Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
1.
Front Immunol ; 15: 1404384, 2024.
Article in English | MEDLINE | ID: mdl-38953035

ABSTRACT

Introduction: Schistosomiasis (SM) is a parasitic disease caused by Schistosoma mansoni. SM causes chronic inflammation induced by parasitic eggs, with collagen/fibrosis deposition in the granuloma process in the liver, spleen, central nervous system, kidneys, and lungs. Pulmonary arterial hypertension (PAH) is a clinical manifestation characterized by high pressure in the pulmonary circulation and right ventricular overload. This study investigated the production of functional autoantibodies (fAABs) against the second loop of the G-protein-coupled receptor (GPCR) in the presence of hepatic and PAH forms of human SM. Methods: Uninfected and infected individuals presenting acute and chronic manifestations (e.g., hepatointestinal, hepato-splenic without PAH, and hepato-splenic with PAH) of SM were clinically evaluated and their blood was collected to identify fAABs/GPCRs capable of recognizing endothelin 1, angiotensin II, and a-1 adrenergic receptor. Human serum was analyzed in rat cardiomyocytes cultured in the presence of the receptor antagonists urapidil, losartan, and BQ123. Results: The fAABs/GPCRs from chronic hepatic and PAH SM individuals, but not from acute SM individuals, recognized the three receptors. In the presence of the antagonists, there was a reduction in beating rate changes in cultured cardiomyocytes. In addition, binding sites on the extracellular domain functionality of fAABs were identified, and IgG1 and/or IgG3 antibodies were found to be related to fAABs. Conclusion: Our data suggest that fAABs against GPCR play an essential role in vascular activity in chronic SM (hepatic and PAH) and might be involved in the development of hypertensive forms of SM.


Subject(s)
Autoantibodies , Receptors, G-Protein-Coupled , Autoantibodies/immunology , Autoantibodies/blood , Humans , Animals , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Rats , Male , Female , Adult , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/etiology , Middle Aged , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , Schistosomiasis mansoni/immunology , Schistosoma mansoni/immunology , Schistosomiasis/immunology
2.
Mol Biol Rep ; 51(1): 775, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904729

ABSTRACT

Acute leukemias (ALs) are the most common cancers in pediatric population. There are two types of ALs: acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Some studies suggest that the Renin Angiotensin System (RAS) has a role in ALs. RAS signaling modulates, directly and indirectly, cellular activity in different cancers, affecting tumor cells and angiogenesis. Our review aimed to summarize the role of RAS in ALs and to explore future perspectives for the treatment of these hematological malignancies by modulating RAS molecules. The database including Pubmed, Scopus, Cochrane Library, and Scielo were searched to find articles about RAS molecules in ALL and in pediatric patients. The search terms were "RAS", "Acute Leukemia", "ALL", "Angiotensin-(1-7)", "Pediatric", "Cancer", "Angiotensin II", "AML". In the bone marrow, RAS has been found to play a key role in blood cell formation, affecting several processes including apoptosis, cell proliferation, mobilization, intracellular signaling, angiogenesis, fibrosis, and inflammation. Local tissue RAS modulates tumor growth and metastasis through autocrine and paracrine actions. RAS mainly acts via two molecules, Angiotensin II (Ang II) and Angiotensin (1-7) [Ang-(1-7)]. While Ang II promotes tumor cell growth and stimulates angiogenesis, Ang-(1-7) inhibits the proliferation of neoplastic cells and the angiogenesis, suggesting a potential therapeutic role of this molecule in ALL. The interaction between ALs and RAS reveals a complex network of molecules that can affect the hematopoiesis and the development of hematological cancers. Understanding these interactions could pave the way for innovative therapeutic approaches targeting RAS components.


Subject(s)
Angiotensin II , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Renin-Angiotensin System , Humans , Renin-Angiotensin System/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Angiotensin II/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Signal Transduction , Angiotensin I/metabolism , Neovascularization, Pathologic/metabolism , Animals , Peptide Fragments/metabolism
3.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892464

ABSTRACT

In this study, we investigated whether severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein may modify angiotensin-converting enzyme 2 (ACE2) activity in the plasma, heart, kidney, liver, lung, and six brain regions (amygdala, brain stem, cortex, hippocampus, hypothalamus, and striatum) of diabetic and hypertensive rats. We determine ACE2 activity in the plasma and lysates of heart, kidney, liver, lung, and six brain regions. MLN-4760 inhibits ACE2 activity in the plasma and all organs. On the other hand, soluble ACE2 (sACE2) activity increased in the plasma of diabetic rats, and there was no change in the plasma of hypertensive rats. ACE2 activity was augmented in the liver, brain stem, and striatum, while it decreased in the kidney, amygdala, cortex, and hippocampus of diabetic rats. ACE2 activity increased in the kidney, liver, and lung, while it decreased in the heart, amygdala, cortex, and hypothalamus of hypertensive rats. We measured the ACE2 content via enzyme-linked immunosorbent assay and found that ACE2 protein levels increased in the heart, while it decreased in the plasma, kidney, brain stem, cortex, hippocampus, hypothalamus, and striatum of diabetic rats. ACE2 protein levels decreased in the brain stem, cortex, hippocampus, and hypothalamus of hypertensive rats. Our data showed that the spike protein enhanced ACE2 activity in the liver and lungs of diabetic rats, as well as in the heart and three of the brain regions (cortex, hypothalamus, and striatum) of hypertensive rats.


Subject(s)
Angiotensin-Converting Enzyme 2 , Hypertension , Spike Glycoprotein, Coronavirus , Animals , Angiotensin-Converting Enzyme 2/metabolism , Rats , Spike Glycoprotein, Coronavirus/metabolism , Male , Hypertension/metabolism , SARS-CoV-2 , Diabetes Mellitus, Experimental/metabolism , Brain/metabolism , Brain/enzymology , COVID-19/metabolism , COVID-19/virology , Carboxypeptidases/metabolism , Kidney/metabolism , Kidney/enzymology , Humans , Imidazoles , Leucine/analogs & derivatives
4.
Vasc Biol ; 6(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38843387

ABSTRACT

Abstract: Renin-angiotensin system plays a critical role in blood pressure control, and the abnormal activation of the AT1 receptor contributes to the development of renovascular hypertension. This study aimed to evaluate the underlying cellular signaling for AT1 receptor activation by Ang II and to compare this mechanism between aortas from 2K-1C and 2K rats. Effects of antagonists and inhibitors were investigated on Ang II-induced contractions in denuded or intact-endothelium aortas. The AT1 receptor antagonist abolished Ang II-induced contraction in 2K-1C and 2K rat aortas, while AT2 and Mas receptors antagonists had no effect. Endothelial nitric oxide synthase inhibition increased the maximal effect (Emax) of Ang II in 2K, which was not changed in 2K-1C aortas. It was associated with lower eNOS mRNA levels in 2K-1C. Endothelium removal increased the Emax of Ang II in 2K-1C and mainly in 2K rat aortas. Nox and COX inhibition did not alter Ang II-induced contraction in 2K and 2K-1C rat aortas. However, AT1 expression was higher in 2K-1C compared to 2K rat aortic rings, whereas expression of phosphorylated (active) IP3 receptors was lower in 2K-1C than in 2K rats. These results demonstrate that endothelium removal impairs Ang II-stimulated contraction in the aorta of 2K-1C rats, which is associated with the reduction of IP3 receptor phosphorylation and activation. In addition, eNOS plays a critical role in Ang II-induced contraction in 2K rat aortas. It is possible that the high Ang II plasma levels could desensitize AT1 receptor in 2K-1C rats, leading to impaired IP3 receptors activation.

5.
Arch Cardiol Mex ; 2024 May 09.
Article in Spanish | MEDLINE | ID: mdl-38724012

ABSTRACT

Hypertension is a major risk of morbidity and mortality in patients when it is uncontrolled. In spite of improved therapies currently available for blood pressure control, their complications are far away from being accomplished. Therefore, chronic renal failure is frequently observed in hypertensive patients. Thus, insights on mechanisms that may contribute to arterial pressure control should be studied to prevent life-threatening cardiovascular disorders. Purinergic receptors have been recognized in the physiopathology of hypertension; this review summarizes their participation in the renal abnormalities of the kidney in hypertension. Several studies have suggested the activation of renal purinergic receptors under an elevated interstitial ATP milieu as a fundamental pathway that leads to generation and maintained hypertension. Elevated ATP concentration alters fundamental mechanisms involved in the long-term control of blood pressure such as pressure natriuresis, autoregulation of glomerular filtration rate and renal blood flow, as well as increased tubule-glomerular feedback responses, overall, these alterations decrease sodium excretion; in addition, the expression of ATP receptors is modified. Under a genetical background, ATP induces the production of vasoactive compounds, decreases renal function and induces tubulointerstitial injury before glomerular damage. Simultaneously, a deleterious interaction between angiotensin II and purinergic receptors lead to the progression of renal damage.


La hipertensión arterial descontrolada es un factor de riesgo muy relevante para el desarrollo de complicaciones cardiovasculares graves. A pesar de los recursos disponibles en la actualidad, el control de la hipertensión arterial y sus complicaciones dista mucho de lograrse. Por ello, sus secuelas continúan siendo catastróficas, como la insuficiencia renal crónica. De ahí la relevancia de reconocer factores que pudieran modificarse para evitar esta complicación. Recientemente se ha propuesto que los receptores purinérgicos contribuyen en forma importante en las alteraciones renales que ocurren en la hipertensión arterial; en esta revisión se resume brevemente su papel. En varios estudios se ha demostrado que cuando existen concentraciones elevadas de ATP en el intersticio renal, la activación de los receptores purinérgicos constituye una vía fundamental en la generación y la persistencia de hipertensión arterial. Las concentraciones elevadas de ATP alteran mecanismos fundamentales asociados en el control de la presión arterial, como el mecanismo de natriuresis de presión, la autorregulación del flujo renal y la filtración glomerular, así como el aumento en la sensibilidad del mecanismo de retroalimentación tubuloglomerular. La alteración de estos mecanismos contribuye a la disminución de la excreción urinaria de sodio. Además, se modifica la expresión de receptores de ATP (purinérgicos). Bajo la influencia de alteraciones genéticas, el ATP estimula la producción de compuestos vasoactivos y en conjunto producen una disminución de la función renal y lesión tubulointersticial antes de que se lesione el glomérulo. Al mismo tiempo, la interacción de la angiotensina II y los receptores purinérgicos favorece la progresión del daño renal.

6.
Am J Physiol Cell Physiol ; 326(4): C1203-C1211, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38581656

ABSTRACT

Cardiometabolic diseases are often associated with heightened levels of angiotensin II (Ang II), which accounts for the observed oxidative stress, inflammation, and fibrosis. Accumulating evidence indicates a parallel upregulation of dipeptidyl dipeptidase 4 (DPP4) activity in cardiometabolic diseases, with its inhibition shown to mitigate oxidative stress, inflammation, and fibrosis. These findings highlight an overlap between the pathophysiological mechanisms used by Ang II and DPP4. Recent evidence demonstrates that targeted inhibition of DPP4 prevents the rise in Ang II and its associated molecules in experimental models of cardiometabolic diseases. Similarly, inhibitors of the angiotensin I-converting enzyme (ACE) or Ang II type 1 receptor (AT1R) blockers downregulate DPP4 activity, establishing a bidirectional relationship between DPP4 and Ang II. Here, we discuss the current evidence supporting the cross talk between Ang II and DPP4, along with the potential mechanisms promoting this cross regulation. A comprehensive analysis of this bidirectional relationship across tissues will advance our understanding of how DPP4 and Ang II collectively promote the development and progression of cardiometabolic diseases.


Subject(s)
Angiotensin II , Cardiovascular Diseases , Humans , Dipeptidyl Peptidase 4 , Peptidyl-Dipeptidase A , Receptor, Angiotensin, Type 1 , Inflammation , Fibrosis , Angiotensin I
7.
Arch Cardiol Mex ; 94(3): 373-380, 2024 03 13.
Article in Spanish | MEDLINE | ID: mdl-38478992

ABSTRACT

Aneurysms are clinical entities that can develop and affect human aorta; and although in most cases they have an asymptomatic course, these pathological dilatations can lead to a lethal outcome when rupture occurs, thus the establishment of predictors is crucial for death prevention. Essential events that take place in the vessel wall have been identified and described, such as inflammation, proteolysis, smooth muscle cell apoptosis, angiogenesis, and vascular remodeling. Porcine and ovine models have been useful for the development and evaluation of endovascular devices of the aorta. However, since the worldwide introduction and adoption of these minimally invasive techniques for aneurysm repair, there is lesser availability of diseased aortic tissue for molecular, cellular, and histopathological analysis, therefore over the last three decades it has been proposed various small species models that have allowed the focal induction of these lesions for the study of physiopathological mechanisms and possible useful biomarkers as diagnostic and therapeutic targets. The present review article presents and discusses the animal models available as their applications, characteristics, advantages, and limitations for the development of preclinical studies, and their importance in the comprehension of this pathology in humans.


Los aneurismas son una de las entidades clínicas que pueden desarrollarse y afectar la aorta humana. Aunque en la mayoría de los casos tienen un carácter asintomático, estas dilataciones patológicas pueden resultar letales cuando se presentan con ruptura, por lo que el reconocimiento de factores predictores de esta complicación es crucial para evitar muertes. Fisiopatológicamente se han identificado eventos esenciales que ocurren en la pared del vaso, como inflamación, proteólisis, apoptosis del músculo liso, angiogénesis y remodelación. Las grandes especies como porcinos y ovinos han sido de utilidad para el desarrollo y evaluación del desempeño de dispositivos endovasculares en la aorta, así como la remodelación; con el advenimiento y disposición de estas técnicas mínimamente invasivas para su reparación existe una menor disponibilidad de tejido aórtico para el análisis molecular, celular e histopatológico, por lo que en las últimas tres décadas se han propuesto e introducido distintos modelos que han permitido, mediante la inducción focal de estas lesiones, el estudio de los mecanismos fisiopatológicos y posibles biomarcadores de utilidad como dianas diagnósticas y terapéuticas. El presente artículo de revisión aborda tipos de modelos animales disponibles, así como sus aplicaciones, consideraciones, ventajas y limitaciones para el desarrollo de estudios preclínicos y su importancia en el entendimiento de esta patología en la especie humana.


Subject(s)
Aortic Aneurysm, Abdominal , Aortic Aneurysm, Thoracic , Disease Models, Animal , Animals , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Thoracic/surgery , Humans
8.
Arch Med Res ; 55(3): 102986, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38492325

ABSTRACT

Fatty liver is a multifactorial disease characterized by excessive accumulation of lipids in hepatocytes (steatosis), insulin resistance, oxidative stress, and inflammation. This disease has a major public health impact because it is the first stage of a chronic and degenerative process in the liver that can lead to steatohepatitis, cirrhosis, and liver cancer. Although this disease is mainly diagnosed in patients with obesity, type 2 diabetes mellitus, and dyslipidemia, recent evidence indicates that vasoactive hormones such as angiotensin II (ANGII) not only promote endothelial dysfunction (ED) and hypertension, but also cause fatty liver, increase adipose tissue, and develop a pro-steatotic environment characterized by a low-grade systemic pro-inflammatory and pro-oxidant state, with elevated blood lipid levels. The role of ANGII in lipid accumulation has been little studied, so this review aims to summarize existing reports on the possible mechanism of action of ANGII in inducing lipid accumulation in hepatocytes.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Humans , Angiotensin II , Diabetes Mellitus, Type 2/complications , Lipids , Liver , Non-alcoholic Fatty Liver Disease/etiology
9.
Natal; s.n; 21 mar. 2024. 52 p. ilus, tab, graf.
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: biblio-1566325

ABSTRACT

O sistema renina angiotensina (SRA) é de grande importância para o equilíbrio hídrico e regulação da pressão arterial do organismo, além de estar associado ao estimulo de vias próinflamatórias. Seu principal peptídeo é a angiotensina II, que interage principalmente com os receptores do tipo 1 (AT1) e do tipo 2 (AT2). Foi encontrado interrelação entre as doenças cardiovasculares e a periodontite. Este estudo teve como objetivo avaliar os aspectos moleculares em camundongos submetidos a um modelo experimental de periodontite, observando a influência dos receptores de Ang II tipo 1 (AT1(-)) e Ang II tipo 2 (AT2(-)) na periodontite. Métodos: A periodontite experimental foi induzida colocando-se uma ligadura com fio de nylon 5.0 ao redor do segundo molar superior esquerdo de camundongos knockoutAT1(-), AT2(-) e selvagem (WT), subdivididos 2 grupos para cada linhagem: sem ligadura e ligadura, totalizando seis grupos: três controles e três experimentais. Após 15 dias da indução da doença os animais foram submetidos à eutanásia. Com o intuito de avaliar se as variações genéticas teriam influência sobre a periodontite foram realizadas as análises de citocinas, peptídeos e enzimas foram analisados a partir de tecidos gengivais por ELISA e RT-PCR. Resultados: Os animais WT e AT2(-) apresentaram resultados semelhantes em relação às citocinas IL-1ß, IL-6, TNF-α, com aumento dos níveis em relação aos saudáveis (p < 0,001). Houve diferenças significativas em IL-ß entre os grupo AT1(-)-L e WT-L (p < 0,05), e em IL-6 e TNF-α os grupos AT1(-)-L apresentaram diferenças significativas (p < 0,001) tanto quando comparado aos grupo WT-L quanto aos grupos AT2(-)-L. Os níveis de IL-10 foram maiores em WT-L (p < 0,01), enquanto os grupos AT2(-) e AT1(-) não apresentaram alterações significativas em relação a essa citocina. Houve diferenças significativas em Angiotensina II entre os grupos AT2(-)-NL e AT2(-)-L (p < 0,01); e em Angiotensina 1-7 entre os grupos AT1(-)-L e AT2(-)-L (p < 0,05). Para TLR2 houve diferenças entre os grupos WT-NL/WT-L (p < 0,05); AT1(-)-NL/AT1(-)-L (p < 0,01) e AT2(-)-NL/AT2(-) - L (p < 0,01). Para o receptor MAS houve diferenças entre os grupos WT-NL/WT-L (p < 0,001) e AT2(-)-NL/AT2(-)-L (p < 0,001), e também em relação ao grupo WT-L/AT1(-)-L (p < 0,001) e AT1(-)-L/AT2(-)-L (p < 0,001). Para a expressão dos peptídeos ECA e ECA2, houve diferença estatística apenas para ECA entre os tipos de grupos WT-NL/WT-L (p < 0,001). Conclusão: Os animais do grupo AT1(-) apresentaram menor inflamação que as demais linhagens doentes, assim como uma menor expressão do receptor Mas e Ang 1-7. Além disso os animais dos grupos WT e AT2(-) demonstraram resultados próximos em diversas análises, evidenciando que o bloqueio do receptor AT1, sobre os efeitos moleculares, é mais positiva (AU).


The renin angiotensin system (RAS) is of great importance for water balance and regulation of blood pressure in the body, in addition to being associated with the stimulation of proinflammatory pathways. Its main peptide is angiotensin II, which interacts mainly with type 1 (AT1) and type 2 (AT2) receptors. An interrelationship was found between cardiovascular diseases and periodontitis. This study aimed to evaluate the molecular aspects in mice subjected to an experimental model of periodontal disease, observing the influence of Ang II type 1 (AT1(-)) and Ang II type 2 (AT2(-)) receptors on periodontitis. Methods: Experimental periodontitis was induced by placing a ligature with 5.0 nylon thread around the upper left second molar of AT1(-), AT2(-) and wild-type (WT) knockout mice, subdivided into 2 groups for each strain: without ligation and ligation, totaling six groups: three controls and three experimental. After 15 days of disease induction, the animals were euthanized. In order to evaluate whether genetic variations would have an influence on periodontal disease, analyzes of cytokines were carried out, peptides and enzymes were analyzed from gingival tissues by ELISA and RT-PCR. Results: WT and AT2(-) animals showed similar results in relation to the cytokines IL-1ß, IL-6, TNF-α, with increased levels compared to healthy ones (p < 0.001). There were significant differences in IL-ß between the AT1(-)-L and WT-L groups (p < 0.05), and in IL-6 and TNF-α the AT1(-)-L groups showed significant differences (p < 0.001) both when compared to the WT-L and AT2(-)-L groups. IL-10 levels were higher in WT-L (p < 0.01), while the AT2(-) and AT1(-) groups did not show significant changes in relation to this cytokine. There were significant differences in Angiotensin II between the AT2(-)-NL and AT2(-)-L groups (p < 0.01); and in Angiotensin 1-7 between the AT1(-)-L and AT2(-)-L groups (p < 0.05). For TLR2 there were differences between the WT-NL/WT-L groups (p < 0.05); AT1(-)-NL/AT1(-)-L (p < 0.01) and AT2(-)-NL/AT2(-)-L (p < 0.01). For the MAS receptor there were differences between the WT-NL/WT-L (p < 0.001) and AT2(-)-NL/AT2(- )-L (p < 0.001) groups, and also in relation to the WT-L group /AT1(-)-L (p < 0.001) and AT1(-)-L/AT2(-)-L (p < 0.001). For the expression of ACE and ACE2 peptides, there was a statistical difference only for ACE between the types of WT-NL/WT-L groups (p < 0.001). Conclusion: The animals in the AT1(-) group showed less inflammation than the other diseased lines, as well as a lower expression of the Mas and Ang 1-7 receptor. Furthermore, animals from the WT and AT2(-) groups demonstrated similar results in several analyses, showing that the blockade of the AT1 receptor, on molecular effects, is more positive (AU).


Subject(s)
Animals , Mice , Periodontal Diseases/pathology , Angiotensins , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/drug effects , In Vitro Techniques/methods , Epidemiology, Descriptive , Analysis of Variance , Statistics, Nonparametric
10.
Hypertension ; 81(5): 977-990, 2024 May.
Article in English | MEDLINE | ID: mdl-38372140

ABSTRACT

To celebrate 100 years of American Heart Association-supported cardiovascular disease research, this review article highlights milestone papers that have significantly contributed to the current understanding of the signaling mechanisms driving hypertension and associated cardiovascular disorders. This article also includes a few of the future research directions arising from these critical findings. To accomplish this important mission, 4 principal investigators gathered their efforts to cover distinct yet intricately related areas of signaling mechanisms pertaining to the pathogenesis of hypertension. The renin-angiotensin system, canonical and novel contractile and vasodilatory pathways in the resistance vasculature, vascular smooth muscle regulation by membrane channels, and noncanonical regulation of blood pressure and vascular function will be described and discussed as major subjects.


Subject(s)
Cardiovascular System , Hypertension , Humans , Signal Transduction , Blood Pressure , Renin-Angiotensin System/physiology , Angiotensin II/metabolism
11.
Clin Exp Nephrol ; 28(5): 359-374, 2024 May.
Article in English | MEDLINE | ID: mdl-38170299

ABSTRACT

BACKGROUND: Post-streptococcal glomerulonephritis (PSGN) is a consequence of the infection by group A beta-hemolytic streptococcus. During this infection, various immunological processes generated by streptococcal antigens are triggered, such as the induction of antibodies and immune complexes. This activation of the immune system involves both innate and acquired immunity. The immunological events that occur at the renal level lead to kidney damage with chronic renal failure as well as resolution of the pathological process (in most cases). Angiotensin II (Ang II) is a molecule with vasopressor and pro-inflammatory capacities, being an important factor in various inflammatory processes. During PSGN some events are defined that make Ang II conceivable as a molecule involved in the inflammatory processes during the disease. CONCLUSION: This review is focused on defining which reported events would be related to the presence of this hormone in PSGN.


Subject(s)
Angiotensin II , Glomerulonephritis , Streptococcal Infections , Streptococcus pyogenes , Humans , Glomerulonephritis/immunology , Glomerulonephritis/microbiology , Glomerulonephritis/etiology , Streptococcal Infections/immunology , Streptococcal Infections/complications , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Animals , Kidney/immunology , Kidney/pathology
12.
J Affect Disord ; 351: 349-355, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38286229

ABSTRACT

OBJECTIVE: Metformin (MET) is a drug used in the treatment of type 2 diabetes due to its insulin receptor sensitizing properties and anti-hepatic gluconeogenesis effect. One of the comorbidities in diabetes is the depression. This review aimed at summarizing the results of the available MET, depression and diabetes studies to clarify the possible role of MET in the depression during diabetes. METHODS: A bibliographic search on PubMed, Embase, PsycINFO, Web of Science, Cochrane Central for studies referring to MET, depression and diabetes. RESULTS: Several studies have associated depression to the chronic inflammation that characterizes diabetes. Additionally MET is an anti-inflammatory molecule that generally acts by activating AMPK and inhibiting the NF-kB factor. In the context of diabetes, MET can act directly as an anti-inflammatory drug as well as inhibiting other pro-inflammatory molecules. In this regard, MET may inhibit the pro-inflammatory effects of angiotensin II. By facilitating the action of insulin and reducing hepatic gluconeogenesis, MET reduces circulating glucose levels, decreasing the formation of advanced glycation end products and therefore inflammation. During diabetes, the gut microbiota and the permeability of the intestinal barrier are altered, causing high levels of circulating lipopolysaccharides (LPS), which induce inflammation. MET can normalize the microbiota and the intestinal barrier permeability reducing the levels of LPS and inflammation. Clinical and experimental studies show the anti-depressant effect of MET mediated by different mechanisms both at the peripheral level and in the central nervous system. CONCLUSION: Therefore, MET as an anti-inflammatory drug can decrease symptoms of depression and represents a therapeutic approach to improve the psychological state of patients with diabetes. Additionally, insulin also has an anti-inflammatory effect that could act together with MET.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Humans , Metformin/pharmacology , Metformin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Lipopolysaccharides , Insulin/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Inflammation/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
13.
Am J Med Sci ; 367(2): 128-134, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37984736

ABSTRACT

Cardiovascular diseases (CVD) are the main causes of death in hemodialysis patients, representing a public health challenge. We investigated the effect of different antihypertensive treatments on circulating levels of renin-angiotensin system (RAS) components in end-stage renal disease (ESRD) patients on hemodialysis. ESRD patients were grouped following the prescribed antihypertensive drugs: ß-blocker, ß-blocker+ACEi and ß-blocker+AT1R blocker. ESDR patients under no antihypertensive drug treatment were used as controls. Blood samples were collected before hemodialysis sessions. Enzymatic activities of the angiotensin-converting enzymes ACE and ACE2 were measured through fluorescence assays and plasma concentrations of the peptides Angiotensin II (Ang II) and Angiotensin-(1-7) [Ang-(1-7)] were quantified using mass spectrometry (LC-MS/MS). ACE activity was decreased only in the ß-blocker+ACEi group compared to the ß-blocker+AT1R, while ACE2 activity did not change according to the antihypertensive treatment. Both Ang II and Ang-(1-7) levels also did not change according to the antihypertensive treatment. We concluded that the treatment of ESRD patients on hemodialysis with different antihypertensive drugs do not alter the circulating levels of RAS components.


Subject(s)
Antihypertensive Agents , Kidney Failure, Chronic , Humans , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Angiotensin-Converting Enzyme 2/pharmacology , Chromatography, Liquid , Tandem Mass Spectrometry , Renin-Angiotensin System , Peptidyl-Dipeptidase A/metabolism , Peptides/pharmacology , Kidney Failure, Chronic/drug therapy , Angiotensin II/pharmacology , Peptide Fragments/metabolism , Renal Dialysis
14.
Int. j. cardiovasc. sci. (Impr.) ; 37: e20230163, 2024. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1558112

ABSTRACT

Abstract Background: Arterial hypertension (AH) is a chronic disease distributed worldwide, and the Angiotensin II receptor type 2 (AGTR2) gene variants are potential DNA markers to study in association with this disease. Objective: This systematic review (SR) aimed to identify single nucleotide variants in the AGTR2 gene as genetic markers associated with AH. Methods: The electronic databases MEDLINE, Web of Science, SCOPUS, Cochrane Central Register, EMBASE, SciELO, and TripDatabase were searched for research up to September 2023. Case-control studies with DNA variants in the AGTR2 gene associated with AH as the outcome were included in the review. Boolean connectors and keywords were used according to each database. Results: After diverse rounds of scrutiny, a final number of eight articles were included for 8911 participants, comprising 5451 cases and 3460 controls. A significant proportion of the selected studies were performed in Asian populations and were heterogeneous. Although 238 variants were shown in the gnomAD v2.1.1 database for September 2023, only six variants were identified in all the analyzed studies. Conclusions: The results obtained were not conclusive that a specific variant located in the AGTR2 gene has a strong association with AH. The study of this gene re-emerged last year as an essential target to investigate due to its participation in the development of agonist therapy to treat mild COVID-19 cases. Future studies with better statistical power are desirable to replicate the primary findings.

15.
Int J Mol Sci ; 24(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38068956

ABSTRACT

The objective of this study was to investigate whether the activity of enzymes involved in sphingolipid catabolism could be biomarkers to predict early renal damage in streptozotocin (STZ)-induced diabetic rats and Angiotensin II (Ang II)-induced hypertension rats. Diabetic and hypertensive rats had no changes in plasma creatinine concentration. However, transmission electron microscopy (TEM) analysis showed slight ultrastructural changes in the glomeruli and tubular epithelial cells from diabetic and hypertensive rats. Our results show that the acid sphingomyelinase (aSMase) and neutral sphingomyelinase (nSMase) activity increased in the urine of diabetic rats and decreased in hypertensive rats. Only neutral ceramidase (nCDase) activity increased in the urine of diabetic rats. Furthermore, the immunofluorescence demonstrated positive staining for the nSMase, nCDase, and sphingosine kinase (SphK1) in glomerular mesangial cells, proximal tubule, ascending thin limb of the loop of Henle, thick ascending limb of Henle's loop, and principal cells of the collecting duct in the kidney. In conclusion, our results suggest that aSMase and nCDase activity in urine could be a novel predictor of early slight ultrastructural changes in the nephron, aSMase and nCDase as glomerular injury biomarkers, and nSMase as a tubular injury biomarker in diabetic and hypertensive rats.


Subject(s)
Diabetes Mellitus, Experimental , Hypertension , Rats , Animals , Sphingomyelin Phosphodiesterase/metabolism , Diabetes Mellitus, Experimental/metabolism , Kidney/metabolism , Nephrons/metabolism , Sphingolipids
16.
Nutrients ; 15(21)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37960332

ABSTRACT

BACKGROUND: Endothelial dysfunction (ED) is a marker of vascular damage and a precursor of cardiovascular diseases such as hypertension, which involve inflammation and organ damage. Nitric oxide (NO), produced by eNOS, which is induced by pAKT, plays a crucial role in the function of a healthy endothelium. METHODS: A combination of subfractions SF1 and SF3 (C4) of the aqueous fraction from Cucumis sativus (Cs-Aq) was evaluated to control endothelial dysfunction in vivo and on HMEC-1 cells to assess the involvement of pAkt in vitro. C57BL/6J mice were injected daily with angiotensin II (Ang-II) for 10 weeks. Once hypertension was established, either Cs-AqC4 or losartan was orally administered along with Ang-II for a further 10 weeks. Blood pressure (BP) was measured at weeks 0, 5, 10, 15, and 20. In addition, serum creatinine, inflammatory status (in the kidney), tissue damage, and vascular remodeling (in the liver and aorta) were evaluated. Cs-AqC4 was also tested in vitro on HMEC-1 cells stimulated by Ang-II to assess the involvement of Akt phosphorylation. RESULTS: Cs-AqC4 decreased systolic and diastolic BP, reversed vascular remodeling, decreased IL-1ß and TGF-ß, increased IL-10, and decreased kidney and liver damage. In HMEC-1 cells, AKT phosphorylation and NO production were increased. CONCLUSIONS: Cs-AqC4 controlled inflammation and vascular remodeling, alleviating hypertension; it also improved tissue damage associated with ED, probably via Akt activation.


Subject(s)
Cucumis sativus , Hypertension , Peptide Hormones , Mice , Animals , Proto-Oncogene Proteins c-akt , Angiotensin II/pharmacology , Vascular Remodeling , Mice, Inbred C57BL , Hypertension/chemically induced , Hypertension/drug therapy , Blood Pressure , Inflammation , Plant Components, Aerial
17.
Biochem Pharmacol ; 217: 115840, 2023 11.
Article in English | MEDLINE | ID: mdl-37783376

ABSTRACT

Ethanol consumption activates renin-angiotensin-aldosterone system (RAAS), which plays a major role in the pro-contractile and hypertensive effects linked to ethanol. We hypothesized that ethanol consumption induces loss of the anticontractile effect of perivascular adipose tissue (PVAT)through RAAS-mediated mechanisms. We examined the contribution of angiotensin II type 1 receptors (AT1R) to ethanol-induced PVAT dysfunction. With this purpose, male Wistar Hannover rats were treated with ethanol 20 % (in volume ratio) and/or losartan (antagonist of AT1R; 10 mg/kg/day, gavage) for 9 weeks. Losartan prevented the increase in blood pressure and the loss of the anticontractile effect of PVAT induced by ethanol consumption. PVAT dysfunction occurred after 3 and 9 weeks of treatment with ethanol in an endothelium-dependent manner. Blockade of AT1R prevented ethanol-induced reduction of adiponectin levels in PVAT from ethanol-treated rats. Functional assays revealed that ethanol impaired the anticontractile effect of PVAT-derived angiotensin (1-7) and endothelial nitric oxide (NO). In conclusion, AT1R are implicated in ethanol-induced loss of the anticontractile effect of PVAT. In PVAT, AT1R activation decreases the production of adiponectin, a PVAT-derived factor that promotes vasorelaxation in an endothelium-dependent manner. In the endothelium, AT1R favors the production of superoxide (O2•-) leading to a reduction in NO bioavailability. These responses impair the vasodilator action induced by PVAT-derived angiotensin (1-7), which occurs via Mas receptors located in endothelial cells. Ethanol-induced PVAT dysfunction favors vascular hypercontractility, a response that could contribute to the hypertensive state associated with ethanol consumption.


Subject(s)
Adiponectin , Hypertension , Male , Rats , Animals , Adiponectin/pharmacology , Losartan/pharmacology , Ethanol/toxicity , Endothelial Cells , Vasoconstriction , Rats, Wistar , Adipose Tissue , Nitric Oxide/pharmacology
18.
Front Physiol ; 14: 1224505, 2023.
Article in English | MEDLINE | ID: mdl-37772054

ABSTRACT

Introduction: The medial preoptic area (mPOA) participates in thermoregulatory control and blood pressure modulation as shown by studies with electrical stimulation of this area or cobalt chloride injection, a non-selective synapse inhibitor. This study aimed to investigate whether angiotensin II (Ang II) and GABA could act or not in the mPOA to mediate the cardiovascular and micturition control pathways. Methods: Female Wistar rats were submitted to stereotaxic surgery for implantation of a guide cannula into the mPOA 7 days prior to the experiments. Afterwards, the animals were isoflurane- anesthetized and submitted to the catheterization of the femoral artery and vein and urinary bladder cannulation for mean arterial pressure (MAP), heart rate (HR), and intravesical pressure (IP) recordings, respectively. After the baseline MAP, HR, and IP recordings for 15 min, Ang II (0.1 nM, 1 µL), losartan (AT-1 receptor antagonist, 100 nM, 1 µL), GABA (50 mM, 1 µL) or saline (1 µL) were injected into the mPOA, and the variables were measured for additional 30 min. In a different group of rats, the AT-1 receptor, angiotensin II converting enzyme (ACE), and GABAa receptor gene expression was evaluated in mPOA samples by qPCR. The data are as mean ± SEM and submitted to One-way ANOVA (Tukey posttest) or paired Student t-test (P <0.05). Results: The injection of Ang II into the mPOA evoked a significant hypotension (-37±10 mmHg, n = 6, p = 0.024) and bradycardia (-47 ± 20 bpm, p = 0.030) compared to saline (+1 ± 1 mmHg and +6 ± 2 bpm, n = 6). A significant increase in IP was observed after Ang II injection into the mPOA (+72.25 ± 17.91%, p = 0.015 vs. -1.80 ± 2.98%, n = 6, saline). No significant changes were observed in MAP, HR and IP after the losartan injection in the mPOA compared to saline injection. Injection of GABA into the mPOA evoked a significant fall in MAP and HR (-68 ± 2 mmHg, n = 6, p < 0.0001 and -115 ± 14 bpm, n = 6, p = 0.0002 vs. -1 ± 1 mmHg and +4 ± 2 bpm, n = 6, saline), but no significant changes were observed in IP. The AT-1 receptor, ACE and GABAa receptor mRNA expression was observed in all mPOA samples. Discussion: Therefore, in female rats, Ang II mediated transmission in the mPOA is involved in the cardiovascular regulation and in the control of central micturition pathways. A phasic control dependent on AT-1 receptors in the mPOA seems to be involved in the regulation of those cardiovascular and intravesical 3 parameters. In contrast, GABAergic transmission in the mPOA participates in the pathways of cardiovascular control in anesthetized female rats, nevertheless, this neurotransmission is not involved in the micturition control.

19.
Can J Physiol Pharmacol ; 101(12): 620-629, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37747059

ABSTRACT

The purpose of this study was to characterize the role of ß1-AR signaling and its cross-talk between cardiac renin-angiotensin system and thyroid-hormone-induced cardiac hypertrophy. T3 was administered at 0.5 mg·kg-1·day-1 for 10 days in ß1-KOT3 and WTT3 groups, while control groups received vehicle alone. Echocardiography and myocardial histology was performed; cardiac and serum ANGI/ANGII and ANP and cardiac levels of p-PKA, p-ERK1/2, p-p38-MAPK, p-AKT, p-4EBP1, and ACE were measured. WTT3 showed decreased IVSTd and increased LVEDD versus WTsal (p < 0.05). ß1-KOT3 exhibited lower LVEDD and higher relative IVSTd versus ß1-KOsal, the lowest levels of ejection fraction, and the highest levels of cardiomyocyte diameter (p < 0.05). Cardiac ANP levels decreased in WTT3 versus ß1-KOT3 (p < 0.05). Cardiac ACE expression was increased in T3-treated groups (p < 0.05). Phosphorylated-p38 MAPK levels were higher in WTT3 versus WTsal or ß1-KOT3, p-4EBP1 was elevated in ß1-KO animals, and p-ERK1/2 was up-regulated in ß1-KOT3. These findings suggest that ß1-AR signaling is crucial for TiCH.


Subject(s)
Cardiomyopathy, Restrictive , Mice , Animals , Cardiomyopathy, Restrictive/metabolism , Cardiomyopathy, Restrictive/pathology , Mice, Knockout , Myocardium/metabolism , Thyroid Hormones , Receptors, Adrenergic/metabolism , Angiotensin II/pharmacology
20.
Behav Brain Res ; 454: 114639, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37652238

ABSTRACT

Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that not only entails alterations in fear behavior and anxiety but also includes neuroendocrine dysfunctions involving the hypothalamic pituitary adrenal (HPA) axis and the renin-angiotensin system. Recent preclinical studies demonstrate that activation of the angiotensin type 1 receptor (AT1R) in the paraventricular region of the hypothalamus (PVR) promotes anxiety-like behaviors and enables microglia proliferation. An increase in microglia and anxiety-like behavior also occurs in the PTSD animal model single-prolonged stress (SPS). In the present study, we tested whether AT1Rs contribute to the effects of SPS on behavior and microglia in brain structures important for HPA axis regulation and fear behavior. To test this, male and female animals were exposed to SPS and then given the oral AT1R antagonist candesartan beginning one week later. Candesartan did not alter auditory fear conditioning or extinction in SPS-exposed male or female animals. However, we found that the male animals exposed to SPS showed increased anxiety-like behavior, which was reversed by candesartan. In contrast, neither SPS nor candesartan altered anxiety-like behavior in the female animals. At the molecular level, SPS increased the cellular expression of AT1Rs in the PVR of male animals and candesartan reversed this effect, whereas AT1Rs in the PVR of females were unaltered by either SPS or candesartan. Iba1-expressing microglia increased in the PVR after SPS exposure and was reversed by candesartan in both sexes suggesting that SPS stimulates AT1Rs to increase microglia in the PVR. Collectively, these results suggest that the contribution of AT1Rs to the molecular and behavioral effects of SPS is sex-dependent.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Hypothalamo-Hypophyseal System , Female , Male , Animals , Angiotensin II Type 1 Receptor Blockers/pharmacology , Pituitary-Adrenal System , Benzimidazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL