Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Physiol ; 238(4): 813-828, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36879552

RESUMEN

Chronic kidney disease (CKD) is a disease with decreased, irreversible renal function. Pruritus is the most common skin symptom in patients with CKD, especially in end-stage renal disease. The underlying molecular and neural mechanism of CKD-associated pruritus (CKD-aP) remains obscure. Our data show that the level of allantoin increases in the serum of CKD-aP and CKD model mice. Allantoin could induce scratching behavior in mice and active DRG neurons. The calcium influx and action potential reduced significantly in DRG neurons of MrgprD KO or TRPV1 KO mice. U73122, an antagonist of phospholipase C, could also block calcium influx in DRG neurons induced by allantoin. Thus, our results concluded that allantoin plays an important role in CKD-aP, mediated by MrgprD and TrpV1, in CKD patients.


Asunto(s)
Alantoína , Prurito , Insuficiencia Renal Crónica , Animales , Ratones , Alantoína/efectos adversos , Calcio , Prurito/inducido químicamente , Prurito/diagnóstico , Receptores Acoplados a Proteínas G , Insuficiencia Renal Crónica/complicaciones
2.
Cell Mol Biol Lett ; 25: 33, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32514269

RESUMEN

BACKGROUND: Peritoneal fibrosis (PF) is a frequent complication caused by peritoneal dialysis (PD). Peritoneal mesothelial cells (PMCs), the first barrier of the peritoneum, play an important role in maintaining structure and function in the peritoneum during PD. Mesothelial-mesenchymal transition (MMT) and oxidative stress of PMCs are two key processes of PF. PURPOSE: To elucidate the efficacy and possible mechanism of asiaticoside inhibition of MMT and ROS generation in TGF-ß1-induced PF in human peritoneal mesothelial cells (HPMCs). METHODS: MMT and ROS generation of HPMCs were induced by TGF-ß1. To explain the anti-MMT and antioxidant role of asiaticoside, varied doses of asiaticoside, oxygen radical scavenger (NAC), TGF-ß receptor kinase inhibitor (LY2109761) and Nrf2 inhibitor (ML385) were used separately. Immunoblots were used to detect the expression of signaling associated proteins. DCFH-DA was used to detect the generation of ROS. Transwell migration assay and wound healing assay were used to verify the capacity of asiaticoside to inhibit MMT. Immunofluorescence assay was performed to observe the subcellular translocation of Nrf2 and expression of HO-1. RESULTS: Asiaticoside inhibited TGF-ß1-induced MMT and suppressed Smad signaling in a dose-dependent manner. Migration and invasion activities of HPMCs were decreased by asiaticoside. Asiaticoside decreased TGF-ß1-induced ROS, especially in a high dose (150 µM) for 6 h. Furthermore, ML385 partly abolished the inhibitory effect of asiaticoside on MMT, ROS and p-Smad2/3. CONCLUSIONS: Asiaticoside inhibited the TGF-ß1-induced MMT and ROS via Nrf2 activation, thus protecting the peritoneal membrane and preventing PF.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Triterpenos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Células Epiteliales/efectos de los fármacos , Humanos , Estrés Oxidativo/efectos de los fármacos , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
3.
J Cell Biochem ; 120(4): 5505-5509, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30474893

RESUMEN

OBJECTIVE: This study aimed to investigate the efficacy of Chinese herb Cistanche Yishen granules (CYG) in the treatment of tinnitus for patients with chronic nephritis. METHODS: A total of 89 adult patients were diagnosed with chronic glomerulonephritis from January 2016 to December 2017. All the patients were randomly divided into two groups, such as the control group and the CYG group. The efficacy of tinnitus was determined using tinnitus handicap inventory (THI), Pittsburgh sleep quality index (PSQI), pure tone audiometry (PTA), speech reception threshold (SRT), and visual analog scale (VAS) for tinnitus loudness and annoyance. RESULTS: In both these two groups of patients, values of THI, PSQI, PTA, SRT, and VAS for tinnitus loudness and annoyance were significantly decreased after the treatment compared with those before treatment. However, all values in CYG group after the treatment were significantly lower than those in the control group. CONCLUSION: CYG could apparently release the tinnitus symptoms in the patients with chronic nephritis. This study might give more clinical evidence for Cistanche in the treatment of tinnitus and give a new treatment method for the patients with tinnitus.


Asunto(s)
Cistanche , Medicamentos Herbarios Chinos/administración & dosificación , Nefritis , Acúfeno , Adulto , Enfermedad Crónica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nefritis/complicaciones , Nefritis/tratamiento farmacológico , Estudios Prospectivos , Acúfeno/tratamiento farmacológico , Acúfeno/etiología
4.
Cell Physiol Biochem ; 51(6): 2794-2813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30562743

RESUMEN

BACKGROUND/AIMS: The epithelial-to-mesenchymal transition (EMT) of peritoneal mesothelial cells (PMCs) is a crucial event in the induction of peritoneal fibrosis (PF), in which canonical Wnt/ß-catenin signaling participates. Smads signaling is reported to interact with ß-catenin and synergistically regulates EMT. This study was aimed to reveal the effect of Astragalus on ß-catenin in EMT of PMCs. METHODS: To obtain the role of ß-catenin in EMT, gene transfer into HMrSV5 cell line and rats has been achieved. After Astragalus treatment, EMT markers and signaling pathway-related indicators were detected by western blotting, immunofluorescence, immunohistochemistry, immunoprecipitation and real time-PCR. RESULTS: ß-catenin knockdown suppressed EMT of HMrSV5 cells. Astragalus alleviated EMT of PMCs characterized by increased E-cadherin and decreased α-SMA and Vimentin. In rat model of peritoneal dialysis (PD), Astragalus attenuated peritoneal thickening and fibrosis. Astragalus down-regulated ß-catenin by stabilizing the Glycogen synthase kinase-3ß (GSK-3ß)/ß-catenin complex and further inhibited the nuclear translocation of ß-catenin. Meanwhile, Astragalus down-regulated ß-catenin by enhancing Smad7 expression. Silencing Smad7 antagonized the EMT-inhibitory effect of Astragalus. CONCLUSION: Astragalus inhibits EMT of PMCs by down-regulating ß-catenin. The modulation of ß-catenin in peritoneum can be a novel tool to prevent PF.


Asunto(s)
Planta del Astrágalo , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Peritoneo/efectos de los fármacos , Extractos Vegetales/farmacología , beta Catenina/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Planta del Astrágalo/química , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/efectos de los fármacos , Humanos , Masculino , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Fibrosis Peritoneal/prevención & control , Peritoneo/citología , Peritoneo/metabolismo , Peritoneo/patología , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Ratas Sprague-Dawley , beta Catenina/metabolismo
5.
Kidney Blood Press Res ; 43(3): 1010-1022, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29940564

RESUMEN

Fibrosis and angiogenesis are the most common processes that result in progressive peritoneal tissue remodeling and, eventually, peritoneal membrane dysfunction. The role of exosomes, which contributes to intercellular communication, in these processes has been neglected. Various biomolecules, including DNA, mRNA, proteins, lipids, and particular certain miRNAs, can be transferred by exosomes to local, neighboring and distal cells. Upon stimulation by cytokines or other microenvironment stimuli, donor cells release a mass of exosomes to peritoneal mesothelial cells, further affecting fibrosis and angiogenesis. This important exosomes-mediated intracellular communication is thought to regulate peritoneal membrane function. Understanding the molecular mechanisms of these processes, targeting changes in exosomes and regulating exosomal miRNAs will advance therapeutic methods for protecting peritoneal membrane function.


Asunto(s)
Exosomas/fisiología , Membranas/fisiopatología , Peritoneo/ultraestructura , Comunicación Celular , Fibrosis , Humanos , Membranas/metabolismo , Neovascularización Patológica , Peritoneo/metabolismo , Peritoneo/patología
6.
Kidney Blood Press Res ; 42(2): 209-219, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28478435

RESUMEN

Chronic inflammation and angiogenesis are the most common complications in patients undergoing maintenance peritoneal dialysis (PD), resulting in progressive peritoneum remolding and, eventually, utrafiltration failure. Contributing to the deeper tissue under the peritoneal membrane, adipocytes play a neglected role in this process. Some adipokines act as inflammatory and angiogenic promoters, while others have the opposite effects. Adipokines, together with inflammatory factors and other cytokines, modulate inflammation and neovascularization in a coordinated fashion. This review will also emphasize cellular regulators and their crosstalk in long-term PD. Understanding the molecular mechanism, targeting changes in adipocytes and regulating adipokine secretion will help extend therapeutic methods for preventing inflammation and angiogenesis in PD.


Asunto(s)
Adipocitos/fisiología , Inflamación/etiología , Neovascularización Patológica/etiología , Diálisis Peritoneal/efectos adversos , Humanos
7.
Cell Physiol Biochem ; 40(6): 1538-1548, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997892

RESUMEN

BACKGROUND/AIMS: Peritoneal fibrosis is a frequent complication of peritoneal dialysis that follows inflammation. It is recognized that epithelial-mesenchymal transition (EMT) of peritoneal mesothelial cells (PMCs), plays a key role in fibrogenesis. However, the relationship between inflammatory macrophages and PMCs remains elusive. In this study, we investigated the effects of different polarized macrophages on EMT of HMrSV5 PMCs. METHODS: Monocytes were polarized to M1/M2 macrophages before being added to HMrSV5 in direct or indirect contact. Morphological changes of HMrSV5 were observed and toll-like receptors 4 (TLR4) on macrophage surfaces was detected using flow cytometry. EMT markers and intracellular signals of HMrSV5 cells were assessed using real time-PCR and WB. RESULTS: The typical epithelial cell morphology of HMrSV5 disappeared after co-culture with M1 macrophages and was accompanied by decreased E-cadherin and increased α-SMA, suggesting HMrSV5 undergo EMT. These effects depended on direct contact between the two cells, as indirect contact or co-culture with M2 macrophages had no effect. Intriguingly, we found TLR4 surface receptors were activated on sorted M1 cells in co-culture, and related signal adaptors, such as TRIF, were obviously upregulated. CONCLUSION: Direct contact with M1 macrophages induces EMT of PMCs, during which TRIF-dependent TLR4 signaling pathway was activated.


Asunto(s)
Polaridad Celular , Transición Epitelial-Mesenquimal , Epitelio/metabolismo , Macrófagos/metabolismo , Peritoneo/citología , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Forma de la Célula , Técnicas de Cocultivo , Regulación hacia Abajo/genética , Fibroblastos/citología , Humanos , Activación de Macrófagos , Monocitos/citología , Transducción de Señal , Regulación hacia Arriba/genética
8.
Cell Physiol Biochem ; 37(1): 43-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26278416

RESUMEN

BACKGROUND/AIMS: To investigate the effect of Astragaloside IV (AS-IV) on the regulation of the TGF-ß1/Smad signaling pathway in peritoneal mesothelial cells with an epithelial-to-mesenchymal transition (EMT). METHODS: EMT of human peritoneal mesothelial cells (HMrSV5) was induced using 2 ng/ml TGF-ß1. Cells were randomly divided into a vehicle group, a vehicle group with AS-IV, a TGF-ß1 treated group, and a TGF-ß1 treated group receiving varied doses of AS-IV or NAC. Real-time quantitative PCR and western blot were used to detect the expression of genes and proteins associated with the TGF-ß1/Smad signaling pathway and EMT. DCFH-DA was used to detect the generation of ROS in HMrSV5 cells, and a transwell migration assay was used to verify the capacity of AS-IV to inhibit EMT in HMrSV5 cells. Lentiviruses were used as carriers for the overexpression or knockdown of the Smad7 gene. RESULTS: Expression levels of E-cadherin (epithelial marker) was decreased and vimentin, α-SMA (EMT markers) and collagen I (extracellular matrix protein) phospho-Smad2/3, Snail1 and Snail2 was increased significantly in the TGF-ß1-treated HMrSV5 cells. AS-IV was associated with downregulated expression of vimentin and phospho-Smad2/3 in a dose-dependent manner, while the expression of Smad7 increased. Silenced or forced expression of Smad7 verified its role in the inhibitory effect of AS-IV on TGF-ß1-induced EMT in HMrSV5 cells. CONCLUSION: AS-IV effectively promotes the upregulation of Smad7 in the TGF-ß1/Smad signaling pathway during the EMT of HMrSV5 cells, indicating its potential therapeutic effect for the control of PF.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Saponinas/farmacología , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Triterpenos/farmacología , Actinas/metabolismo , Cadherinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Vimentina/metabolismo
9.
Int J Mol Sci ; 15(7): 12959-71, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25054320

RESUMEN

Inflammation and transforming growth factor-ß1 (TGF-ß1) contribute to the development of peritoneal fibrosis (PF), which is associated with peritoneal dialysis (PD). Astragalus membranaceus (Astragalus) has anti-inflammatory and anti-fibrotic effects in many diseases. The goal of this study was to determine the anti-fibrotic effects of Astragalus on the PF response to PD. A rat model of PD was induced using standard PD fluid, and PF was verified by HE and Masson's staining, as well as through the expression of fibroblast surface protein (FSP) and collagen III. The expression levels of monocyte chemoattractant protein (MCP)-1, F4/80 (macrophage/monocyte marker in rat), TGF-ß1 and the downstream proteins phospho-SMAD 2/3 in dialyzed peritoneal tissue treated with or without Astragalus was evaluated using immunohistochemistry analysis. Overall correlations between MCP-1 and TGF-ß1 staining were analyzed using both the Spearman and Pearson methods. The results showed that Astragalus could inhibit the recruitment and activation of monocytes/macrophages, thereby reducing the production of TGF-ß1 in the dialyzed peritoneal membrane. PF was also significantly decreased following treatment with Astragalus. MCP-1 expression had a strong positive correlation with TGF-ß1 sensitivity, suggesting that the anti-fibrotic function of Astragalus was mediated by MCP-1 and the TGF-ß1 pathway. Our results indicate that Astragalus could be a useful agent against PD-induced PF.


Asunto(s)
Astragalus propinquus/química , Quimiocina CCL2/metabolismo , Fibrosis Peritoneal/prevención & control , Extractos Vegetales/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Astragalus propinquus/metabolismo , Quimiocina CCL2/genética , Inmunohistoquímica , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Monocitos/citología , Monocitos/metabolismo , Diálisis Peritoneal , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/fisiología , Extractos Vegetales/química , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/genética
10.
Front Physiol ; 15: 1331976, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38390449

RESUMEN

Long-term peritoneal dialysis (PD) causes structural and functional alterations of the peritoneal membrane. Peritoneal deterioration and fibrosis are multicellular and multimolecular processes. Under stimulation by deleterious factors such as non-biocompatibility of PD solution, various cells in the abdominal cavity show differing characteristics, such as the secretion of different cytokines, varying protein expression levels, and transdifferentiation into other cells. In this review, we discuss the role of various cells in the abdominal cavity and their interactions in the pathogenesis of PD. An in-depth understanding of intercellular communication and inter-organ communication in PD will lead to a better understanding of the pathogenesis of this disease, enabling the development of novel therapeutic targets.

11.
Front Pharmacol ; 15: 1418485, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239655

RESUMEN

Background: To anticipate the potential molecular mechanism of Astragalus membranaceus (AM) and its monomer, Calycosin, against peritoneal fibrosis (PF) and related muscle atrophy using mRNA-seq, network pharmacology, and serum pharmacochemistry. Methods: Animal tissues were examined to evaluate a CKD-PF mice model construction. mRNA sequencing was performed to find differential targets. The core target genes of AM against PF were screened through network pharmacology analysis, and CKD-PF mice models were given high- and low-dose AM to verify common genes. Serum pharmacochemistry was conducted to clarify which components of AM can enter the blood circulation, and the selected monomer was further validated through cell experiments for the effect on PF and mesothelial mesenchymal transition (MMT) of peritoneal mesothelial cells (PMCs). Results: The CKD-PF mice models were successfully constructed. A total of 31,184 genes were detected in the blank and CKD-PF groups, and 228 transcription factors had significant differences between the groups. Combined with network pharmacology analysis, a total of 228 AM-PF-related targets were identified. Androgen receptor (AR) was the remarkable transcription factor involved in regulating transforming growth factor-ß1 (TGF-ß1). AM may be involved in regulating the AR/TGF-ß1 signaling pathway and may alleviate peritoneal dialysis-related fibrosis and muscle atrophy in CKD-PF mice. In 3% peritoneal dialysis solution-stimulated HMrSV5 cells, AR expression levels were dramatically reduced, whereas TGF-ß1/p-smads expression levels were considerably increased. Conclusion: AM could ameliorate PF and related muscle atrophy via the co-target AR and modulated AR/TGF-ß1 pathway. Calycosin, a monomer of AM, could partially reverse PMC MMT via the AR/TGF-ß1/smads pathway. This study explored the traditional Chinese medicine theory of "same treatment for different diseases," and supplied the pharmacological evidence of "AM can treat flaccidity syndrome."

12.
Phytomedicine ; 129: 155683, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38701543

RESUMEN

BACKGROUND: Peritoneal dialysis (PD) is a successful renal replacement therapy for end-stage renal disease. Long-term PD causes mesothelial-mesenchymal transition (MMT) of peritoneal mesothelial cells (PMCs), leading to peritoneal fibrosis (PF), which reduces the efficiency of PD. Macrophages are thought to play a role in the onset and perpetuation of peritoneal injury. However, the mechanisms by which macrophages-PMCs communication regulates peritoneal fibrosis are not fully understood resulting in a lack of disease-modifying drugs. Astragaloside IV (AS-IV) possessed anti-fibrotic effect towards PF in PD whereas the mechanistic effect of AS-IV in PD is unknown. METHODS: The primary macrophages were extracted and treated with LPS or AS-IV, then co-cultured with primary PMCs in transwell plates. The macrophage-derived exosomes were extracted and purified by differential centrifugation, then co-cultured with primary PMCs. Small RNA-seq was used to detect differential miRNAs in exosomes, and then KEGG analysis and q-PCR were performed for validation. In vivo PD rat models were established by inducing with high-glucose peritoneal dialysis fluid and different concentrations of AS-IV and exosomes were intraperitoneal injection. Through qRT-PCR, western blotting, and luciferase reporting, candidate proteins and pathways were validated in vivo and in vitro. The functions of the validated pathways were further investigated using the mimic or inhibition strategy. PF and inflammatory situations were assessed. RESULTS: We found AS-IV reversed the MMT of PMCs caused by LPS-stimulated macrophages and the improving effect was mediated by macrophage-derived exosomes in vitro. We also demonstrated that AS-IV significantly reduced the MMT of PMCs in vitro or PF in a rat PD model via regulating exosome-contained miR-204-5p which targets Foxc1/ß-catenin signaling pathway. CONCLUSION: AS-IV attenuates macrophage-derived exosomes induced fibrosis in PD through the miR-204-5p/Foxc1 pathway.


Asunto(s)
Exosomas , Macrófagos , MicroARNs , Fibrosis Peritoneal , Ratas Sprague-Dawley , Saponinas , Triterpenos , Fibrosis Peritoneal/tratamiento farmacológico , Animales , Exosomas/metabolismo , Exosomas/efectos de los fármacos , Saponinas/farmacología , Triterpenos/farmacología , Ratas , MicroARNs/metabolismo , Masculino , Macrófagos/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Modelos Animales de Enfermedad , Células Cultivadas , Técnicas de Cocultivo
13.
BMC Complement Med Ther ; 24(1): 204, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789949

RESUMEN

PURPOSE: This study aimed to evaluate the potential of astragalus polysaccharide (APS) pretreatment in enhancing the homing and anti-peritoneal fibrosis capabilities of bone marrow mesenchymal stromal cells (BMSCs) and to elucidate the underlying mechanisms. METHODS: Forty male Sprague-Dawley rats were allocated into four groups: control, peritoneal dialysis fluid (PDF), PDF + BMSCs, and PDF + APSBMSCs (APS-pre-treated BMSCs). A peritoneal fibrosis model was induced using PDF. Dil-labeled BMSCs were administered intravenously. Post-transplantation, BMSC homing to the peritoneum and pathological alterations were assessed. Stromal cell-derived factor-1 (SDF-1) levels were quantified via enzyme-linked immunosorbent assay (ELISA), while CXCR4 expression in BMSCs was determined using PCR and immunofluorescence. Additionally, a co-culture system involving BMSCs and peritoneal mesothelial cells (PMCs) was established using a Transwell setup to examine the in vitro effects of APS on BMSC migration and therapeutic efficacy, with the CXCR4 inhibitor AMD3100 deployed to dissect the role of the SDF-1/CXCR4 axis and its downstream impacts. RESULTS: In vivo and in vitro experiments confirmed that APS pre-treatment notably facilitated the targeted homing of BMSCs to the peritoneal tissue of PDF-treated rats, thereby amplifying their therapeutic impact. PDF exposure markedly increased SDF-1 levels in peritoneal and serum samples, which encouraged the migration of CXCR4-positive BMSCs. Inhibition of the SDF-1/CXCR4 axis through AMD3100 application diminished BMSC migration, consequently attenuating their therapeutic response to peritoneal mesenchyme-to-mesothelial transition (MMT). Furthermore, APS upregulated CXCR4 expression in BMSCs, intensified the activation of the SDF-1/CXCR4 axis's downstream pathways, and partially reversed the AMD3100-induced effects. CONCLUSION: APS augments the SDF-1/CXCR4 axis's downstream pathway activation by increasing CXCR4 expression in BMSCs. This action bolsters the targeted homing of BMSCs to the peritoneal tissue and amplifies their suppressive influence on MMT, thereby improving peritoneal fibrosis.


Asunto(s)
Planta del Astrágalo , Quimiocina CXCL12 , Células Madre Mesenquimatosas , Fibrosis Peritoneal , Polisacáridos , Ratas Sprague-Dawley , Receptores CXCR4 , Animales , Receptores CXCR4/metabolismo , Quimiocina CXCL12/metabolismo , Ratas , Masculino , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/metabolismo , Polisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Modelos Animales de Enfermedad , Ciclamas/farmacología
14.
J Ethnopharmacol ; 331: 118335, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754644

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis in the world, it is one of the most common causes of kidney disease and can lead to end-stage kidney disease, however, its pathogenesis is still complicated. The Shen-yan-yi-hao oral solution (SOLI) is an effective prescription for the clinical treatment of IgAN while its specific mechanism remains to be further elucidated. AIM OF THE STUDY: This study investigates SOLI's effects on IgAN in rats, particularly on the intestinal mucosal barrier, and identifies potential therapeutic targets through network pharmacology and molecular docking, validated experimentally. MATERIALS AND METHODS: Target genes for SOLI in IgAN were identified and analysed through molecular docking and KEGG pathway enrichment. An IgAN rat model examined SOLI's effect on renal biomarkers and cytokines involved in specific pathways, ileum mucosal lesions, and the intestinal immune system. The IL-17 pathway's role was studied in IEC-6 cells with SOLI in vitro. RESULT: Rats developed increased proteinuria and kidney damage marked by IgA deposition and inflammation. SOLI treatment significantly ameliorated these symptoms, reduced galactose-deficient Ig A1 (Gd-IgA1), and decreased cytokines like IL-17, TNF-α, IL-6 and IL-1ß etc. SOLI also normalized intestinal tight junction protein expression, ameliorated intestinal damage, and regulated intestinal immune response (focused on IL-17/NF-κB signal pathway). SOLI moderated the abnormally activated IL-17 pathway, which damages intestinal epithelial cells, suggesting IgAN treatment potential. CONCLUSION: SOLI reduces proteinuria and enhances intestinal mucosal function in IgAN rats, kidney protection in the IgAN rat model may initiate from modulating the intestinal IL-17/NF-κB pathway and subsequent Gd-IgA1 accumulation.


Asunto(s)
Medicamentos Herbarios Chinos , Glomerulonefritis por IGA , Interleucina-17 , Mucosa Intestinal , Simulación del Acoplamiento Molecular , FN-kappa B , Transducción de Señal , Animales , Glomerulonefritis por IGA/tratamiento farmacológico , FN-kappa B/metabolismo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/administración & dosificación , Interleucina-17/metabolismo , Ratas , Masculino , Transducción de Señal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratas Sprague-Dawley , Administración Oral , Línea Celular , Modelos Animales de Enfermedad , Farmacología en Red , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Citocinas/metabolismo
15.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 33(12): 1636-41, 2013 Dec.
Artículo en Zh | MEDLINE | ID: mdl-24517060

RESUMEN

OBJECTIVE: To evaluate the clinical efficacy and safety of treatment of chronic primary glomerulopathy (CPG) patients of Shen deficiency and dampness heat syndrome (SDDHS) by Yishen Qingli Granule (YQG) combined with low-dose Tripterygium Wilfordii multiglycoside Tablet (TWT). METHODS: Totally 231 CPG patients of SDDHS were enrolled in this study (including 60 patients from First Affiliated Hospital of Nanjing University of Chinese Medicine, 58 from First Affiliated Hospital of Nanjing Medical University, 46 from Xinqiao Hospital of Third Military Medical University, 35 from First Affiliated Hospital of Guangzhou University of Chinese Medicine, 14 from First Affiliated Hospital of Soochow University, and 18 from Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine). They were randomly assigned to the control group (116 cases) and the trial group (115 cases) according to block group method. There were 217 cases in the safety analysis set (109 cases in the trial group vs 108 cases in the control group), and 203 cases in the full analysis set (99 cases in the trial group vs 104 cases in the control group). All patients received basic treatment such as ACEI/ARB. Furthermore, YQG (consisting of raw astragalus 10 g, prepared Polygonum Multiflorum 10 g, Pyrrosia 10 g, 1.5 g each package, containing 10 g of crude drugs) was additionally given to patients in the trial group, each package, twice daily. The TWT (10 mg) was given, twice a day. The TWT dose was adjusted according to 24 h urinary total protein (UTP). The placebos of YQG and TWT were administered to those in the control group. The treatment course consisted of 24 weeks and the follow-up visit lasted for 24 weeks. The biochemical indices were observed before and after treatment including 24 h UTP, urine red cell count (U(RBC)), renal functions (BUN, SCr), blood routine test (WBC), and liver functions (SGPT, SGOT). Reverse reactions such as gastrointestinal discomfort, skin rash, and irregular menstruation were also observed. RESULTS: Compared with the control group, the total effective rate was better in the trial group (82.83% vs 61.54%, P < 0.01). Results of stratified comparison of UTP showed better efficacy in the trial group (0.8-3.0 g/24 h, P < 0.01). The UTP decline occurred in the trial group after 8 weeks of treatment, with stable action, showing statistical difference when compared with the control group (P < 0.01). In the trial group, U(RBC) level decreased after treatment but changed more significantly. But there was no statistical difference in the changes when compared with the control group (P > 0.05). After treatment, there were no statistical difference in safety indicators such as WBC, SGPT, and SGOT between the two groups after treatment (P > 0.05). CONCLUSION: On the basis of basic treatment such as ACEI/ARB, application of YQG combined with low-dose TWT had better effect in controlling proteinuria of CPG patients, and could help stabilizing their conditions with less adverse reactions.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Enfermedades Renales/diagnóstico , Enfermedades Renales/tratamiento farmacológico , Fitoterapia/métodos , Adulto , Femenino , Humanos , Glomérulos Renales/patología , Masculino , Medicina Tradicional China , Persona de Mediana Edad , Resultado del Tratamiento , Tripterygium
16.
J Ethnopharmacol ; 309: 116343, 2023 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-36906159

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese medicine, Centella asiatica (L.) Urb., has been extensively utilized in clinics to treat a variety of fibrotic disorders. Asiaticoside (ASI), as an important active ingredient, has attracted much attention in this field. However, the effect of ASI on peritoneal fibrosis (PF) is still unclear. Therefore, we evaluated the benefits of ASI for PF and mesothelial-mesenchymal transition (MMT) and revealed the underlying mechanisms. AIM OF STUDY: The objective of this investigation was to anticipate the potential molecular mechanism of ASI against peritoneal mesothelial cells (PMCs) MMT employing proteomics and network pharmacology, and to confirm it using in vivo and in vitro studies. MATERIALS AND METHODS: The mesentery of peritoneal fibrosis mice and normal mice were analyzed quantitatively for proteins that were differentially expressed using a technique tandem mass tag (TMT). Next, the core target genes of ASI against PF were screened through network pharmacology analysis, and PPI and C-P‒T networks were constructed by Cytoscape Version 3.7.2. According to the findings of a GO and KEGG enrichment analysis of differential proteins and core target genes, the signaling pathway with a high correlation degree was selected as the key signaling pathway of ASI inhibiting the PMCs MMT for further molecular docking analysis and experimental verification. RESULTS: TMT-based quantitative proteome analysis revealed the identification of 5727 proteins, of which 70 were downregulated and 178 were upregulated. Among them, the levels of STAT1, STAT2, and STAT3 in the mesentery of mice with peritoneal fibrosis were considerably lower than in the control group, indicating a role for the STAT family in the pathogenesis of peritoneal fibrosis. Then, a total of 98 ASI-PF-related targets were identified by network pharmacology analysis. JAK2 is one of the top 10 core target genes representing a potential therapeutic target. JAK/STAT signaling may represent a core pathway mediating PF effects by ASI. Molecular docking studies showed that ASI had the potential to interact favorably with target genes involved in the JAK/STAT signaling pathway, such as JAK2 and STAT3. The experimental results showed that ASI could significantly alleviate Chlorhexidine Gluconate (CG)-induced peritoneal histopathological changes and increase JAK2 and STAT3 phosphorylation levels. In TGF-ß1-stimulated HMrSV5 cells, E-cadherin expression levels were dramatically reduced whereas Vimentin, p-JAK2, α-SMA, and p-STAT3 expression levels were considerably increased. ASI inhibited the TGF-ß1-induced HMrSV5 cell MMT, decreased the activation of JAK2/STAT3 signaling, and increased the nuclear translocation of p-STAT3, which was consistent with the effect of the JAK2/STAT3 pathway inhibitor AG490. CONCLUSION: ASI can inhibit PMCs MMT and alleviate PF by regulating the JAK2/STAT3 signaling pathway.


Asunto(s)
Fibrosis Peritoneal , Ratones , Animales , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/genética , Factor de Crecimiento Transformador beta1/metabolismo , Simulación del Acoplamiento Molecular , Farmacología en Red , Proteómica , Línea Celular , Transición Epitelial-Mesenquimal , Transducción de Señal
17.
Nutrients ; 15(2)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36678231

RESUMEN

BACKGROUND: Targeting the gut microbiota may become a new therapeutic to prevent and delay the progression of chronic kidney disease (CKD). Nonetheless, the causal relationship between specific intestinal flora and CKD is still unclear. MATERIALS AND METHOD: To identify genetically predicted microbiota, we used summary data from genome-wide association studies on gut microbiota in 18340 participants from 24 cohorts. Furthermore, we genetically predicted the causal relationship between 211 gut microbiotas and six phenotypes (outcomes) (CKD, estimated glomerular filtration rate (eGFR), urine albumin to creatinine ratio (UACR), dialysis, rapid progress to CKD, and rapid decline of eGFR). Four Mendelian randomization (MR) methods, including inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode were used to investigate the casual relationship between gut microbiotas and various outcomes. The result of IVW was deemed as the primary result. Then, Cochrane's Q test, MR-Egger, and MR-PRESSO Global test were used to detect heterogeneity and pleiotropy. The leave-one method was used for testing the stability of MR results and Bonferroni-corrected was used to test the strength of the causal relationship between exposure and outcome. RESULTS: Through the MR analysis of 211 microbiotas and six clinical phenotypes, a total of 36 intestinal microflora were found to be associated with various outcomes. Among them, Class Bacteroidia (=-0.005, 95% CI: -0.001 to -0.008, p = 0.002) has a strong causality with lower eGFR after the Bonferroni-corrected test, whereas phylum Actinobacteria (OR = 1.0009, 95%CI: 1.0003-1.0015, p = 0.0024) has a strong causal relationship with dialysis. The Cochrane's Q test reveals that there is no significant heterogeneity between various single nucleotide polymorphisms. In addition, no significant level of pleiotropy was found according to MR-Egger and MR-PRESSO Global tests. CONCLUSIONS: Through the two-sample MR analysis, we identified the specific intestinal flora that has a causal relationship with the incidence and progression of CKD at the level of gene prediction, which may provide helpful biomarkers for early disease diagnosis and potential therapeutic targets for CKD.


Asunto(s)
Microbioma Gastrointestinal , Insuficiencia Renal Crónica , Humanos , Microbioma Gastrointestinal/genética , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Diálisis Renal , Insuficiencia Renal Crónica/genética , Polimorfismo de Nucleótido Simple , Riñón/fisiología
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(5): 466-471, 2022 May.
Artículo en Zh | MEDLINE | ID: mdl-35603656

RESUMEN

Fibrosis is a pathological healing process which occurs in the progression of various chronic diseases after tissue injury. Extracellular vesicles are involved in tissue fibrosis through communication between cells. Macrophages are important immune cells that maintain tissue integrity by eliminating or repairing damaged cells and matrix. Macrophages can directly regulate tissue fibrosis by secreting extracellular vesicles and transporting microRNAs, lncRNAs and fibrogenic factors to target cells of lungs, heart, liver, kidney and other organs. At the same time, chronic diseases are often accompanied by inflammation. A large number of macrophages are recruited to the site of injury, and local or distal cells transport extracellular vesicles to macrophages to regulate the polarization of macrophages, thus affecting the development and prognosis of tissue fibrosis.


Asunto(s)
Vesículas Extracelulares , Macrófagos , Fibrosis , Humanos , Riñón/patología , Hígado/patología
19.
FEBS Open Bio ; 12(4): 784-797, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35226797

RESUMEN

As a potential source of myofibroblasts, pericytes may play a role in human peritoneal fibrosis. The culture of primary vascular pericytes in animals has previously been reported, most of which are derived from cerebral and retinal microvasculature. Here, in the field of peritoneal dialysis, we describe a method to isolate and characterize mouse peritoneal microvascular pericytes. The mesenteric tissues of five mice were collected and digested by type II collagenase and type I DNase. After cell attachment, the culture fluid was replaced with pericyte-conditioned medium. Pericytes with high purity (99.0%) could be isolated by enzymatic disaggregation combined with conditional culture and magnetic activated cell sorting. The primary cells were triangular or polygonal with protrusions, and confluent cell culture could be established in 3 days. The primary pericytes were positive for platelet-derived growth factor receptor-ß, α-smooth muscle actin, neuron-glial antigen 2, and CD13. Moreover, they promoted formation of endothelial tubes, and pericyte-myofibroblast transition occurred after treatment with transforming growth factor-ß1. In summary, we describe here a reproducible isolation protocol for primary peritoneal pericytes, which may be a powerful tool for in vitro peritoneal fibrosis studies.


Asunto(s)
Miofibroblastos , Pericitos , Animales , Ratones , Miofibroblastos/metabolismo , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
20.
FEMS Microbiol Lett ; 369(1)2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36368696

RESUMEN

Acute kidney injury (AKI) is a global public health hazard with high morbidity and mortality. Sepsis accounts for nearly half of all causes of AKI. Scientists have made a great effort to explore effective therapeutic agents with limited side effects in the treatment of AKI, but have had little success. With the development of gut flora study, Akkermansia muciniphila (A. muciniphila) has been proven to prevent different organs by regulating the inflammatory response. However, the reno-protective function is still unknown. Here, the AKI model was induced using lipopolysaccharide (LPS) in mice with or without pretreatment of A. muciniphila. Renal function and histological change were measured. Inflammatory factors were detected by ELISA and rt-PCR. TLR4/NF-κB signaling factors and NLRP3 inflammasome were tested by western blot and immunohistochemistry. Pretreatment of A. muciniphila markedly inhibited inflammatory response and ameliorated kidney histopathological changes. Furthermore, the TLR4, p-NF-κB p65, and downstream IκBα were notably activated in the model group and inhibited by A. muciniphila. A similar effect was found in the regulation of NLRP3 inflammasome. In conclusion, pretreatment with A. muciniphila could protect against LPS-induced AKI by inhibition of the TLR4/NF-κB pathway and NLRP3 inflammasome activation. It may be a new therapeutic strategy for AKI prevention and treatment in the future.


Asunto(s)
Lesión Renal Aguda , Akkermansia , FN-kappa B , Animales , Ratones , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/prevención & control , Inflamasomas/metabolismo , Lipopolisacáridos/toxicidad , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor Toll-Like 4 , Akkermansia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA