Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ren Fail ; 46(1): 2349113, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38721900

RESUMO

BACKGROUND: Type 3 cardiorenal syndrome (CRS type 3) triggers acute cardiac injury from acute kidney injury (AKI), raising mortality in AKI patients. We aimed to identify risk factors for CRS type 3 and develop a predictive nomogram. METHODS: In this retrospective study, 805 AKI patients admitted at the Department of Nephrology, Second Hospital of Shanxi Medical University from 1 January 2017, to 31 December 2021, were categorized into a study cohort (406 patients from 2017.1.1-2021.6.30, with 63 CRS type 3 cases) and a validation cohort (126 patients from 1 July 2021 to 31 Dec 2021, with 22 CRS type 3 cases). Risk factors for CRS type 3, identified by logistic regression, informed the construction of a predictive nomogram. Its performance and accuracy were evaluated by the area under the curve (AUC), calibration curve and decision curve analysis, with further validation through a validation cohort. RESULTS: The nomogram included 6 risk factors: age (OR = 1.03; 95%CI = 1.009-1.052; p = 0.006), cardiovascular disease (CVD) history (OR = 2.802; 95%CI = 1.193-6.582; p = 0.018), mean artery pressure (MAP) (OR = 1.033; 95%CI = 1.012-1.054; p = 0.002), hemoglobin (OR = 0.973; 95%CI = 0.96--0.987; p < 0.001), homocysteine (OR = 1.05; 95%CI = 1.03-1.069; p < 0.001), AKI stage [(stage 1: reference), (stage 2: OR = 5.427; 95%CI = 1.781-16.534; p = 0.003), (stage 3: OR = 5.554; 95%CI = 2.234-13.805; p < 0.001)]. The nomogram exhibited excellent predictive performance with an AUC of 0.907 in the study cohort and 0.892 in the validation cohort. Calibration and decision curve analyses upheld its accuracy and clinical utility. CONCLUSIONS: We developed a nomogram predicting CRS type 3 in AKI patients, incorporating 6 risk factors: age, CVD history, MAP, hemoglobin, homocysteine, and AKI stage, enhancing early risk identification and patient management.


Assuntos
Injúria Renal Aguda , Síndrome Cardiorrenal , Nomogramas , Humanos , Feminino , Masculino , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/sangue , Estudos Retrospectivos , Pessoa de Meia-Idade , Fatores de Risco , Síndrome Cardiorrenal/diagnóstico , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/etiologia , Idoso , Medição de Risco/métodos , China/epidemiologia , Modelos Logísticos , Adulto
2.
Circ Res ; 128(10): 1468-1486, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33983837

RESUMO

Acute decompensated heart failure (ADHF) is one of the leading admission diagnoses worldwide, yet it is an entity with incompletely understood pathophysiology and limited therapeutic options. Patients admitted for ADHF have high in-hospital morbidity and mortality, as well as frequent rehospitalizations and subsequent cardiovascular death. This devastating clinical course is partly due to suboptimal medical management of ADHF with persistent congestion upon hospital discharge and inadequate predischarge initiation of life-saving guideline-directed therapies. While new drugs for the treatment of chronic HF continue to be approved, there has been no new therapy approved for ADHF in decades. This review will focus on the current limited understanding of ADHF pathophysiology, possible therapeutic targets, and current limitations in expanding available therapies in light of the unmet need among these high-risk patients.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Doença Aguda , Líquidos Corporais/fisiologia , Síndrome Cardiorrenal/complicações , Cardiotoxinas/uso terapêutico , Comorbidade , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/etiologia , Hospitalização , Humanos , Mediadores da Inflamação/metabolismo , Contração Miocárdica/fisiologia , Peptídeo Natriurético Encefálico/metabolismo , Alta do Paciente , Readmissão do Paciente , Sistema Renina-Angiotensina/fisiologia , Avaliação de Sintomas , Resistência Vascular , Vasoconstrição/fisiologia , Vasodilatadores/uso terapêutico
3.
Urologiia ; (2): 130-134, 2023 May.
Artigo em Russo | MEDLINE | ID: mdl-37401718

RESUMO

A lecture on the pathogenesis and treatment of cardiorenal syndrome, which is a combination of various variants of renal and heart failure, is presented in the article. Currently, there are five types of this syndrome. All of them are discussed in detail from the view of relevance for urological practice. In patients of the urological profile, II type, to a lesser extent III and V types of cardiorenal syndrome are most common. Moreover, type II, which is the simultaneous coexistence of chronic heart failure and chronic renal failure due to different (unrelated causal relationships) conditions, can significantly influence on the choice of surgical tactics. This question requires further research. Type III of cardiorenal syndrome, which is a cardiac complication of a prolonged acute phase of acute renal failure, in most cases can be prevented through drug treatment and timely renal replacement therapy. Type V cardiorenal syndrome, which represents a combined damage to the heart and kidneys within the same condition, apparently, occurs in urological practice in the most severe patients with metabolic syndrome, which allows to combine uric acid stones and other variants of gouty nephropathy into one nosology, naturally leading to progressive renal failure, ischemic heart disease and chronic heart failure. In the section on treatment tactics, it is mentioned that there are no standard approaches to the treatment of cardiorenal syndrome in the literature. The restrictions in the choice and dosing regimen of cardiotropic drugs due to renal failure are considered in detail. The importance of timely hemodialysis is especially emphasized. In conclusion, the authors suggest that the development of cardiorenal syndrome is due to the effect of potentiation with a significantly higher rate of progression of both renal and heart failure compared to isolated forms of both conditions.


Assuntos
Síndrome Cardiorrenal , Insuficiência Cardíaca , Falência Renal Crônica , Humanos , Síndrome Cardiorrenal/terapia , Síndrome Cardiorrenal/complicações , Insuficiência Cardíaca/terapia , Insuficiência Cardíaca/tratamento farmacológico , Rim , Falência Renal Crônica/complicações , Doença Crônica
4.
Georgian Med News ; (342): 54-57, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37991957

RESUMO

The purpose of this paper is to analyses the cases with cardiorenal syndrome, and the ratio of cardiovascular disease and COVID-19. Prospective methods were used to conduct this research, including the period (January 2020-December 2021). Cases of patients treated at the Nephrology Clinic at the University Clinical Center of Kosovo (UCCK) have been studied. The categorical variables were analyzed with the X² test and the Fisher exact test. The study included 120 patients with acute renal disease treated at the Nephrology Clinic at the University Clinical Center of Kosovo (UCCK), of which 46 (38.3%) female and 74 (61.6%) male. Of the 120 patients included in the study 4 were 18-34 years old, 8 were 35-49 years old, 30 were 50-64 years old, and 78 were > 65 years old. There is a strong link between cardiorenal syndrome and age. Regarding cardiorenal syndrome and its association with other diseases in this prospective study were found these concomitant diseases such as: diabetes mellitus type 2, secondary anemia, hypothyroidism, hyperparathyroidism, pneumonia, sepsis, ascites, mesenteric tumor, hyperkalemia, and Covid-19 Infection. There is a strong link between cardiorenal syndrome and COVID-19 Infection. In recent decades various studies have been done against the definition of cardiorenal syndrome, the understanding of pathophysiology, the use of new biomarkers that represent a new dimension in the diagnostic algorithm, and the difficulties in treating this syndrome.


Assuntos
Injúria Renal Aguda , COVID-19 , Síndrome Cardiorrenal , Humanos , Masculino , Feminino , Adolescente , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Síndrome Cardiorrenal/diagnóstico , Síndrome Cardiorrenal/epidemiologia , Síndrome Cardiorrenal/complicações , Estudos Prospectivos , COVID-19/complicações , Doença Crônica
5.
FASEB J ; 35(10): e21907, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34516679

RESUMO

This study investigated the effect of apela on renal function and anti-inflammatory effect on whole body and kidney tissue in mice with type I cardiorenal syndrome (CRS). The murine type I CRS model was established and apela was subcutaneously infused for two weeks. Cardiac and renal functions were evaluated by echocardiography and blood biochemistry, respectively. The systemic and renal inflammatory responses were examined with molecular biological and histological methods. Human renal glomerular endothelial cells (RGECs) were used to evaluate the adhesion effect of monocytes in vitro. Compared to mice from the control group (CRS + vehicle), the plasma levels of N-terminal pro-brain natriuretic peptide, blood urea nitrogen and creatinine were significantly decreased, while the mean left ventricular ejection fraction was increased in apela-treated CRS mice at the 4th week. The expression of monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α) in the circulation and kidney was decreased in apela-treated mice compared with control mice, and apela improved cardio-renal pathology in mice with type I CRS. Additionally, Apela significantly suppressed the expression of MCP-1, TNF-α, intercellular adhesion molecule-1 and vascular intercellular adhesion molecule-1 in RGECs induced by angiotensin II (Ang II), and inhibited the promoting effect of Ang II on the adhesion of THP-1 cells to RGECs. Western blot results showed that the expression of phosphorylated nuclear factor kappa B (phospho-NFκB) in CRS mice was increased, but the expression of phospho-NFκB was down-regulated after apela treatment. Furthermore, apela significantly inhibited the Ang II-mediated increase in phospho-NFκB expression in RGECs in vitro, but the administration of an apelin peptide jejunum receptor (APJ) inhibitor blocked the inhibitory effect of apela. This study revealed that apela improves cardiorenal function and reduces systemic and renal inflammatory response in type I CRS mice and the apela/APJ system may alleviate renal inflammatory responses by inhibiting the NFκB signalling pathway.


Assuntos
Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/patologia , Inflamação/complicações , Inflamação/prevenção & controle , Rim , Hormônios Peptídicos/metabolismo , Animais , Coração/fisiologia , Coração/fisiopatologia , Humanos , Inflamação/patologia , Rim/patologia , Rim/fisiologia , Rim/fisiopatologia , Glomérulos Renais/citologia , Camundongos , NF-kappa B/metabolismo , Fosforilação , Células THP-1
6.
J Am Soc Nephrol ; 32(6): 1513-1526, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33883251

RESUMO

BACKGROUND: Few studies have thoroughly investigated the causes of kidney graft loss (GL), despite its importance. METHODS: A novel approach assigns each persistent and relevant decline in renal function over the lifetime of a renal allograft to a standardized category, hypothesizing that singular or multiple events finally lead to GL. An adjudication committee of three physicians retrospectively evaluated indication biopsies, laboratory testing, and medical history of all 303 GLs among all 1642 recipients of transplants between January 1, 1997 and December 31, 2017 at a large university hospital to assign primary and/or secondary causes of GL. RESULTS: In 51.2% of the patients, more than one cause contributed to GL. The most frequent primary or secondary causes leading to graft failure were intercurrent medical events in 36.3% of graft failures followed by T cell-mediated rejection (TCMR) in 34% and antibody-mediated rejection (ABMR) in 30.7%. In 77.9%, a primary cause could be attributed to GL, of which ABMR was most frequent (21.5%). Many causes for GL were identified, and predominant causes for GL varied over time. CONCLUSIONS: GL is often multifactorial and more complex than previously thought.


Assuntos
Aloenxertos/fisiopatologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto , Falência Renal Crônica/cirurgia , Transplante de Rim , Adulto , Idoso , Aloenxertos/patologia , Aloenxertos/estatística & dados numéricos , Inibidores de Calcineurina/efeitos adversos , Síndrome Cardiorrenal/complicações , Bases de Dados Factuais , Morte , Feminino , Rejeição de Enxerto/prevenção & controle , Humanos , Imunidade Celular , Imunidade Humoral , Imunossupressores/uso terapêutico , Transplante de Rim/normas , Transplante de Rim/estatística & dados numéricos , Masculino , Adesão à Medicação/estatística & dados numéricos , Pessoa de Meia-Idade , Infecções por Polyomavirus/complicações , Recidiva , Estudos Retrospectivos , Taxa de Sobrevida , Linfócitos T , Trombose/complicações , Fatores de Tempo , Infecções Tumorais por Vírus/complicações
7.
Ren Fail ; 44(1): 1243-1262, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35930297

RESUMO

OBJECTIVES: Several studies have shown that cholecalciferol supplementation (25OHD-S) in chronic kidney disease (CKD) improves kidney injury by reducing fibrosis-related vascular calcification and declining apoptosis-linked nephron damage. METHODS: The oral 25OHD-S was evaluated in 60,000 IU/month/36 weeks versus in 2000 IU/d/24 weeks in CKD Stage 3 with serum 25OHD level < 20 ng/mL. The study was undertaken on 156 black subjects and 150 white subjects Southern Sahara (SS). All biomarkers of cardiometabolic (CMet) and cardiorenal (CRenal) syndrome, Renin-angiotensin-aldosterone system (RAAS) profile, secondary hyperparathyroidism (SHPT), N-terminal pro B-type natriuretic peptide (NT-proBNP), Troponin T (cTnT) and atherogenicity risk were assessed by biochemical methods. Estimate glomerular filtration rate (eGFR) by chronic CKD-EPI equation formula. Total serum vitamin D by liquid chromatography-tandem mass spectrometry (MS). RESULTS: Vitamin D deficiency alters in the same manner CMet, CRenal, and others biomarkers in both groups SS; however, these disorders are more acute in blacks compared to whites SS. Oral 25OHD-S a highlighted improvement of eGFR drop, SHPT decrease, decline proteinuria, and cardiac failure risk (NT-proBNP and cTnT) attenuation. Concomitantly, 25OHD-S normalizes Renin, Aldosterone, and Angiotensin System (RAAS) activity. Nevertheless, homocysteine and Lp (a) do not modulate by 25OHD-S. CONCLUSIONS: The oral vitamin D3 supplementation, according the dose, and the treatment duration does not like in black-skinned people versus to white-skinned inhabitants, while the 02 groups are native to the same Saharan environment. It emerge that a high intermittent dose through an extensive supplementation (60,000 IU/36 weeks) was more effective in black subjects. At opposite, a lower dose during a short period supplementation is sufficient (2000 IU/24 weeks) in white subjects.


Assuntos
Síndrome Cardiorrenal , Hiperparatireoidismo Secundário , Insuficiência Renal Crônica , Deficiência de Vitamina D , Biomarcadores , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/etnologia , Síndrome Cardiorrenal/etiologia , Colecalciferol/administração & dosagem , Colecalciferol/uso terapêutico , Suplementos Nutricionais , Humanos , Hiperparatireoidismo Secundário/complicações , Hiperparatireoidismo Secundário/etnologia , Troponina T
8.
Am J Physiol Renal Physiol ; 320(4): F569-F577, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33522414

RESUMO

Ischemic heart disease is the leading cause of death worldwide and is frequently comorbid with chronic kidney disease. Physiological communication is known to occur between the heart and the kidney. Although primary dysfunction in either organ can induce dysfunction in the other, a clinical entity known as cardiorenal syndrome, mechanistic details are lacking. Here, we used a model of experimental myocardial infarction (MI) to test effects of chronic cardiac ischemia on acute and chronic kidney injury. Surprisingly, chronic cardiac damage protected animals from subsequent acute ischemic renal injury, an effect that was accompanied by evidence of chronic kidney hypoxia. The protection observed post-MI was similar to protection observed in a separate group of healthy animals housed in ambient hypoxic conditions prior to kidney injury, suggesting a common mechanism. There was evidence that chronic cardiac injury activates renal hypoxia-sensing pathways. Increased renal abundance of several glycolytic enzymes following MI suggested that a shift toward glycolysis may confer renal ischemic preconditioning. In contrast, effects on chronic renal injury followed a different pattern, with post-MI animals displaying worsened chronic renal injury and fibrosis. These data show that although chronic cardiac injury following MI protected against acute kidney injury via activation of hypoxia-sensing pathways, it worsened chronic kidney injury. The results further our understanding of cardiorenal signaling mechanisms and have implications for the treatment of heart failure patients with associated renal disease.NEW & NOTEWORTHY Experimental myocardial infarction (MI) protects from subsequent ischemic acute kidney injury but worsens chronic kidney injury. Observed protection from ischemic acute kidney injury after MI was accompanied by chronic kidney hypoxia and increased renal abundance of hypoxia-inducible transcripts. These data support the idea that MI confers protection from renal ischemic injury via chronic renal hypoxia and activation of downstream hypoxia-inducible signaling pathways.


Assuntos
Injúria Renal Aguda/metabolismo , Síndrome Cardiorrenal/complicações , Hipóxia/metabolismo , Precondicionamento Isquêmico , Infarto do Miocárdio/complicações , Injúria Renal Aguda/complicações , Injúria Renal Aguda/patologia , Animais , Síndrome Cardiorrenal/fisiopatologia , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Rim/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo
9.
Biochem Biophys Res Commun ; 524(1): 184-189, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-31982132

RESUMO

Plasma aldosterone concentration increases in proportion to the severity of heart failure, even during treatment with renin-angiotensin system inhibitors. This study investigated alternative regulatory mechanisms of aldosterone production that are significant in heart failure. Dahl salt-sensitive rats on a high-salt diet, a rat model of heart failure with cardio-renal syndrome, had high plasma aldosterone levels and elevated ß3-adrenergic receptor expression in hypoxic zona glomerulosa cells. In H295R cells (a human adrenocortical cell line), hypoxia-induced ß3-adrenergic receptor expression. Hypoxia-mediated ß3-adrenergic receptor expression augmented aldosterone production by facilitating hydrolysis of lipid droplets though ERK-mediated phosphorylation of hormone-sensitive lipase, also known as cholesteryl ester hydrolase. Hypoxia also accelerated the synthesis of cholesterol esters by acyl-CoA:cholesterol acyltransferase, thereby increasing the cholesterol ester content in lipid droplets. Thus, hypoxia enhanced aldosterone production by zona glomerulosa cells via promotion of the accumulation and hydrolysis of cholesterol ester in lipid droplets. In conclusion, hypoxic zona glomerulosa cells with heart failure show enhanced aldosterone production via increased catecholamine responsiveness and activation of cholesterol trafficking, irrespective of the renin-angiotensin system.


Assuntos
Córtex Suprarrenal/patologia , Aldosterona/biossíntese , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Hipóxia/metabolismo , Hipóxia/patologia , Córtex Suprarrenal/efeitos dos fármacos , Animais , Síndrome Cardiorrenal/complicações , Catecolaminas/farmacologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Colesterol/metabolismo , Modelos Animais de Doenças , Humanos , Hipóxia/complicações , Masculino , Fosforilação/efeitos dos fármacos , Ratos Endogâmicos Dahl , Receptores Adrenérgicos beta 3/metabolismo , Esterol Esterase/metabolismo , Zona Glomerulosa/metabolismo , Zona Glomerulosa/patologia
10.
J Pharmacol Exp Ther ; 375(3): 398-405, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33008869

RESUMO

Glucocorticoids are extensively used for a variety of conditions, including those associated with dysregulation of immune and inflammatory responses as primary etiopathogenic factors. Indeed, the proinflammatory cytokine storm of coronavirus disease 2019 (COVID-19) is the latest condition for which the use of a glucocorticoid has been advocated. Recognition of serious adverse effects of glucocorticoids has led to research aimed at unraveling molecular basis by which they impact immune and inflammatory events with the ultimate objective of devising novel therapies to circumvent glucocorticoids-related adverse outcomes. Consequently, glucocorticoid-induced leucine zipper (GILZ) protein was discovered and is increasingly recognized as the pivotal regulator of the effects of glucocorticoids on immune and inflammatory responses. Importantly, the advent of GILZ-based options raises the prospect of their eventual therapeutic use for a variety of conditions accompanied with dysregulation of immune and inflammatory responses and associated target organ complications. Thus, the objective of this minireview is to describe our current understanding of the role of GILZ in the cardiovascular system and the kidney along with outcome of GILZ-based interventions on associated disorders. This information is also of relevance for emerging complications of COVID-19. SIGNIFICANCE STATEMENT: Glucocorticoid-induced leucine zipper (GILZ) was initially discovered as the pivotal mediator of immune regulatory/suppressive effects of glucocorticoids. Since the use of glucocorticoids is associated with serious adverse effects, GILZ-based formulations could offer therapeutic advantages. Thus, this minireview will describe our current understanding of the role of GILZ in the kidney and the cardiovascular system, which is of relevance and significance for pathologies affecting them, including the multiorgan complications of coronavirus disease 2019.


Assuntos
COVID-19/metabolismo , Síndrome Cardiorrenal/complicações , Sistema Cardiovascular/metabolismo , Coronavirus/metabolismo , Rim/metabolismo , Fatores de Transcrição/metabolismo , Animais , COVID-19/complicações , COVID-19/terapia , Regulação da Expressão Gênica , Glucocorticoides/metabolismo , Humanos , Zíper de Leucina , Macrófagos/metabolismo , Transporte Proteico , RNA Mensageiro , Receptores Toll-Like/metabolismo
11.
Am J Nephrol ; 51(1): 74-82, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31812955

RESUMO

BACKGROUND: We evaluated the incremental contribution of chronic kidney disease (CKD) to the risk of major adverse cardiovascular (CV) events (MACE), heart failure (HF), and all-cause mortality (ACM) in type 2 diabetes mellitus (T2DM) patients and its importance relative to the presence of other cardio-renal-metabolic (CaReMe) comorbidities. METHODS: Patients (≥40 years) were identified at the time of T2DM diagnosis from US (Humedica/Optum) and UK (Clinical Practice Research Datalink) databases. Patients were monitored post-diagnosis for modified MACE (myocardial infarction, stroke, ACM), HF, and ACM. Adjusted hazard ratios were obtained using Cox proportional-hazards regression to evaluate the relative risk of modified MACE, HF, and ACM due to CKD. Patients were stratified by the presence or absence of atherosclerotic CV disease (ASCVD) and age. RESULTS: Between 2011 and 2015, of 227,224 patients identified with incident T2DM, 40,063 (17.64%) had CKD. Regardless of prior ASCVD, CKD was associated with higher risk of modified MACE, HF, and ACM; this excess hazard was more pronounced in older patients with prior ASCVD. In time-to-event analyses in the overall cohort, patients with T2DM + CKD or T2DM + CKD + hypertension + hyperlipidemia had increased risks for modified MACE, HF, and ACM versus patients with T2DM and no CaReMe comorbidities. Patients with CKD had higher risks for and shorter times to modified MACE, HF, and ACM than those without CKD. CONCLUSION: In T2DM patients, CKD presence was associated with higher risk of modified MACE, HF, and ACM. This may have risk-stratification implications for T2DM patients based on background CKD and highlights the potential importance of novel renoprotective strategies.


Assuntos
Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/mortalidade , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/mortalidade , Diabetes Mellitus Tipo 2/complicações , Angiopatias Diabéticas/complicações , Angiopatias Diabéticas/mortalidade , Cardiomiopatias Diabéticas/complicações , Cardiomiopatias Diabéticas/mortalidade , Síndrome Metabólica/complicações , Síndrome Metabólica/mortalidade , Adulto , Idoso , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
12.
Ann Hepatol ; 19(2): 145-152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31594758

RESUMO

Renal dysfunction is a common finding in cirrhotic patients and has a great physiologic, and therefore, prognostic relevance. The combination of liver disease and renal dysfunction can occur as a result of systemic conditions that affect both the liver and the kidney, although primary disorders of the liver complicated by renal dysfunction are much more common. As most of the renal dysfunction scenarios in cirrhotic patients correspond to either prerenal azotemia or hepatorenal syndrome (HRS), physicians tend to conceive renal dysfunction in cirrhotic patients as mainly HRS. However, there are many systemic conditions that may cause both a "baseline" chronic kidney damage and a superimposed kidney dysfunction when this systemic condition worsens. The main aim of this article is to review some of the most important non prerenal non-HRS considerations regarding acute on chronic kidney dysfunction in cirrhotic patients, including renal manifestation of related to non-alcoholic steatohepatitis (NASH) viral hepatitis, the effect of cardiorenal syndrome in cirrhotics and corticosteroid-deficiency associated renal dysfunction.


Assuntos
Injúria Renal Aguda/metabolismo , Síndrome Cardiorrenal/metabolismo , Hepatite Viral Humana/metabolismo , Cirrose Hepática/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Insuficiência Renal Crônica/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Corticosteroides/deficiência , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/fisiopatologia , Hepatite Viral Humana/complicações , Hepatite Viral Humana/fisiopatologia , Humanos , Cirrose Hepática/complicações , Cirrose Hepática/fisiopatologia , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/fisiopatologia
13.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R563-R570, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30840486

RESUMO

The genetic background of a mouse strain determines its susceptibility to disease. C57BL/6J and Balb/CJ are two widely used inbred mouse strains that we found react dramatically differently to angiotensin II and high-salt diet (ANG II + Salt). Balb/CJ show increased mortality associated with anuria and edema formation while C57BL/6J develop arterial hypertension but do not decompensate and die. Clinical symptoms of heart failure in Balb/CJ mice gave the hypothesis that ANG II + Salt impairs cardiac function and induces cardiac remodeling in male Balb/CJ but not in male C57BL/6J mice. To test this hypothesis, we measured cardiac function using echocardiography before treatment and every day for 7 days during treatment with ANG II + Salt. Interestingly, pulsed wave Doppler of pulmonary artery flow indicated increased pulmonary vascular resistance and right ventricle systolic pressure in Balb/CJ mice, already 24 h after ANG II + Salt treatment was started. In addition, Balb/CJ mice showed abnormal diastolic filling indicated by reduced early and late filling and increased isovolumic relaxation time. Furthermore, Balb/CJ exhibited lower cardiac output compared with C57BL/6J even though they retained more sodium and water, as assessed using metabolic cages. Left posterior wall thickness increased during ANG II + Salt treatment but did not differ between the strains. In conclusion, ANG II + Salt treatment causes early restriction of pulmonary flow and reduced left ventricular filling and cardiac output in Balb/CJ, which results in fluid retention and peripheral edema. This makes Balb/CJ a potential model to study the adaptive capacity of the heart for identifying new disease mechanisms and drug targets.


Assuntos
Angiotensina II/metabolismo , Síndrome Cardiorrenal/fisiopatologia , Dieta , Hipertensão/fisiopatologia , Animais , Pressão Sanguínea/fisiologia , Síndrome Cardiorrenal/complicações , Insuficiência Cardíaca/fisiopatologia , Hipertensão/complicações , Hipertensão Pulmonar/complicações , Masculino , Camundongos Endogâmicos BALB C , Miocárdio/metabolismo , Cloreto de Sódio na Dieta/metabolismo , Cloreto de Sódio na Dieta/farmacologia , Fatores de Tempo , Desequilíbrio Hidroeletrolítico/tratamento farmacológico , Desequilíbrio Hidroeletrolítico/metabolismo
14.
Nephrol Dial Transplant ; 34(Suppl 3): iii36-iii44, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31800079

RESUMO

Renin-angiotensin-aldosterone system inhibitors (RAASi) are now a standard treatment in most patients with cardiovascular disease, especially in those with heart failure (HF). The European Society of Cardiology and the American College of Cardiology/American Heart Association gave a Class IA recommendation for the use of RAASi in the treatment of Classes II-IV symptomatic HF with reduced ejection fraction (HFREF), based on their strong clinical benefits of lowering all-cause mortality and HF hospitalizations in these subjects. However, RAASi therapy or adding mineralocorticoid receptor antagonists in subjects receiving background angiotensin-converting enzyme inhibitors or angiotensin receptor blockers may be associated with an increased risk of hyperkalemia (HK), especially in those with reduced kidney function. As a result, a significant proportion of these subjects either have RAASi dose reduced or more often discontinued when they develop HK. Discontinuation of RAASi in patients hospitalized with HFREF was associated with higher postdischarge mortality and rehospitalization rates, while optimal dosing of RAASi significantly reduced median hospital stays, outpatient visits and related costs. Thus, effective treatment is required to lower potassium level and maintain normokalemia in subjects with HF and reduced kidney disease who develop or are at risk of HK, thus enabling them to continue their RAASi therapy and maximize benefits from RAASi. In this review, we provide an up-to-date review of the prevalence and significance of HK in patients with cardiorenal syndrome, as well as their optimal management of HK with recent novel therapies.


Assuntos
Antagonistas de Receptores de Angiotensina/uso terapêutico , Síndrome Cardiorrenal/complicações , Gerenciamento Clínico , Hiperpotassemia/terapia , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Potássio/sangue , Sistema Renina-Angiotensina/fisiologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Síndrome Cardiorrenal/sangue , Humanos , Hiperpotassemia/sangue , Hiperpotassemia/tratamento farmacológico , Hiperpotassemia/etiologia
15.
Blood Purif ; 48(4): 289-298, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31454818

RESUMO

Left ventricular assist devices (LVADs) are increasingly used for the management of patients with advanced heart failure (AHF) due to their established salutary impact on hemodynamic status and survival benefit. Impairment in kidney function is common in the setting of AHF and is associated with adverse impact on the outcomes. Cardiorenal interactions represent a complex pattern in these patients rendering their care a challenge that needs to be addressed by multidisciplinary approaches. Following LVAD implantation, AHF patients have the potential to achieve marked improvement in kidney function due to increased cardiac output and kidney perfusion as well as reduction in renal venous congestion. However, a subset of these patients is also at risk for acute kidney injury and resurgence of kidney dysfunction on continued mechanical circulatory support. Herein, we provide an overview of various aspects of changes in kidney function pre- and post-LVAD implantation, review potential underlying pathophysiologic mechanisms, and the impact on the outcomes. Moreover, the currently available data on renal replacement therapy of LVAD-treated patients, whether in the acute setting or as a maintenance therapy, are discussed in detail along with areas of high clinical relevance where a clear gap in knowledge exists.


Assuntos
Injúria Renal Aguda/complicações , Síndrome Cardiorrenal/complicações , Insuficiência Cardíaca/complicações , Coração Auxiliar , Injúria Renal Aguda/fisiopatologia , Injúria Renal Aguda/terapia , Animais , Síndrome Cardiorrenal/fisiopatologia , Síndrome Cardiorrenal/terapia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Ventrículos do Coração/fisiopatologia , Coração Auxiliar/efeitos adversos , Humanos , Rim/fisiopatologia , Terapia de Substituição Renal
16.
J Biol Regul Homeost Agents ; 32(1): 163-166, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29504382

RESUMO

Cardiorenal syndrome (CRS) describes the concurrent failure of cardiac and renal function, each influencing the other. Malnutrition and cachexia frequently develop in patients with heart failure or kidney failure. However, no information is currently available on the prevalence of malnutrition in CRS patients. We studied CRS patients admitted to an internal medicine ward during a 5-month period and evaluated their clinical characteristics and nutritional status. Malnutrition risk was assessed by using the validated screening tool NRS-2002 whilst body composition was assessed by bioimpedance analysis and muscle function was measured by handgrip (HG) strength. Cardiac mass was also recorded. Length of stay, hospital readmission and 6-month mortality were registered. During the study period, 22 CRS patients were studied. Twenty patients were diagnosed with either CRS type 1 or CRS type 5. In CRS patients, fat-free mass showed a trend toward representing a protective factor for 6-month mortality (OR=0.904; p=0.06). Also, fat-free mass correlated with HG strength and cardiac ejection fraction. Malnutrition risk was diagnosed in 45% of the patients, whereas 8 patients met the definition of cachexia. Even without statistical significance, CRS patients with malnutrition had lower BMI (Body Mass Index) (p=0.038) and fat-free mass (p= n.s.). However, CRS malnutrition was associated to higher 6-month mortality (p= 0.05), and appears to negatively influence the outcome in CRS (OR= 9; p= 0.06). Our results show that malnutrition is prevalent in CRS patients and influences the clinical outcome. The assessment of nutritional status, and particularly body composition, should be implemented in daily practice of patients with CRS.


Assuntos
Síndrome Cardiorrenal , Força da Mão , Desnutrição , Estado Nutricional , Volume Sistólico , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/mortalidade , Síndrome Cardiorrenal/fisiopatologia , Impedância Elétrica , Feminino , Humanos , Masculino , Desnutrição/etiologia , Desnutrição/mortalidade , Desnutrição/fisiopatologia , Fatores de Risco
17.
Nephrology (Carlton) ; 23(5): 411-417, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28240799

RESUMO

AIM: The aim of the study is to determine whether the apparent benefit of revascularization of renal artery stenosis for 'flash' pulmonary oedema extends to heart failure patients without a history of prior acute pulmonary oedema. METHODS: A prospective study of patients with renal artery stenosis and heart failure at a single centre between 1 January 1995 and 31 December 2010. Patients were divided into those with and without previous acute pulmonary oedema/decompensation. Survival analysis compared revascularization versus medical therapy in each group using Cox regression adjusted for age, estimated glomerular filtration rate, blood pressure and co-morbidities. RESULTS: There were 152 patients: 59% male, 36% diabetic, age 70 ± 9 years, estimated glomerular filtration rate 29 ± 17 mL/min per 1.73 m2 ; 52 had experienced previous acute pulmonary oedema (34%), whereas 100 had no previous acute pulmonary oedema (66%). The revascularization rate was 31% in both groups. For heart failure without previous acute pulmonary oedema, the hazard ratio for death after revascularization compared with medical therapy was 0.76 (0.58-0.99, P = 0.04). In heart failure with previous acute pulmonary enema, the hazard ratio was 0.73 (0.44-1.21, P = 0.22). For those without previous acute pulmonary oedema, the hazard ratio for heart failure hospitalization after revascularization compared with medical therapy was 1.00 (0.17-6.05, P = 1.00). In those with previous acute pulmonary oedema, it was 0.51 (0.08-3.30, P = 0.48). CONCLUSION: The benefit of revascularization in heart failure may extend beyond the current indication of acute pulmonary oedema. However, findings derive from an observational study.


Assuntos
Angioplastia , Síndrome Cardiorrenal/complicações , Insuficiência Cardíaca/complicações , Edema Pulmonar/etiologia , Obstrução da Artéria Renal/terapia , Doença Aguda , Idoso , Idoso de 80 Anos ou mais , Angioplastia/efeitos adversos , Angioplastia/instrumentação , Angioplastia/mortalidade , Síndrome Cardiorrenal/diagnóstico , Síndrome Cardiorrenal/mortalidade , Síndrome Cardiorrenal/fisiopatologia , Distribuição de Qui-Quadrado , Doença Crônica , Comorbidade , Feminino , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Edema Pulmonar/diagnóstico , Edema Pulmonar/mortalidade , Edema Pulmonar/fisiopatologia , Obstrução da Artéria Renal/diagnóstico , Obstrução da Artéria Renal/mortalidade , Obstrução da Artéria Renal/fisiopatologia , Estudos Retrospectivos , Fatores de Risco , Stents , Fatores de Tempo , Resultado do Tratamento
18.
Georgian Med News ; (282): 65-73, 2018 Sep.
Artigo em Russo | MEDLINE | ID: mdl-30358543

RESUMO

Aim - to study the predictors of cardiovascular events in patients with chronic heart failure (CHF) with preserved left ventricular ejection fraction role of traditional and non-traditional risk factors, to analyze the prognostic role of hyponatremia. Analysis of the clinical features of CHF was provided by retrospective research of medical cards in 308 patients, with endpoint verification during long-term period from 2010 to 2015. Among the examined patients 81 addmitted to the intensive care unit. The study showed that in obesity patients with body mass index (BMI) more than 30 kg/m2, the incidence of CHF decompensation was higher (p<0,05), but a significant mortality in CHF is only possible with abdominal obesity. It was proved the significant increase of CHF decompensation at patients with diabetes mellitus (p<0,05), the gender specificity of the disease was the higher risk of cardiovascular mortality in males (p<0,05).It was found that the most often in patients with CHF decompensation was reduced systolic pressure with an absolute risk of 82,7%. It was established a significantly higher risk of cardiovascular mortality in CHF with a sodium level less than 135 mmol/l, an increase from 19,0 to 45,0%, the "cut-off point" was established at the Na+ level between 115.0-125.0 mmol/l. After analyzing of the "combined" risk with hyponatremia and obesity, cardiovascular mortality increased to 23,0%.In the group with severe decompensation of CHF, it was set the lower hemoglobin level (p<0,05), lower hematocrit (p<0,05), higher ESR (p<0,05) and total leukocyte count(p<0,05).Absolute risk of cardiovascular mortality with hemoglobin level before 120 g/l was 48,0% vs. 16,0% of patients with normal hemoglobin level. Significant factors, combined with frequent hospitalization were age over 65 years (p<0,05), body mass index more than 30 kg/m2 (p<0,05), III and IV functional classes of CHF (p<0,05),hemoglobin level less than 120 g/l (p<0,05), hyponatremia (p<0,05).It was set the reliable influence of combined hyponatremia and obesity on the frequency of hospitalizations in CHF patients with an increase of absolute risk with 55,0%, reliable RR and OR (p<0,05).


Assuntos
Insuficiência Cardíaca/diagnóstico , Hiponatremia/diagnóstico , Obesidade/diagnóstico , Idoso , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/diagnóstico , Doença Crônica , Feminino , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/mortalidade , Humanos , Hiponatremia/complicações , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Prognóstico , Risco , Fatores Sexuais
19.
Anesth Prog ; 63(1): 34-41, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26866410

RESUMO

Patients with multiple comorbid conditions visit the dental office every day, and although rare, complications from their conditions may occur during treatment. A case is presented of a 65-year-old African American woman with a history of severe cardiovascular disease, renal disease, and a reported local anesthetic allergy who experienced complete cardiovascular collapse during routine dental treatment from which she was successfully resuscitated. Treating clinicians should recognize the emerging symptoms and be proficient with a basic and advanced cardiac life support protocol to care for their patients safely and effectively until they can be transported to more advanced care facilities.


Assuntos
Assistência Odontológica , Insuficiência Cardíaca/complicações , Hipertensão/complicações , Falência Renal Crônica/complicações , Choque Cardiogênico/etiologia , Idoso , Síndrome Cardiorrenal/complicações , Reanimação Cardiopulmonar/métodos , Desfibriladores , Feminino , Parada Cardíaca/etiologia , Humanos , Insuficiência Respiratória/etiologia
20.
Curr Opin Nephrol Hypertens ; 24(2): 154-62, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25636143

RESUMO

PURPOSE OF REVIEW: The purpose of this study is to review current perspectives regarding the pathogenesis of cardiorenal syndrome (CRS) in chronic kidney disease (CKD), and current treatment guidelines for this condition. RECENT FINDINGS: The pathophysiological mechanisms underlying the development of CRS in CKD include neurohumoral, haemodynamic and CKD-related mechanisms. Recent evidence suggests that sympathetic nerve activity plays a role in CRS, but the SYMPLICITY HTN-3 trial failed to show a reduction of blood pressure after catheter-based renal denervation in patients with resistant hypertension. Kidney injury in patients with heart failure was previously considered to result from arterial underfilling due to low cardiac output, but the role of renal venous hypertension in this process has also recently been investigated. It would be useful to develop a reliable treatment option for CRS due to haemodynamic mechanism other than volume control using diuretics. Fibroblast growth factor 23 (FGF23) is a phosphaturic hormone that has recently been identified as a CKD-related factor affecting CRS. FGF23 treatment has both advantages and disadvantages in terms of CRS progression. SUMMARY: Multiple disorders underlie the development of CRS. Current treatment options include renin-angiotensin system blockade and volume control, but remain limited. A multidisciplinary approach is required to prevent CRS, including renal sympathetic denervation, treatment of renal venous hypertension and FGF23 treatment.


Assuntos
Síndrome Cardiorrenal/terapia , Rim/fisiopatologia , Guias de Prática Clínica como Assunto , Insuficiência Renal Crônica/terapia , Sistema Renina-Angiotensina/fisiologia , Animais , Síndrome Cardiorrenal/complicações , Síndrome Cardiorrenal/diagnóstico , Fator de Crescimento de Fibroblastos 23 , Humanos , Hipertensão/complicações , Hipertensão/terapia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA