Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Free Radic Biol Med ; 212: 49-64, 2024 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-38141891

RESUMEN

Releasing unilateral ureteral obstruction (RUUO) is the gold standard for decreasing renal damage induced during unilateral ureteral obstruction (UUO); however, the complete recovery after RUUO depends on factors such as the time and severity of obstruction and kidney contralateral compensatory mechanisms. Interestingly, previous studies have shown that kidney damage markers such as oxidative stress, inflammation, and apoptosis are present and even increase after removal obstruction. To date, previous therapeutic strategies have been used to potentiate the recovery of renal function after RUUO; however, the mechanisms involving renal damage reduction are poorly described and sometimes focus on the recovery of renal functionality. Furthermore, using natural antioxidants has not been completely studied in the RUUO model. In this study, we selected sulforaphane (SFN) because it activates the nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that induces an antioxidant response, decreasing oxidative stress and inflammation, preventing apoptosis. Thus, we pre-administrated SFN on the second day after UUO until day five, where we released the obstruction on the three days after UUO. Then, we assessed oxidative stress, inflammation, and apoptosis markers. Interestingly, we found that SFN administration in the RUUO model activated Nrf2, inducing its translocation to the nucleus to activate its target proteins. Thus, the Nrf2 activation upregulated glutathione (GSH) content and the antioxidant enzymes catalase, glutathione peroxidase (GPx), and glutathione reductase (GR), which reduced the oxidative stress markers. Moreover, the improvement of antioxidant response by SFN restored S-glutathionylation in the mitochondrial fraction. Activated Nrf2 also reduced inflammation by lessening the nucleotide-binding domain-like receptor family pyrin domain containing 3 and interleukin 1ß (IL-1ß) production. Reducing oxidative stress and inflammation prevented apoptosis by avoiding caspase 3 cleavage and increasing B-cell lymphoma 2 (Bcl2) levels. Taken together, the obtained results in our study showed that the upregulation of Nrf2 by SFN decreases oxidative stress, preventing inflammation and apoptosis cell death during the release of UUO.


Asunto(s)
Antioxidantes , Sulfóxidos , Obstrucción Ureteral , Humanos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Riñón/metabolismo , Isotiocianatos/farmacología , Inflamación/metabolismo , Apoptosis , Antiinflamatorios/farmacología
2.
Int J Mol Sci ; 24(21)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37958859

RESUMEN

Cardiorenal syndrome type 4 (CRS type 4) occurs when chronic kidney disease (CKD) leads to cardiovascular damage, resulting in high morbidity and mortality rates. Mitochondria, vital organelles responsible for essential cellular functions, can become dysfunctional in CKD. This dysfunction can trigger inflammatory responses in distant organs by releasing Damage-associated molecular patterns (DAMPs). These DAMPs are recognized by immune receptors within cells, including Toll-like receptors (TLR) like TLR2, TLR4, and TLR9, the nucleotide-binding domain, leucine-rich-containing family pyrin domain-containing-3 (NLRP3) inflammasome, and the cyclic guanosine monophosphate (cGMP)-adenosine monophosphate (AMP) synthase (cGAS)-stimulator of interferon genes (cGAS-STING) pathway. Activation of these immune receptors leads to the increased expression of cytokines and chemokines. Excessive chemokine stimulation results in the recruitment of inflammatory cells into tissues, causing chronic damage. Experimental studies have demonstrated that chemokines are upregulated in the heart during CKD, contributing to CRS type 4. Conversely, chemokine inhibitors have been shown to reduce chronic inflammation and prevent cardiorenal impairment. However, the molecular connection between mitochondrial DAMPs and inflammatory pathways responsible for chemokine overactivation in CRS type 4 has not been explored. In this review, we delve into mechanistic insights and discuss how various mitochondrial DAMPs released by the kidney during CKD can activate TLRs, NLRP3, and cGAS-STING immune pathways in the heart. This activation leads to the upregulation of chemokines, ultimately culminating in the establishment of CRS type 4. Furthermore, we propose using chemokine inhibitors as potential strategies for preventing CRS type 4.


Asunto(s)
Síndrome Cardiorrenal , Insuficiencia Renal Crónica , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal , Mitocondrias/metabolismo , Nucleotidiltransferasas/metabolismo , Receptores Inmunológicos/metabolismo , Alarminas/metabolismo , Quimiocinas/metabolismo , Insuficiencia Renal Crónica/metabolismo
3.
Life Sci ; 332: 122098, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37734433

RESUMEN

Otto Warburg hypothesized that some cancer cells reprogram their metabolism, favoring glucose metabolism by anaerobic glycolysis (Warburg effect) instead of oxidative phosphorylation, mainly because the mitochondria of these cells were damaged or dysfunctional. It should be noted that mitochondrial apoptosis is decreased because of the dysfunctional mitochondria. Strategies like mitochondrial transplantation therapy, where functional mitochondria are transplanted to cancer cells, could increase cell death, such as apoptosis, because the intrinsic apoptosis mechanisms would be reactivated. In addition, mitochondrial transplantation is associated with the redox state, which could promote synergy with common anticancer treatments such as ionizing radiation, chemotherapy, or radiotherapy, increasing cell death due to the presence or decrease of oxidative stress. On the other hand, mitochondrial transfer, a natural process for sharing mitochondrial between cells, induces an increase in chemoresistance and invasiveness in cancer cells that receive mitochondria from cells of the tumor microenvironment (TME), which indicates an antitumor therapeutic target. This review focuses on understanding mitochondrial transplantation as a therapeutic outcome induced by a procedure in aspects including oxidative stress, metabolism shifting, mitochondrial function, auto-/mitophagy, invasiveness, and chemoresistance. It also explores how these mechanisms, such as apoptosis, necroptosis, and parthanatos, impact cell death pathways. Finally, it discusses the chemoresistance and invasiveness in cancer cells associated with mitochondria transfer, indicating an antitumor therapeutic target.


Asunto(s)
Mitocondrias , Neoplasias , Humanos , Mitocondrias/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Glucólisis , Estrés Oxidativo , Apoptosis , Autofagia , Microambiente Tumoral
4.
Methods Mol Biol ; 2664: 283-308, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37423995

RESUMEN

Proper kidney function depends highly on mitochondria homeostasis. This organelle is the primary source of ATP production in the kidney and regulates other cellular processes such as redox and calcium homeostasis. Although the mitochondria's primary recognized function is cellular energy production, through the function of the Krebs cycle, electron transport system (ETS), as well as oxygen and electrochemical gradient consumption, this function is interconnected with multiple signaling and metabolic pathways, making bioenergetics a central hub in renal metabolism. Furthermore, mitochondrial biogenesis, dynamics, and mass are also strongly related to bioenergetics. This central role is not surprising given that mitochondrial impairment, including functional and structural alterations, has been recently reported in several kidney diseases. Here, we describe assessment of mitochondrial mass, structure, and bioenergetics in kidney tissue and renal-derived cell lines. These methods allow investigation of mitochondrial alterations in kidney tissue and renal cells under different experimental conditions.


Asunto(s)
Metabolismo Energético , Mitocondrias , Mitocondrias/metabolismo , Riñón/metabolismo , Técnicas Histológicas , Microscopía Electrónica de Transmisión
5.
Foods ; 12(5)2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36900438

RESUMEN

Chronic kidney disease (CKD) is a health problem that is constantly growing. This disease presents a diverse symptomatology that implies complex therapeutic management. One of its characteristic symptoms is dyslipidemia, which becomes a risk factor for developing cardiovascular diseases and increases the mortality of CKD patients. Various drugs, particularly those used for dyslipidemia, consumed in the course of CKD lead to side effects that delay the patient's recovery. Therefore, it is necessary to implement new therapies with natural compounds, such as curcuminoids (derived from the Curcuma longa plant), which can cushion the damage caused by the excessive use of medications. This manuscript aims to review the current evidence on the use of curcuminoids on dyslipidemia in CKD and CKD-induced cardiovascular disease (CVD). We first described oxidative stress, inflammation, fibrosis, and metabolic reprogramming as factors that induce dyslipidemia in CKD and their association with CVD development. We proposed the potential use of curcuminoids in CKD and their utilization in clinics to treat CKD-dyslipidemia.

6.
Antioxidants (Basel) ; 11(10)2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36290577

RESUMEN

Unilateral ureteral obstruction (UUO) is an animal rodent model that allows the study of obstructive nephropathy in an accelerated manner. During UUO, tubular damage is induced, and alterations such as oxidative stress, inflammation, lipid metabolism, and mitochondrial impairment favor fibrosis development, leading to chronic kidney disease progression. Sulforaphane (SFN), an isothiocyanate derived from green cruciferous vegetables, might improve mitochondrial functions and lipid metabolism; however, its role in UUO has been poorly explored. Therefore, we aimed to determine the protective effect of SFN related to mitochondria and lipid metabolism in UUO. Our results showed that in UUO SFN decreased renal damage, attributed to increased mitochondrial biogenesis. We showed that SFN augmented peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) and nuclear respiratory factor 1 (NRF1). The increase in biogenesis augmented the mitochondrial mass marker voltage-dependent anion channel (VDAC) and improved mitochondrial structure, as well as complex III (CIII), aconitase 2 (ACO2) and citrate synthase activities in UUO. In addition, lipid metabolism was improved, observed by the downregulation of cluster of differentiation 36 (CD36), sterol regulatory-element binding protein 1 (SREBP1), fatty acid synthase (FASN), and diacylglycerol O-acyltransferase 1 (DGAT1), which reduces triglyceride (TG) accumulation. Finally, restoring the mitochondrial structure reduced excessive fission by decreasing the fission protein dynamin-related protein-1 (DRP1). Autophagy flux was further restored by reducing beclin and sequestosome (p62) and increasing B-cell lymphoma 2 (Bcl2) and the ratio of microtubule-associated proteins 1A/1B light chain 3 II and I (LC3II/LC3I). These results reveal that SFN confers protection against UUO-induced kidney injury by targeting mitochondrial biogenesis, which also improves lipid metabolism.

7.
Chem Biol Interact ; 361: 109961, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35500868

RESUMEN

Cadmium (Cd) is one of the most toxic metals without biological function, and its accumulation in living organisms has been reported. The kidney is a target organ in Cd toxicity; it has been observed that Cd causes kidney damage even at low concentrations, and Cd damage can quickly progress to chronic kidney disease. The mitochondria play a fundamental role in the nephrotoxicity of Cd; Cd enters the mitochondria and affects the electron transport system (ETS), increases the production of reactive oxygen species (ROS), decreases the mitochondrial membrane potential (Δψm), alters mitochondrial dynamics, induces mutations in mitochondrial deoxyribonucleic acid (mtDNA) and decreased biogenesis leading to increased mitophagy, autophagy, and inevitably apoptosis. Existing therapies to treat Cd nephrotoxicity are currently based on antioxidant and chelating compounds, but despite their promising effects, they have some limitations; therefore, Cd nephrotoxicity continues to represent a global health problem. Mitochondrial transplantation is a new experimental approach with positive results by reversing mitochondrial alterations in cardiac and kidney dysfunction mainly caused by oxidative stress. Hence, the current review discusses the role of mitochondria in Cd-induced toxicity in the kidney and proposes mitochondrial transference as a novel therapy based on transplanting healthy mitochondria to cells with Cd-compromised mitochondria. This review is the first to propose mitochondrial transplantation as a treatment for heavy metal-induced kidney damage.


Asunto(s)
Cadmio , Enfermedades Renales , Apoptosis , Cadmio/toxicidad , Humanos , Riñón , Enfermedades Renales/inducido químicamente , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
8.
FASEB J ; 35(8): e21761, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34245616

RESUMEN

Uremic cardiomyopathy is a common complication in chronic kidney disease (CKD) patients, accounting for a high mortality rate. Several mechanisms have been proposed to link CKD and cardiac alterations; however, the early cardiac modifications that occur in CKD that may trigger cardiac remodeling and dysfunction remain largely unexplored. Here, in a mouse model of CKD induced by 5/6 nephrectomy, we first analyzed the early transcriptional and inflammatory changes that occur in the heart. Five days after 5/6 nephrectomy, RNA-sequencing showed the upregulation of 54 genes in the cardiac tissue of CKD mice and the enrichment of biological processes related to immune system processes. Increased cardiac infiltration of T-CD4+ lymphocytes, myeloid cells, and macrophages during early CKD was observed. Next, since CC chemokine ligand-8 (CCL8) was one of the most upregulated genes in the heart of mice with early CKD, we investigated the effect of acute and transient CCL8 inhibition on uremic cardiomyopathy severity. An increase in CCL8 protein levels was confirmed in the heart of early CKD mice. CCL8 inhibition attenuated the early infiltration of T-CD4+ lymphocytes and macrophages to the cardiac tissue, leading to a protection against chronic cardiac fibrotic remodeling, inflammation and cardiac dysfunction induced by CKD. Altogether, our data show the occurrence of transcriptional and inflammatory changes in the heart during the early phases of CKD and identify CCL8 as a key contributor to the early cardiac inflammatory state that triggers further cardiac remodeling and dysfunction in uremic cardiomyopathy.


Asunto(s)
Cardiomiopatías/metabolismo , Quimiocina CCL8/biosíntesis , Miocardio/metabolismo , Insuficiencia Renal Crónica/metabolismo , Regulación hacia Arriba , Uremia/metabolismo , Animales , Cardiomiopatías/patología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Miocardio/patología , Insuficiencia Renal Crónica/patología , Uremia/patología
9.
Int J Mol Sci ; 20(13)2019 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-31252520

RESUMEN

The epithelial sodium channel (ENaC) has a key role in modulating endothelial cell stiffness and this in turn regulates nitric oxide (NO) synthesis. The physiological relevance of endothelial ENaC in pathological conditions where reduced NO bioavailability plays an essential role remains largely unexplored. Renal ischemia/reperfusion (IR) injury is characterized by vasoconstriction and sustained decrease in renal perfusion that is partially explained by a reduction in NO bioavailability. Therefore, we aimed to explore if an endothelial ENaC deficiency has an impact on the severity of renal injury induced by IR. Male mice with a specific endothelial sodium channel α (αENaC) subunit gene inactivation in the endothelium (endo-αENaCKO) and control littermates were subjected to bilateral renal ischemia of 22 min and were studied after 24 h of reperfusion. In control littermates, renal ischemia induced an increase in plasma creatinine and urea, augmented the kidney injury molecule-1 (Kim-1) and neutrophil gelatinase associated lipocalin-2 (NGAL) mRNA levels, and produced severe tubular injury. The absence of endothelial αENaC expression prevented renal tubular injury and renal dysfunction. Moreover, endo-αENaCKO mice recovered faster from renal hypoxia after the ischemia episode as compared to littermates. In human endothelial cells, pharmacological ENaC inhibition promoted endothelial nitric oxide synthase (eNOS) coupling and activation. Altogether, these data suggest an important role for endothelial αENaC in kidney IR injury through improving eNOS activation and kidney perfusion, thus, preventing ischemic injury.


Asunto(s)
Canales Epiteliales de Sodio/genética , Daño por Reperfusión/metabolismo , Animales , Células Cultivadas , Canales Epiteliales de Sodio/deficiencia , Receptor Celular 1 del Virus de la Hepatitis A/genética , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Lipocalina 2/genética , Lipocalina 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Daño por Reperfusión/genética
10.
Am J Physiol Renal Physiol ; 317(2): F275-F285, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31116605

RESUMEN

Major cardiovascular events are a common complication in patients with chronic kidney disease (CKD). Endothelial dysfunction can contribute to the cardiovascular injury observed in CKD. Here, we used a rat model of acute kidney injury to CKD transition to investigate heart alterations in the pathway activating endothelial nitric oxide synthase (eNOS) and its impact on the cardiac injury observed during CKD progression. Fifty male Wistar rats were subjected to sham surgery (n = 25) or bilateral renal ischemia-reperfusion (IR-CKD) for 45 min (n = 25). Rats were studied on a monthly basis up to 5 mo (n = 5). In another set of sham and IR-CKD rats, l-arginine was administered starting on the third month after renal ischemia. CKD development and cardiac alterations were monitored in all groups. CKD was characterized by a progressive increase in proteinuria and renal dysfunction that was evident after the fifth month of followup. Heart hypertrophy was observed starting on the fourth month after ischemia-reperfusion. There was a significant increase in brain natriuretic peptide levels. In the heart, IR-CKD rats had increased eNOS phosphorylation at threonine 495 and reduced eNOS-heat shock protein-90α interactions. l-Arginine administration prevented the heart alterations observed during CKD and increased eNOS coupling/dimerization and activation. In summary, CKD progression is accompanied by cardiac hypertrophy, fibrosis, oxidative stress, and increased brain natriuretic peptide levels. These alterations were associated with limited eNOS activation in the heart, which may result in reduced nitric oxide bioavailability and contribute to cardiac injury during CKD.


Asunto(s)
Lesión Renal Aguda/complicaciones , Cardiomegalia/etiología , Miocardio/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Insuficiencia Renal Crónica/etiología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/enzimología , Animales , Arginina/farmacología , Cardiomegalia/enzimología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Activación Enzimática , Fibrosis , Proteínas HSP90 de Choque Térmico/metabolismo , Masculino , Péptido Natriurético Encefálico/metabolismo , Estrés Oxidativo , Fosforilación , Ratas Wistar , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/prevención & control , Treonina , Factores de Tiempo
11.
Nephrol Dial Transplant ; 34(5): 794-801, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107561

RESUMEN

BACKGROUND: Acute kidney injury (AKI) is not as harmless as previously thought since it may lead to chronic kidney disease (CKD). Because most of the time ischemic AKI occurs unexpectedly, it is difficult to prevent its occurrence and there are no specific therapeutic approaches to prevent the AKI to CKD transition. We aimed to determine whether mineralocorticoid receptor blockade (MRB) in the first days after ischemia/reperfusion (IR) can prevent progression to CKD. METHODS: Four groups of male Wistar rats were included: sham and three groups of bilateral renal ischemia for 45 min, one without treatment and the other two receiving spironolactone for 5 or 10 days, starting 24 h after IR. The rats were studied at 10 days or 5 months after ischemia induction. RESULTS: After 5 months of follow-up, the untreated group exhibited clear evidence of AKI to CKD progression, such as proteinuria, reduced renal blood flow, tubulointerstitial fibrosis, glomerulosclerosis and glomerular hypertrophy. All these alterations were prevented by both spironolactone treatments initiated 24 h after IR, the 10-day treatment being more effective. Within the early mechanisms of the MRB protective effect are the reduction of inflammation and increased endothelin-B-receptor expression and endothelial nitric oxide synthase activation in the first 10 days after IR. CONCLUSIONS: We propose that MRB, administered 24 h after the ischemic injury that leads to AKI, reduces inflammation and promotes efficient tissue repair that avoids the AKI to CKD transition. These data highlight a therapeutic window to preclude CKD development after AKI.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Inflamación/metabolismo , Riñón/patología , Receptor de Endotelina B/metabolismo , Insuficiencia Renal Crónica/prevención & control , Espironolactona/administración & dosificación , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inflamación/patología , Riñón/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...