Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 192
Filtrer
1.
Saudi Med J ; 45(7): 675-684, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38955454

RÉSUMÉ

OBJECTIVES: To evaluate the correlation between different attributes, levels of biomarkers, and the probability of developing cardiorenal syndrome (CRS) in patients who have been diagnosed with type 2 diabetes mellitus (T2DM) and liver cirrhosis (LC). The hypothesis suggests that liver illness may be linked to renal impairment, cardiac dysfunction, and the development of cardiorenal syndrome METHODS: The current study retrospectively assessed the medical records of patients who had LC and T2DM diagnoses and were hospitalized at Al Madina Al Munwara hospitals in 2022 and 2023. RESULTS: This research investigated T2DM patients with physician-confirmed to have LC. Poor glycemic control is indicated by high blood glucose and glycated hemoglobin (HbA1c) readings in research participants. High blood pressure, atherogenic plasma indicator (AIP), and obesity plagued most of these individuals. High creatinine, moderate estimated Glomerular Filtration Rate (eGFR) decline, and a modest urinary albumin-to-creatinine (UACR) rise were the most prevalent variables in LC and T2DM patients. Cardiorenal syndrome risk factors, including elevated blood pressure, triglyceride levels, body mass index (BMI), and high-sensitivity C-reactive protein (hs-CRP) concentrations, were identified through logistic regression. It has been demonstrated that the prevalence of these risk factors increases with age; women may be at a greater risk for developing CRS. Specific biomarker evaluations classified 108 (22.6%) LC and T2DM patients at high risk for chronic kidney disease (CKD), 100 (20%) at risk for cardiovascular disease (CVD), and 91 (18.2%) at risk for CRS. CONCLUSION: The current assessment included 500 patients with T2DM and LC. The risk factors for CRS identified in this study included elevated cholesterol and triglyceride levels, high BMI, and elevated blood pressure, with age being a significant factor, particularly in female patients. Early identification of these characteristics in patients with LC and T2DM could aid in mitigating the progression of chronic illnesses and their associated complications.


Sujet(s)
Marqueurs biologiques , Syndrome cardiorénal , Diabète de type 2 , Cirrhose du foie , Humains , Diabète de type 2/complications , Diabète de type 2/épidémiologie , Femelle , Cirrhose du foie/complications , Cirrhose du foie/épidémiologie , Mâle , Marqueurs biologiques/sang , Arabie saoudite/épidémiologie , Adulte d'âge moyen , Syndrome cardiorénal/épidémiologie , Syndrome cardiorénal/étiologie , Facteurs de risque , Études rétrospectives , Sujet âgé , Adulte , Indice de masse corporelle , Protéine C-réactive/analyse , Protéine C-réactive/métabolisme , Débit de filtration glomérulaire , Hémoglobine glyquée/analyse , Créatinine/sang
2.
Ren Fail ; 46(1): 2349113, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38721900

RÉSUMÉ

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.


Sujet(s)
Atteinte rénale aigüe , Syndrome cardiorénal , Nomogrammes , Humains , Femelle , Mâle , Atteinte rénale aigüe/diagnostic , Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/sang , Études rétrospectives , Adulte d'âge moyen , Facteurs de risque , Syndrome cardiorénal/diagnostic , Syndrome cardiorénal/complications , Syndrome cardiorénal/étiologie , Sujet âgé , Appréciation des risques/méthodes , Chine/épidémiologie , Modèles logistiques , Adulte
3.
Methods Mol Biol ; 2803: 145-162, 2024.
Article de Anglais | MEDLINE | ID: mdl-38676891

RÉSUMÉ

Cardiorenal syndrome (CRS) is a clinical disorder involving combined heart and kidney dysfunction, which leads to poor clinical outcomes. To understand the complex pathophysiology and mechanisms that lie behind this disease setting, and design/evaluate appropriate treatment strategies, suitable animal models are required. Described here are the protocols for establishing surgically induced animal models of CRS including important methods to determine clinically relevant measures of cardiac and renal function, commonly used to assess the degree of organ dysfunction in the model and treatment efficacy when evaluating novel therapeutic strategies.


Sujet(s)
Syndrome cardiorénal , Modèles animaux de maladie humaine , Syndrome cardiorénal/physiopathologie , Syndrome cardiorénal/diagnostic , Syndrome cardiorénal/étiologie , Animaux , Rats , Rein/physiopathologie , Rein/anatomopathologie , Coeur/physiopathologie , Mâle , Humains
4.
Sci Rep ; 13(1): 9130, 2023 06 05.
Article de Anglais | MEDLINE | ID: mdl-37277538

RÉSUMÉ

Cardiorenal syndromes type 1 and 2 are complex disorders in which cardiac dysfunction leads to kidney dysfunction. However, the mechanisms remain incompletely explained, during pulmonary hypertension in particular. The objective of this study is to develop an original preclinical model of cardiorenal syndrome secondary to a pulmonary hypertension in piglets. Twelve 2-month-old Large White piglets were randomized in two groups: (1) induction of pulmonary hypertension by ligation of the left pulmonary artery and iterative embolizations of the right lower pulmonary artery, or (2) Sham interventions. We evaluated the cardiac function using right heart catheterization, echocardiography and measurement of biochemistry markers). Kidney was characterized using laboratory blood and urine tests, histological evaluation, immunostainings for renal damage and repair, and a longitudinal weekly assessment of the glomerular filtration rate using creatinine-based estimation and intravenous injection of an exogenous tracer on one piglet. At the end of the protocol (6 weeks), the mean pulmonary artery pressure (32 ± 10 vs. 13 ± 2 mmHg; p = 0.001), pulmonary vascular resistance (9.3 ± 4.7 vs. 2.5 ± 0.4 WU; p = 0.004) and central venous pressure were significantly higher in the pulmonary hypertension group while the cardiac index was not different. Piglets with pulmonary hypertension had higher troponin I. We found significant tubular damage and an increase in albuminuria in the pulmonary hypertension group and negative correlation between pulmonary hypertension and renal function. We report here the first porcine model of cardiorenal syndrome secondary to pulmonary hypertension.


Sujet(s)
Syndrome cardiorénal , Cardiopathies , Défaillance cardiaque , Hypertension pulmonaire , Animaux , Syndrome cardiorénal/étiologie , Rein , Suidae
5.
Hum Exp Toxicol ; 42: 9603271231165678, 2023.
Article de Anglais | MEDLINE | ID: mdl-36960691

RÉSUMÉ

BACKGROUND: Syringaresinol processes anti-inflammatory and antioxidative activity. However, the effects of syringaresinol on cardiorenal fibrosis caused by cardiorenal syndrome type 2 (CRS2) are unclear. METHODS: Molecular docking predicted binding activity of syringaresinol to heat shock protein 90 (HSP90). The toxicity of a 4-weeks treatment with 20 mg/kg of syringaresinol was observed by measuring serum pro-inflammatory cytokines levels and by cardiorenal pathology. A CRS2 rad model was established by myocardial infarction using ligation over an 8 week-period. Rats were divided into five groups, including sham, CRS2, pimitespib, syringaresinol, and HSP90 + syringaresinol. Rats were received a 4-weeks daily treatment with 10 mg/kg pimitespib (a HSP90 inhibitor) or 20 mg/kg syringaresinol. Recombinant adeno-associated virus (rAAV) carrying a periostin (PE) promoter driving the expression of wild-type HSP90 (rAAV9-PE-HSP90, 1 × 1011 µg) was treated intravenously once in CRS2 model rats. Cardiorenal function and pathology were assessed. Expressions of HSP90 and TGF-ß1 in the myocardium and kidney were measured by immunohistochemistry and western blotting. RESULTS: Syringaresinol showed good binding activity with HSP90, and no signs of toxicity in rats following treatment. Pimitespib or syringaresinol significantly improved the cardiorenal function and fibrosis in rats with CRS2. Meanwhile, the rAAV9-PE-HSP90 injection obviously blocked the effects of syringaresinol. CONCLUSIONS: Syringaresinol targets HSP90 to suppress CRS2-induced cardiorenal fibrosis, providing a promising therapeutic drug for CRS2.


Sujet(s)
Syndrome cardiorénal , Rats , Animaux , Syndrome cardiorénal/traitement médicamenteux , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/métabolisme , Simulation de docking moléculaire , Rein/anatomopathologie , Fibrose
6.
Ren Fail ; 45(1): 2157285, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-36763000

RÉSUMÉ

Left ventricular assist device (LVAD) has been highlighted as a new treatment option in the end-stage heart failure (HF). Kidney outcome after LVAD in severe cardiorenal syndrome (CRS) patients requiring kidney replacement therapy (KRT) is unclear. We investigated the impact of preoperative KRT on kidney function and survival in LVAD patients with severe CRS. A total of 50 patients followed up for at least 1 year after LVAD implantation was analyzed. The primary outcomes were estimated glomerular filtration rate and survival rate. Patients were divided into two groups depending on in-hospital KRT before LVAD implantation: the control group (n = 33) and the KRT group (n = 17). Postoperative KRT was performed for 76.5% of patients in the KRT group, and all of them discontinued KRT before discharge. There were no statistically significant differences in the degree of eGFR decline in survivors according to preoperative KRT. Although there were no statistically significant differences in the degree of eGFR decline in survivors regardless of preoperative KRT, old age (ß -0.94, p < 0.01), preexisting chronic kidney disease (ß -21.89, p < 0.01), and high serum creatinine (ß -13.95, p < 0.01) were identified as independent predictors of post-LVAD eGFR decline. Mortality rate was higher, and more patients progressed to end-stage kidney disease in KRT group than control group. However, LVAD still can be considered as the treatment option in end-stage HF patients with severe CRS requiring KRT, especially in those with young age and previous normal kidney function.


Sujet(s)
Azotémie , Syndrome cardiorénal , Défaillance cardiaque , Dispositifs d'assistance circulatoire , Humains , Dispositifs d'assistance circulatoire/effets indésirables , Défaillance cardiaque/complications , Défaillance cardiaque/chirurgie , Études rétrospectives , Facteurs de risque , Rein , Syndrome cardiorénal/étiologie , Traitement substitutif de l'insuffisance rénale , Azotémie/étiologie , Résultat thérapeutique
8.
Curr Probl Cardiol ; 48(3): 101509, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36402213

RÉSUMÉ

Cardiorenal syndrome (CRS) is an increasingly recognized diagnostic entity associated with high morbidity and mortality among acutely ill heart failure (HF) patients with acute and/ or chronic kidney diseases (CKD). While traditionally viewed as a state of decline in glomerular filtration rate (GFR) due to decreased renal perfusion, mainly due to therapeutic interventions to relieve congestive in HF, recent insights into the underlying pathophysiologic mechanisms of CRS led to a broader definition and further classification of CRS into 5 distinct types. In this comprehensive review, we discuss the classification of CRS, highlighting the underlying common pathogenetic pathways of heart failure and kidney injury, including increased congestion, neurohormonal dysregulation, oxidative stress as well as inflammation, and cytokine storm that are particularly evident in COVID-19 patients with multiorgan failure and also in those with other disorders including sepsis, systemic lupus erythematosus and amyloidosis. In this review we also present the recent advances in the diagnostic strategies of CRS including cardiac and renal biomarkers as well as advanced cardiac and renal imaging techniques that are available to aid in the diagnosis as well as in the prognostication of this disorder. Finally, we discuss the various therapeutic options available to-date, including fluid optimization, hemofiltration, renal replacement therapy as well as the role of SGLT2 inhibitors in light of recent data from RCTs. It is important to note that, CRS population are either excluded or underrepresented, at best, in major RCTs and therefore, therapeutic recommendations are largely extrapolated from HF and CKD clinical trials.


Sujet(s)
COVID-19 , Syndrome cardiorénal , Défaillance cardiaque , Insuffisance rénale chronique , Humains , Syndrome cardiorénal/diagnostic , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/thérapie , COVID-19/complications , Défaillance cardiaque/diagnostic , Défaillance cardiaque/étiologie , Défaillance cardiaque/thérapie , Insuffisance rénale chronique/complications , Marqueurs biologiques
9.
G Ital Cardiol (Rome) ; 23(9): 716-727, 2022 Sep.
Article de Italien | MEDLINE | ID: mdl-36039723

RÉSUMÉ

Chronic kidney disease (CKD) and cardiovascular (CV) disease are highly prevalent conditions in the general population and are strictly connected to each other with a bidirectional interaction. In patients affected by CKD, the leading cause of morbidity and mortality is represented by CV disease, since CKD promotes the atherosclerotic process increasing inflammation, and modifying lipid and bone mineral metabolism. On the other side, a strict relationship exists between CKD and CV risk factors, which are prevalent in nephropathic patients and impose a stringent assessment of the risk of CV events in this population together with an optimized pharmacological approach, complicated by the coexistence of the two pathological conditions. The first part of this consensus document focuses on the mechanisms of cardio-renal damage and on the impact, as well as the management, of the main CV risk factors in the context of CKD.


Sujet(s)
Syndrome cardiorénal , Cardiologie , Maladies cardiovasculaires , Néphrologie , Insuffisance rénale chronique , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/prévention et contrôle , Maladies cardiovasculaires/étiologie , Maladies cardiovasculaires/prévention et contrôle , Consensus , Facteurs de risque de maladie cardiaque , Humains , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/épidémiologie , Facteurs de risque
10.
Ren Fail ; 44(1): 1243-1262, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-35930297

RÉSUMÉ

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.


Sujet(s)
Syndrome cardiorénal , Hyperparathyroïdie secondaire , Insuffisance rénale chronique , Carence en vitamine D , Marqueurs biologiques , Syndrome cardiorénal/complications , Syndrome cardiorénal/ethnologie , Syndrome cardiorénal/étiologie , Cholécalciférol/administration et posologie , Cholécalciférol/usage thérapeutique , Compléments alimentaires , Humains , Hyperparathyroïdie secondaire/complications , Hyperparathyroïdie secondaire/ethnologie , Troponine T
12.
Nephrol Dial Transplant ; 37(12): 2386-2397, 2022 11 23.
Article de Anglais | MEDLINE | ID: mdl-35438794

RÉSUMÉ

BACKGROUND: Cardiorenal syndromes (CRSs) are reputed to result in worse prognosis than isolated heart failure (HF) and chronic kidney disease (CKD). Whether it is true for all major outcomes over the long-term regardless of CRS chronology (simultaneous, cardiorenal and renocardiac CRS) is unknown. METHODS: The 5-year adjusted risk of major outcomes was assessed in this nationwide retrospective cohort study in all 385 687 with either CKD or HF (out of 5 123 193 patients who were admitted in a French hospital in 2012). RESULTS: Overall, 84.0% patients had HF and 8.9% had CKD (they had similar age, sex ratio, diabetes and hypertension prevalence), while 7.1% had CRS (cardiorenal: 44.6%, renocardiac: 14.5%, simultaneous CRS: 40.8%).The incidence of major outcomes was 57.3%, 53.0%, 79.2% for death; 18.8%, 10.9%, 27.5% for cardiovascular death; 52.6%, 34.7%, 64.3% for HF; 6.2%, 5.5%, 5.6% for myocardial infarction (MI); 6.1%, 5.8%, 5.3% for ischaemic stroke; and 23.1%, 4.8%, 16.1% for end-stage kidney disease (ESKD) for isolated CKD, isolated HF and CRS, respectively.As compared with isolated CKD or HF, the risk of death, cardiovascular death and HF was markedly increased in CRS, the worse phenotype being cardiorenal CRS, while the increased risk of MI and ischaemic stroke associated with CRS subtypes was statistically but not clinically significant. As compared with isolated CKD, the risk of ESKD was similar for cardiorenal CRS only and marginally increased for renocardiac and simultaneous CRS. We could not find a synergy between HF and CKD on major clinical outcomes in the whole population (n = 5 123 193 patients). CONCLUSIONS: The additional impact of CRS versus isolated HF or CKD on long-term kidney and cardiovascular risk is highly heterogenous, depending of the event considered and CRS chronology. No synergy between HF and CKD could be demonstrated.


Sujet(s)
Encéphalopathie ischémique , Syndrome cardiorénal , Défaillance cardiaque , Accident vasculaire cérébral ischémique , Défaillance rénale chronique , Infarctus du myocarde , Insuffisance rénale chronique , Accident vasculaire cérébral , Humains , Syndrome cardiorénal/épidémiologie , Syndrome cardiorénal/étiologie , Études de cohortes , Études rétrospectives , Accident vasculaire cérébral/complications , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/épidémiologie , Défaillance cardiaque/complications , Défaillance cardiaque/épidémiologie , Défaillance rénale chronique/épidémiologie , Défaillance rénale chronique/complications , Infarctus du myocarde/complications
13.
JACC Heart Fail ; 10(3): 175-183, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35241245

RÉSUMÉ

Renal dysfunction is one of the strongest predictors of outcome in heart failure. Several studies have revealed that both reduced perfusion and increased congestion (and central venous pressure) contribute to worsening renal function in heart failure. This paper proposes a novel factor in the link between cardiac and renal dysfunction: "renal tamponade" or compression of renal structures caused by the limited space for expansion. This space can be limited either by the rigid renal capsule that encloses the renal interstitial tissue or by the layer of fat around the kidneys or by the peritoneal space exerting pressure on the retroperitoneal kidneys. Renal decapsulation in animal models of heart failure and acute renal ischemia has been shown effective in alleviating pressure-related injury within the kidney itself, thus supporting this concept and making it a potentially interesting novel treatment in heart failure.


Sujet(s)
Syndrome cardiorénal , Défaillance cardiaque , Animaux , Syndrome cardiorénal/étiologie , Femelle , Coeur , Humains , Rein/physiologie , Mâle , Pression
14.
Am J Cardiol ; 168: 99-104, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35045927

RÉSUMÉ

This study aimed to investigate the role of secondary mitral regurgitation (MR) and tricuspid regurgitation (TR) in the pathogenesis of cardiorenal syndrome (CRS). Worsening renal function in patients with acute decompensated heart failure receiving diuretic therapy is defined as CRS and is related to central venous congestion. The role of secondary MR and TR is not well studied. We retrospectively reviewed the electronic medical records of 80 consecutive patients hospitalized with acute decompensated heart failure. Patients were divided into 2 groups: group 1 (CRS) if creatinine increased >0.3 mg/dl from baseline and group 2 (no CRS) if creatinine remained stable or improved with diuretic therapy. Admission creatinine was higher in group 1 compared with group 2 (1.5 vs 1.2 mg/dl, p = 0.033). The magnitude of MR and TR were higher by both visual assessment (moderate to severe [3+] or severe [4+] MR in 68% of patients in group 1 vs 3% in group 2, p <0.0001; 3+ or 4+ TR in 48% of patients in group 1 vs 10% in group 2, p = 0.0004) and by vena contracta (MR 0.6 ± 0.2 cm in group 1 vs 0.4 ± 0.1 cm in group 2, p <0.0001; TR 0.5 ± 0.2 cm in group 1 vs 0.4 ± 0.2 cm in group 2, p = 0.0013). By using receiver operating characteristic curves, MR and TR were the most sensitive parameters in predicting CRS. In conclusion, renal function on admission and moderate to severe or severe MR and TR are highly predictive of the risk of developing CRS.


Sujet(s)
Syndrome cardiorénal , Défaillance cardiaque , Insuffisance mitrale , Insuffisance tricuspide , Syndrome cardiorénal/complications , Syndrome cardiorénal/étiologie , Créatinine , Diurétiques/usage thérapeutique , Femelle , Défaillance cardiaque/complications , Défaillance cardiaque/épidémiologie , Humains , Mâle , Insuffisance mitrale/complications , Insuffisance mitrale/épidémiologie , Études rétrospectives , Résultat thérapeutique , Insuffisance tricuspide/complications
15.
Kidney Int ; 101(2): 214-216, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35065687

RÉSUMÉ

Chronic kidney disease (CKD) and cardiovascular disease frequently run in parallel. Herein, Soppert et al. provide an interesting meta-analysis of the effects of CKD on cardiac remodeling and/or function in mice based on the model, strain, and duration. The authors sought to determine the most appropriate experimental model to unravel the specific underlying pathologic mechanisms involved in cardiac damage in CKD (single hit) or to investigate new strategies to prevent CKD-induced cardiovascular disease (multifactorial hits representing cardiovascular comorbidities of patients with CKD).


Sujet(s)
Syndrome cardiorénal , Cardiomyopathies , Insuffisance rénale chronique , Animaux , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/anatomopathologie , Cardiomyopathies/étiologie , Modèles animaux de maladie humaine , Coeur/physiopathologie , Humains , Souris , Insuffisance rénale chronique/anatomopathologie
16.
Clin Appl Thromb Hemost ; 28: 10760296211072820, 2022.
Article de Anglais | MEDLINE | ID: mdl-35018865

RÉSUMÉ

BACKGROUND: Patients with end-stage renal disease (ESRD) often present with an increased risk of cardiovascular disease. Conditions of compromised cardiovascular health such as atrial fibrillation (AFIB) and peripheral arterial disease (PAD) may alter biomarker levels in a way that reflects worsening ESRD. This study profiled biomarkers and laboratory parameters of endothelium dysfunction in patients with ESRD, categorized by additional AFIB and PAD conditions. METHODS: Citrated blood samples were collected from 95 patients with ESRD. Biomarker levels were measured from plasma samples using sandwich ELISAs, including tissue plasminogen activator (tPA), D-dimer, and nitrotyrosine. Lab parameters, including BUN, calcium, creatinine, parathyroid hormone, phosphate, alkaline phosphatase, ferritin, transferrin, and total iron capacity, and patient comorbidities were obtained from patient medical records. The comorbidities were determined through provider notes, and evidence of applicable testing. RESULTS: 14.89% of patients were found to have atrial fibrillation (n = 14), 30.85% of patients were found to have peripheral arterial disease (n = 29), and 6.38% of patients were found to have both peripheral arterial disease and atrial fibrillation (n = 6). When compared to patients with only ESRD, patients with ESRD and PAD showed elevated levels of D-Dimer (p = .0314) and nitrotyrosine (p = .0330). When compared to patients with only ESRD, patients with atrial fibrillation showed elevated levels of D-Dimer (p = .0372), nitrotyrosine (p = .0322), and tPA (p = .0198). CONCLUSION: When compared to patients with just ESRD, patients with concomitant PAD had elevated levels of Nitrotyrosine and D-dimer; while patients with concomitant Afib had elevated levels of nitrotyrosine, D-dimer, as well as tPA.


Sujet(s)
Syndrome cardiorénal/étiologie , Produits de dégradation de la fibrine et du fibrinogène/métabolisme , Défaillance rénale chronique/complications , Sujet âgé , Marqueurs biologiques/sang , Syndrome cardiorénal/sang , Syndrome cardiorénal/épidémiologie , Femelle , Humains , Incidence , Défaillance rénale chronique/épidémiologie , Défaillance rénale chronique/thérapie , Mâle , Adulte d'âge moyen , Dialyse rénale/méthodes , Facteurs de risque , États-Unis/épidémiologie
17.
Biomolecules ; 11(9)2021 09 16.
Article de Anglais | MEDLINE | ID: mdl-34572583

RÉSUMÉ

Acute renal failure (ARF) is a clinical critical syndrome with rapid and severe decline of renal function. Complications of ARF, especially its cardiac complications (cardiorenal syndrome type 3, CRS-3), are the main causes of death in patients with ARF. However, the shortage and limited efficacy of therapeutic drugs make it significant to establish new large-scale drug screening models. Based on the Nitroreductase/Metronidazole (NTR/MTZ) cell ablation system, we constructed a Tg(cdh17:Dendra2-NTR) transgenic zebrafish line, which can specifically ablate renal tubular epithelial cells. The absence of renal tubular epithelial cells can lead to ARF in zebrafish larvae. The ARF symptoms, such as heart enlargement, slow heart rate and blood stasis, are similar to the clinical manifestations of human CRS-3. Furthermore, two therapeutic drugs (digoxin and enalapril) commonly used in the clinical treatment of heart failure were also effective in alleviating the symptoms of CRS-3 in zebrafish, which proved the effectiveness of this model. Drug screening further discovered a potential drug candidate, α-lipoic acid, which can effectively alleviate the symptoms of CRS-3 through its antioxidant function. Accordingly, we established a new ARF model of zebrafish, which laid a foundation for large-scale screening of new therapeutic drugs for its complications.


Sujet(s)
Atteinte rénale aigüe/complications , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/étiologie , Évaluation préclinique de médicament , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/physiopathologie , Animaux , Animal génétiquement modifié , Syndrome cardiorénal/traitement médicamenteux , Syndrome cardiorénal/étiologie , Maladies cardiovasculaires/anatomopathologie , Digoxine/pharmacologie , Digoxine/usage thérapeutique , Modèles animaux de maladie humaine , Énalapril/pharmacologie , Énalapril/usage thérapeutique , Cellules épithéliales/anatomopathologie , Humains , Tubules rénaux/anatomopathologie , Tubules rénaux/physiopathologie , Larve/physiologie , Métronidazole , Débit sanguin régional/effets des médicaments et des substances chimiques , Acide lipoïque/pharmacologie , Acide lipoïque/usage thérapeutique , Résultat thérapeutique , Danio zébré
18.
Int Heart J ; 62(5): 1052-1056, 2021 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-34544987

RÉSUMÉ

Cardio-renal syndrome is a challenging clinical entity to manage, and is often associated with increased morbidity and mortality. We hypothesized that adaptive servo-ventilation (ASV), non-invasive positive pressure ventilation that ameliorates systemic/pulmonary congestion, may improve renal function in patients with symptomatic heart failure complicated by the cardio-renal syndrome. Patients with symptomatic congestive heart failure who underwent ASV therapy for over 1 month were included in this retrospective study. The trajectory of the estimated glomerular filtration ratio (eGFR) between the pre-1 month period and the post-one-month period (on ASV) were compared. A total of 81 patients (median 65 years old, 65 men) were included. eGFR decreased during the pre-1 month period from 52.7 (41.7, 64.6) down to 49.9 (37.3, 63.5) mL/minute/1.73 m2 (P < 0.001) whereas we observed an increase following one-month of ASV therapy up to 53.4 (38.6, 68.6) mL/minute/1.73 m2 (P = 0.022). A reduction in furosemide equivalent dose following the initiation of ASV therapy was independently associated with increases in eGFR with an adjusted odds ratio of 13.72 (95% confidence interval 3.40-55.3, P < 0.001). In conclusion, short-term ASV therapy was associated with the preservation of renal function, particularly when the dose of loop diuretics was concomitantly reduced.


Sujet(s)
Syndrome cardiorénal/thérapie , Défaillance cardiaque/thérapie , Rein/physiopathologie , Ventilation non effractive/instrumentation , Ventilation à pression positive/méthodes , Sujet âgé , Syndrome cardiorénal/épidémiologie , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/mortalité , Diurétiques/administration et posologie , Diurétiques/usage thérapeutique , Diminution progressive de la dose du médicament/statistiques et données numériques , Femelle , Furosémide/administration et posologie , Furosémide/usage thérapeutique , Débit de filtration glomérulaire/physiologie , Défaillance cardiaque/complications , Défaillance cardiaque/physiopathologie , Hémodynamique/effets des médicaments et des substances chimiques , Hémodynamique/physiologie , Humains , Tests de la fonction rénale , Mâle , Adulte d'âge moyen , Études rétrospectives
19.
Contrib Nephrol ; 199: 71-79, 2021.
Article de Anglais | MEDLINE | ID: mdl-34428764

RÉSUMÉ

Clinical Background: The heart can cause kidney disease, and the kidney can cause heart disease. As an example of the first situation, we can mention dilated cardiomyopathies, which can lead to renal failure of the pre-renal type due to the state of renal hypoflow. As an example of the second situation, we can remember that renal failure is a risk factor for cardiovascular diseases, such as coronary heart disease, due to the acceleration in the process of atherosclerosis that it promotes. Epidemiology: In this chapter, we will address what we consider to be the two main aspects of the interrelationships between heart and kidney disease that are "cardiorenal syndrome (CRS)" and "chronic kidney disease (CKD) and coronary heart disease (CHD)." Challenges: For CRS, we discuss its epidemiology, types, pathophysiological mechanisms common to CRS types 1, 2, 3 and 4 and pathogenesis of CSR type 5. Treatment: For "CKD and CHD" we discuss the association of CKD and CHD in community-based populations, traditional risk factor in CKD, non-traditional risk factor in CKD, reduced risk of CHD in patients with CKD, statin treatment, hypertension treatment, anti-platelet aggregation therapy, treatment of CHD in patients with CKD and prognosis of CHDF in CKD patients.


Sujet(s)
Athérosclérose , Syndrome cardiorénal , Insuffisance rénale chronique , Syndrome cardiorénal/complications , Syndrome cardiorénal/étiologie , Humains , Rein , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/épidémiologie , Facteurs de risque
20.
Sci Rep ; 11(1): 15036, 2021 07 22.
Article de Anglais | MEDLINE | ID: mdl-34294750

RÉSUMÉ

Renal injury caused by renal ischemia and reperfusion strongly influences heart morphology, electrophysiology, and redox unbalance. The so-called cardiorenal syndrome is an important class of dysfunction since heart and kidneys are responsible for hemodynamic stability and organ perfusion through a complex network. In the present work we investigate the vibrational spectral features probed by Fourier-Transform Raman (FT-Raman) spectroscopy due to physiological alterations induced by renal ischemic reperfusion aiming to detect molecular markers related to progression of acute to chronic kidney injury and mortality predictors as well. C57BL/6J mice were subjected to unilateral occlusion of the renal pedicle for 60 min and reperfusion for 5, 8, and 15 days. Biopsies of heart and kidney tissues were analyzed. Our findings indicated that cysteine/cystine, fatty acids, methyl groups of Collagen, α-form of proteins, Tyrosine, and Tryptophan were modulated during renal ischemia and reperfusion process. These changes are consistent with fibroblast growth factors and Collagen III contents changes. Interestingly, Tyrosine and Tryptophan, precursor molecules for the formation of uremic toxins such as indoxyl sulfate and p-cresyl sulfate were also modulated. They are markers of kidney injury and their increase is strongly correlated to cardiovascular mortality. Regarding this aspect, we notice that monitoring the Tyrosine and Tryptophan bands at 1558, 1616, and 1625 cm-1 is a viable and and advantageous way to predict fatality in cardiovascular diseases both "in vivo" or "in vitro", using the real-time, multiplexing, and minimally invasive advantages of FT-Raman spectroscopy.


Sujet(s)
Marqueurs biologiques , Maladies du rein/étiologie , Maladies du rein/métabolisme , Tryptophane/métabolisme , Tyrosine/métabolisme , Animaux , Syndrome cardiorénal/diagnostic , Syndrome cardiorénal/étiologie , Syndrome cardiorénal/métabolisme , Prise en charge de la maladie , Prédisposition aux maladies , Maladies du rein/diagnostic , Mâle , Souris , Spécificité d'organe , Lésion d'ischémie-reperfusion/complications , Analyse spectrale/méthodes , Tryptophane/analyse , Tyrosine/analyse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE