aggregation of vascular smooth muscle cells (VSMCs) in intima. Previous studies have demonstrated that dehydroepiandrosterone (DHEA), a steroid hormone, can reverse vascular remodeling. However, it is still far clear that whether and how DHEA participates in the modulation of VSMCs activation and vascular remodeling. VSMCs were obtained from the thoracic aorta of SD rats. Cell proliferation was evaluated by CCK-8 assay and BrdU assay. To measure VSMCs migration activity, a transwell chamber assay was performed. Quantitative real-time RT-PCR and western blot were used to explore the molecular mechanisms. ROS generation by VSMCs was measured by DCF fluorescence. NADPH oxidase activity and SOD activity were measured by the corresponding kits. NF-κB activity was detected by NF-κB luciferase reporter gene assay. A rat carotid artery balloon injury model was built to evaluate the neointimal formation, and plasma PGF2 was measured by ELISA. Our results showed that DHEA significantly inhibited VSMCs proliferation after angiotensin (Ang II) stimulation by down-regulation of NADPH oxidase activity and ERK1/2 phosphorylation. Ang II can increase IL-6 and MCP-1 expression, but DHEA reverses these changes via inhibiting p38-MAPK/NF-κB (p65) signaling pathway. DHEA has no significant effects on VSMCs phenotype transition, but can reduce the neointimal to media area ratio after balloon injury. DHEA can alleviate oxidative stress and inflammation in VSMCs via ERK1/2 and NF-κB signaling pathway, but has no effect on VSMCs phenotype transition. Furthermore, DHEA attenuates VSMCs activation and neointimal formation after carotid injury in vivo. Taken together, DHEA might be a promising treatment for vascular injury under pathological condition.


Assuntos
Aorta Torácica/lesões , Lesões das Artérias Carótidas/tratamento farmacológico , Desidroepiandrosterona/farmacologia , Músculo Liso Vascular/patologia , Lesões do Sistema Vascular/tratamento farmacológico , Angiotensina II/biossíntese , Animais , Aorta Torácica/patologia , Lesões das Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Dinoprosta/sangue , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inflamação/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Músculo Liso Vascular/citologia , NADP/metabolismo , NADPH Oxidases/biossíntese , Neointima/metabolismo , Neointima/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Regeneração/efeitos dos fármacos , Superóxido Dismutase/biossíntese , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/biossíntese , Lesões do Sistema Vascular/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
15.
Am J Physiol Heart Circ Physiol ; 305(8): H1141-8, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23934850

RESUMO

Cathepsin G is a serine protease with a broad range of catalytic activities, including production of angiotensin II, degradation of extracellular matrix and cell-cell junctions, modulation of chemotactic responses, and induction of apoptosis. Cathepsin G mRNA expression is increased in human coronary atheroma vs. the normal vessel. To assess whether cathepsin G modulates atherosclerosis, cathepsin G knockout (Cstg(-/-)) mice were bred with apolipoprotein E knockout (Apoe(-/-)) mice to obtain Ctsg(+/-)Apoe(-/-) and Ctsg(+/+)Apoe(-/-) mice. Heterozygous cathepsin G deficiency led to a 70% decrease in cathepsin G activity in bone marrow cells, but this reduced activity did not impair generation of angiotensin II in bone marrow-derived macrophages (BMDM). Atherosclerotic lesions were compared in male Cstg(+/-)Apoe(-/-) and Cstg(+/+)Apoe(-/-) mice after 8 wk on a high-fat diet. Plasma cholesterol levels and cholesterol distribution within serum lipoprotein fractions did not differ between genotypes nor did the atherosclerotic lesion areas in either the aortic root or aortic arch. Cstg(+/-)Apoe(-/-) mice, however, showed a lower percentage of complex lesions within the aortic root and a smaller number of apoptotic cells compared with Cstg(+/+)Apoe(-/-) littermates. Furthermore, apoptotic Cstg(-/-) BMDM were more efficiently engulfed by phagocytic BMDM than were apoptotic Ctsg(+/+) BMDM. Thus cathepsin G activity may impair efferocytosis, which could lead to an accumulation of lesion-associated apoptotic cells and the accelerated progression of early atherosclerotic lesions to more complex lesions in Apoe(-/-) mice.


Assuntos
Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Catepsina G/genética , Macrófagos/metabolismo , Fagocitose/genética , Placa Aterosclerótica/genética , Angiotensina II/biossíntese , Animais , Apolipoproteínas E/deficiência , Apoptose/genética , Aterosclerose/patologia , Dieta Hiperlipídica , Masculino , Camundongos , Camundongos Knockout , Placa Aterosclerótica/patologia
16.
Am J Physiol Regul Integr Comp Physiol ; 304(8): R588-98, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23427083

RESUMO

The role of intracellular ANG II in proximal tubules of the kidney remains poorly understood. We tested the hypothesis that proximal tubule-dominant transfer of AT(1a) receptors in the cortex mediates intracellular ANG II-induced blood pressure responses in AT(1a) receptor-deficient (Agtr1a-/-) mice. A GFP-tagged AT(1a) receptor, AT(1a)R/GFP, and an enhanced cyan fluorescent intracellular ANG II fusion protein, ECFP/ANG II, were expressed in proximal tubules of Agtr1a-/- mouse kidneys via the adenoviral transfer using a sodium and glucose cotransporter 2 promoter. Transfer of AT(1a)R/GFP alone or with ECFP/ANG II induced proximal tubule-dominant expression of AT(1a)R/GFP and/or ECFP/ANG II with a peak response at 2 wk. No significant AT(1a)R/GFP and/or ECFP/ANG II expression was observed in the glomeruli, medulla, or extrarenal tissues. Transfer of AT(1a)R/GFP alone, but not ECFP/ANG II, increased systolic blood pressure by 12 ± 2 mmHg by day 14 (n = 9, P < 0.01). However, cotransfer of AT(1a)R/GFP with ECFP/ANG II increased blood pressure by 18 ± 2 mmHg (n = 12, P < 0.01). Twenty-four hour urinary sodium excretion was decreased by day 7 with proximal tubule-dominant transfer of AT(1a)R/GFP alone (P < 0.01) or with AT(1a)R/GFP and ECFP/ANG II cotransfer (P < 0.01). These responses were associated with twofold increases in phosphorylated ERK1/2, lysate, and membrane NHE-3 proteins in freshly isolated proximal tubules (P < 0.01). By contrast, transfer of control CMV-GFP (a recombinant human adenovirus type 5 expresses enhanced green fluorescent protein under the control of a cytomegalovirus (CMV) promoter), ECFP/ANG II, or a scrambled control ECFP/ANG IIc alone in proximal tubules had no effect on all indices. These results suggest that AT(1a) receptors and intracellular ANG II in proximal tubules of the kidney play an important physiological role in blood pressure regulation.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/fisiologia , Túbulos Renais Proximais/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Vasoconstritores/farmacologia , Angiotensina II/biossíntese , Animais , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Dependovirus , Ingestão de Líquidos/fisiologia , Eletrólitos/urina , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Túbulos Renais Proximais/efeitos dos fármacos , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/biossíntese , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Tipo 1 de Angiotensina/fisiologia , Transportador 2 de Glucose-Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/biossíntese , Trocadores de Sódio-Hidrogênio/genética
17.
Clin Exp Metastasis ; 29(8): 915-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22552372

RESUMO

We examined the effects of hyperglycemic conditions on liver metastasis of colorectal cancer (CRC). Angiotensin (A)-II increased growth, invasion, and anti-apoptotic survival in HT29 and CT26 cells. In contrast, angiotensinogen (ATG) increased these features in HT29 cells but not in CT26 cells. HT29 cells expressed A-II type 1 receptor, chymase, and rennin, whereas CT26 cells did not express renin. Renin expression and ATG-induced cell growth, invasion, and survival induced and increased as glucose concentration increased in HT29 cells and also CT26 cells. An inhibitor of renin or chymase abrogated A-II production in HT29 cells. Reduction of hepatic ATG production by cholesterol-conjugated antisense S-oligodeoxynucleotide suppressed liver metastasis of HT29 cells. An examination of 121 CRC patients showed that diabetes in CRC cases was associated with higher blood HbA1c, higher renin and A-II concentrations in the primary tumors, and higher incidence of liver metastasis than in nondiabetic cases. These results suggest that diabetes-associated angiotensin activation enhances liver metastasis of CRC and may therefore provide a possible target for antimetastatic therapy in CRC.


Assuntos
Angiotensina II/metabolismo , Neoplasias do Colo/metabolismo , Complicações do Diabetes/metabolismo , Diabetes Mellitus/metabolismo , Neoplasias Hepáticas/secundário , Angiotensina I/biossíntese , Angiotensina I/metabolismo , Angiotensina II/biossíntese , Bloqueadores do Receptor Tipo 1 de Angiotensina II , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Quimases/biossíntese , Quimases/genética , Quimases/metabolismo , Neoplasias do Colo/complicações , Neoplasias do Colo/patologia , Glucose/análise , Hemoglobinas Glicadas/análise , Células HT29 , Humanos , Hiperglicemia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Oligorribonucleotídeos Antissenso , Interferência de RNA , RNA Interferente Pequeno , Receptor Tipo 1 de Angiotensina/metabolismo , Renina/genética , Renina/metabolismo
18.
Toxicology ; 291(1-3): 139-45, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22120037

RESUMO

Candesartan cilexetil, an angiotensin (Ang) II receptor 1 blocker was reported to suppress the myocardial damage in various cardiovascular complications but the mode by which it is effective in preventing the progression of dilated cardiomyopathy (DCM) is unknown. Emerging evidences suggest that, at least, part of the benefits observed with the use of AT1 receptor blockers could be attributed to the increased Ang (1-7) levels observed during administration of these agents. Identification of the novel components of the RAS, ACE2 and Ang (1-7) receptor mas, provided essential elements for considering the existence of a vasodilator arm of the RAS, represented by the ACE2-Ang (1-7)-mas axis. In this study, rat model of DCM was prepared by injection with porcine cardiac myosin. Twenty-eight days after immunization, candesartan cilexetil was administered intraperitoneally at 1 or 10mg/kg/day to rats for four weeks. Myocardial expression of Ang receptors and markers of calcium homeostasis, endoplasmic reticulum (ER) stress and apoptosis were measured by Western blotting and histopathological staining techniques. Candesartan improved the functional markers in a dose-dependent manner and also upregulated Ang (1-7), ACE2 and mas1 in the myocardium of DCM rats. Various ER stress and apoptosis markers were attenuated and the number apoptotic cells were significantly lower in the candesartan treated rats compared with those of the vehicle group. These findings suggest that candesartan treatment prevented the progression of DCM by activation of the counter regulatory arm of the RAS and possibly through modulation of ER stress and subsequently, cardiac apoptosis.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Angiotensina I/farmacologia , Apoptose/efeitos dos fármacos , Benzimidazóis/farmacologia , Compostos de Bifenilo/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Cardiopatias/induzido quimicamente , Cardiopatias/prevenção & controle , Miosinas/antagonistas & inibidores , Miosinas/toxicidade , Fragmentos de Peptídeos/farmacologia , Peptidil Dipeptidase A/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Tetrazóis/farmacologia , Angiotensina II/biossíntese , Enzima de Conversão de Angiotensina 2 , Animais , Proteínas Reguladoras de Apoptose/biossíntese , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/prevenção & controle , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Miocardite/induzido quimicamente , Miocardite/prevenção & controle , Proto-Oncogene Mas , Ratos , Ratos Endogâmicos Lew , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , Transdução de Sinais/fisiologia
19.
Braz J Med Biol Res ; 44(9): 933-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21956536

RESUMO

Ouabain, an endogenous digitalis compound, has been detected in nanomolar concentrations in the plasma of several mammals and is associated with the development of hypertension. In addition, plasma ouabain is increased in several hypertension models, and the acute or chronic administration of ouabain increases blood pressure in rodents. These results suggest a possible association between ouabain and the genesis or development and maintenance of arterial hypertension. One explanation for this association is that ouabain binds to the α-subunit of the Na(+) pump, inhibiting its activity. Inhibition of this pump increases intracellular Na(+), which reduces the activity of the sarcolemmal Na(+)/Ca(2+) exchanger and thereby reduces Ca(2+) extrusion. Consequently, intracellular Ca(2+) increases and is taken up by the sarcoplasmic reticulum, which, upon activation, releases more calcium and increases the vascular smooth muscle tone. In fact, acute treatment with ouabain enhances the vascular reactivity to vasopressor agents, increases the release of norepinephrine from the perivascular adrenergic nerve endings and promotes increases in the activity of endothelial angiotensin-converting enzyme and the local synthesis of angiotensin II in the tail vascular bed. Additionally, the hypertension induced by ouabain has been associated with central mechanisms that increase sympathetic tone, subsequent to the activation of the cerebral renin-angiotensin system. Thus, the association with peripheral mechanisms and central mechanisms, mainly involving the renin-angiotensin system, may contribute to the acute effects of ouabain-induced elevation of arterial blood pressure.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Cardiotônicos/farmacologia , Hipertensão/induzido quimicamente , Ouabaína/farmacologia , Angiotensina II/biossíntese , Animais , Cálcio/metabolismo , Cardiotônicos/administração & dosagem , Cardiotônicos/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Humanos , Hipertensão/metabolismo , Injeções Intravenosas , Norepinefrina/metabolismo , Ouabaína/administração & dosagem , Ouabaína/metabolismo , Peptidil Dipeptidase A/metabolismo , Ratos , Sistema Renina-Angiotensina/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/fisiologia
20.
Transl Res ; 158(4): 235-48, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21925120

RESUMO

Excessive renin-angiotensin system (RAS) activation within the kidney induces not only renal oxidative stress but also renal scarring and dysfunction. This study examined the effects of an angiotensin II (Ang II) type I receptor (AT1R) blocker (ARB) on the progression of renal injury in rat anti-glomerular basement membrane glomerulonephritis (GN), with a particular focus on the participation of glomerular RAS activation in glomerular structural alterations, inflammation, and oxidative stress. Nephritic rats were divided into 2 groups and treated with vehicle or ARB until day 28. Treatment with ARB improved proteinuria significantly in nephritic rats. Vehicle-treated nephritic rats developed crescentic GN accompanied by marked macrophage infiltration and the enhanced expression of glomerular α-smooth muscle actin (α-SMA), angiotensinogen (AGT), Ang II, AT1R, and NADPH oxidase (Nox2) on days 7 and 28 of GN. ARB improved pathologic alterations such as crescent formation and glomerulosclerosis, and it had a significant inhibitory effect on the levels of these parameters on day 28 of GN. Enhanced superoxide production in nephritic glomeruli was decreased also by ARB. Moreover, Ang II and transforming growth factor beta (TGF-ß) in the supernatant of cultured glomeruli was increased significantly in vehicle-treated nephritic rats whereas ARB inhibited the production of these compounds significantly on day 28. These results indicate that increased glomerular RAS activity and the resulting Ang II play important roles in progressive glomerular injury-the induction of oxidative stress and TGF-ß expression, and they suggest that AT1R blockade attenuates proteinuria and progressive glomerular remodeling via the suppression of glomerular RAS activation in GN.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Doença Antimembrana Basal Glomerular/tratamento farmacológico , Sistema Renina-Angiotensina/efeitos dos fármacos , Actinas/metabolismo , Angiotensina II/biossíntese , Angiotensinogênio/biossíntese , Animais , Doença Antimembrana Basal Glomerular/metabolismo , Doença Antimembrana Basal Glomerular/patologia , Membrana Basal Glomerular/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Glicoproteínas de Membrana/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Superóxidos/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Pesquisa Translacional Biomédica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA