Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
BMC Complement Med Ther ; 24(1): 204, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38789949

RESUMO

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.


Assuntos
Astrágalo , Quimiocina CXCL12 , Células-Tronco Mesenquimais , Fibrose Peritoneal , Polissacarídeos , Ratos Sprague-Dawley , Receptores CXCR4 , Animais , Receptores CXCR4/metabolismo , Quimiocina CXCL12/metabolismo , Ratos , Masculino , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/metabolismo , Polissacarídeos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Modelos Animais de Doenças , Ciclamos/farmacologia
2.
J Ethnopharmacol ; 331: 118335, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38754644

RESUMO

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.


Assuntos
Medicamentos de Ervas Chinesas , Glomerulonefrite por IGA , Interleucina-17 , Mucosa Intestinal , Simulação de Acoplamento Molecular , NF-kappa B , Transdução de Sinais , Animais , Glomerulonefrite por IGA/tratamento farmacológico , NF-kappa B/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/administração & dosagem , Interleucina-17/metabolismo , Ratos , Masculino , Transdução de Sinais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Ratos Sprague-Dawley , Administração Oral , Linhagem Celular , Modelos Animais de Doenças , Farmacologia em Rede , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Citocinas/metabolismo
3.
Phytomedicine ; 129: 155683, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38701543

RESUMO

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.


Assuntos
Exossomos , Macrófagos , MicroRNAs , Fibrose Peritoneal , Ratos Sprague-Dawley , Saponinas , Triterpenos , Fibrose Peritoneal/tratamento farmacológico , Animais , Exossomos/metabolismo , Exossomos/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Ratos , MicroRNAs/metabolismo , Masculino , Macrófagos/efeitos dos fármacos , Diálise Peritoneal/efeitos adversos , Modelos Animais de Doenças , Células Cultivadas , Técnicas de Cocultura
4.
Front Physiol ; 15: 1331976, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38390449

RESUMO

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.

5.
J Cell Physiol ; 238(4): 813-828, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36879552

RESUMO

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.


Assuntos
Alantoína , Prurido , Insuficiência Renal Crônica , Animais , Camundongos , Alantoína/efeitos adversos , Cálcio , Prurido/induzido quimicamente , Prurido/diagnóstico , Receptores Acoplados a Proteínas G , Insuficiência Renal Crônica/complicações
6.
J Ethnopharmacol ; 309: 116343, 2023 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-36906159

RESUMO

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.


Assuntos
Fibrose Peritoneal , Camundongos , Animais , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/genética , Fator de Crescimento Transformador beta1/metabolismo , Simulação de Acoplamento Molecular , Farmacologia em Rede , Proteômica , Linhagem Celular , Transição Epitelial-Mesenquimal , Transdução de Sinais
7.
Nutrients ; 15(2)2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36678231

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Insuficiência Renal Crônica , Humanos , Microbioma Gastrointestinal/genética , Estudo de Associação Genômica Ampla , Análise da Randomização Mendeliana , Diálise Renal , Insuficiência Renal Crônica/genética , Polimorfismo de Nucleotídeo Único , Rim/fisiologia
8.
FEMS Microbiol Lett ; 369(1)2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36368696

RESUMO

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.


Assuntos
Injúria Renal Aguda , Akkermansia , NF-kappa B , Animais , Camundongos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/prevenção & controle , Inflamassomos/metabolismo , Lipopolissacarídeos/toxicidade , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptor 4 Toll-Like , Akkermansia/fisiologia
9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(5): 466-471, 2022 May.
Artigo em Chinês | MEDLINE | ID: mdl-35603656

RESUMO

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.


Assuntos
Vesículas Extracelulares , Macrófagos , Fibrose , Humanos , Rim/patologia , Fígado/patologia
10.
FEBS Open Bio ; 12(4): 784-797, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35226797

RESUMO

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.


Assuntos
Miofibroblastos , Pericitos , Animais , Camundongos , Miofibroblastos/metabolismo , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
11.
Am J Chin Med ; 50(1): 261-274, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34983328

RESUMO

Peritoneal fibrosis (PF) is a disease caused by prolonged exposure of the peritoneum to high levels of dialysis fluid. Astragalus total saponins (ATS) is a phytochemical naturally occurring in Radix Astragali that has anti-inflammatory and anti-oxidant properties. In this study, we constructed an in vivo model of PF using 4.25% glucose-containing administered intraperitoneally to rats and incubated peritoneal mesothelial cells (PMCs) with 4.25% glucose-containing peritoneal dialysis fluid to construct an in vitro model of PF. Furthermore, siRNA of PGC-1[Formula: see text] was used to inhibit the expression of PGC-1[Formula: see text] to further investigate the mechanism of the protective effect of ATS on PF. In both in vivo and in vitro models, ATS treatment showed a protective effect against PF, with ATS reducing the thickness of peritoneal tissues in PF rats, increasing the viability of PMCs, increasing the mitochondrial membrane potential and reducing apoptosis ratio. ATS treatment also reduced the expressions of peritoneal fibrosis markers (Smad2, p-Smad2 and [Formula: see text]-SMA) and apoptosis markers (Caspase3, cleaved-Caspase3 and Bax) and restored the expressions of mitochondrial synthesis proteins (PGC-1[Formula: see text], NRF1 and TFAM) in ATS-treated peritoneal tissues or PMCs. Furthermore, in the presence of PGC-1[Formula: see text] inhibition, the protective effect of ATS on PF was blocked. In conclusion, ATS treatment may be an effective therapeutic agent to inhibit high glucose-induced in peritoneal fibrosis through PGC-1[Formula: see text]-mediated apoptosis.


Assuntos
Fibrose Peritoneal , Saponinas , Animais , Apoptose , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/tratamento farmacológico , Fibrose Peritoneal/prevenção & controle , Peritônio/metabolismo , Peritônio/patologia , Ratos , Saponinas/metabolismo , Saponinas/farmacologia , Transdução de Sinais
12.
Front Pharmacol ; 12: 649581, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33927624

RESUMO

Angiogenesis of human peritoneal vascular endothelial cells (HPVECs), linked to vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) signaling, is a complication of peritoneal fibrosis (PF). Hippo/YAP signaling interacts with VEGF/VEGFR2 signaling, but the effect on peritoneal angiogenesis and PF has not been studied. We tested VEGF/Hippo/YAP inhibition by tetramethylpyrazine (TMP) in PF mice and HPVECs. This treatment ameliorated peritoneal dialysis (PD)-induced angiogenesis and PF. In mice, PF was associated with upregulation of VEGF, and TMP ameliorated submesothelial fibrosis, perivascular bleeding, and Collagen I abundance. In HPVECs, angiogenesis occurred due to human peritoneal mesothelial cells (HPMCs)-conditioned medium, and TMP alleviated HPVECs migration, tube formation, and YAP nuclear translocation. YAP knockdown PF mouse and HPVEC models were established to further confirm our finding. YAP deletion attenuated the PD-induced or VEGF-induced increase in angiogenesis and PF. The amount of CYR61 and CTGF was significantly less in the YAP knockdown group. To study the possibility that TMP could benefit angiogenesis, we measured the HPVECs migration and tube formation and found that both were sharply increased in YAP overexpression; TMP treatment partly abolished these increases. As well, the amount of VEGFR localized in the trans-Golgi network was lower by double immunofluorescence; VEGFR and its downstream signaling pathways including p-ERK, p-P38, and p-Akt were more in HPVECs with YAP overexpression. Overall, TMP treatment ameliorated angiogenesis, PF, and peritoneum injury. These changes were accompanied by inhibition of VEGF/Hippo/YAP.

13.
Artigo em Inglês | MEDLINE | ID: mdl-33144869

RESUMO

BACKGROUND: It is generally considered that traditional Chinese medicine (TCM) therapy postpones the progression of some chronic kidney diseases (CKDs). Chinese medicine herbs are widely applied in TCM therapy. We aimed to evaluate clinical efficacy and safety of Chinese herbal formula granules in patients with CKD stage 3 through a prospective randomized controlled study. METHODS: A total of 343 participants with CKD stage 3 were recruited from 9 hospitals in Jiangsu Province between April 2014 and October 2016. Participants were randomly assigned to a treatment or control group. Patients in the treatment group orally took Chinese herbal formula granules twice a day, while controls received placebo granules. The duration of intervention was 24 weeks. Primary outcomes were 24-hour proteinuria, serum creatinine, and eGFR, which were measured every 4 weeks. RESULTS: There was no statistical difference in 24-hour proteinuria between the two groups (0.97 ± 1.14 g/d vs. 0.97 ± 1.25 g/d). Patients in the treatment group had significantly lower serum creatinine level (130.78 ± 32.55 µmol/L versus 149.12 ± 41.27 µmol/L) and significantly higher eGFR level (55.74 ± 50.82 ml/min/1.73·m2 versus 44.46 ± 12.60 ml/min/1.73·m2) than those in the control group (P < 0.05). There was no significant difference between two groups in the incidence of adverse events. CONCLUSION: The treatment adopting Chinese herbal formula granules for 24 weeks improved kidney function of patients with CKD stage 3.

14.
Artigo em Inglês | MEDLINE | ID: mdl-32802122

RESUMO

OBJECTIVE: Tai Chi and Qigong are the two similar traditional Chinese wellness exercises. A strong body of published clinical randomized controlled trials (RCTs) has investigated the health benefits of Tai Chi and Qigong exercises (TCQE) in patients with essential hypertension (EH). This is the first meta-analysis to evaluate the efficacy of TCQE on blood pressure (BP) and blood levels of nitric oxide (NO) and endothelin-1 (ET-1) in EH patients and explore the potential antihypertensive mechanism of TCQE. METHODS: We conducted a literature retrieval for Chinese and English studies in seven databases from their respective inceptions until January 14, 2020. All RCTs examining clinical efficacy of TCQE for EH patients were considered. The major therapeutic outcomes of TCQE were changes in the blood levels of NO, ET-1, and BP in EH patients. Methodological quality of the included RCTs was detected via The Cochrane Risk of Bias tool. We evaluated the data reported and performed the meta-analysis by Review Manager 5.3 software. RESULTS: 9 RCTs involving 516 EH patients were included. The intervention duration lasted from 1.5 months to 6 months. The results of comprehensive analysis showed that compared with control interventions, experimental interventions were more effective in reducing the systolic blood pressure and the diastolic blood pressure and contributed higher blood levels of NO and lower blood levels of ET-1. CONCLUSIONS: TCQE could be an effective complementary and alternative therapy for EH. The lower BP in EH patients who practice TCQE may have some connection with exercise-related increased blood NO levels and decreased blood ET-1 levels. However, further research is needed to make clear the efficacy of TCQE in management of EH and the mechanism of lowering BP in TCQE.

15.
Cell Mol Biol Lett ; 25: 33, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32514269

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Humanos , Estresse Oxidativo/efeitos dos fármacos , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
16.
J Cell Biochem ; 120(4): 5505-5509, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30474893

RESUMO

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.


Assuntos
Cistanche , Medicamentos de Ervas Chinesas/administração & dosagem , Nefrite , Zumbido , Adulto , Doença Crônica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nefrite/complicações , Nefrite/tratamento farmacológico , Estudos Prospectivos , Zumbido/tratamento farmacológico , Zumbido/etiologia
17.
Cell Physiol Biochem ; 51(6): 2794-2813, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30562743

RESUMO

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.


Assuntos
Astrágalo , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Peritônio/efeitos dos fármacos , Extratos Vegetais/farmacologia , beta Catenina/genética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Astrágalo/química , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/efeitos dos fármacos , Humanos , Masculino , Fibrose Peritoneal/genética , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Fibrose Peritoneal/prevenção & controle , Peritônio/citologia , Peritônio/metabolismo , Peritônio/patologia , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Ratos Sprague-Dawley , beta Catenina/metabolismo
18.
Kidney Blood Press Res ; 43(3): 1010-1022, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29940564

RESUMO

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.


Assuntos
Exossomos/fisiologia , Membranas/fisiopatologia , Peritônio/ultraestrutura , Comunicação Celular , Fibrose , Humanos , Membranas/metabolismo , Neovascularização Patológica , Peritônio/metabolismo , Peritônio/patologia
19.
Int J Clin Exp Pathol ; 11(9): 4296-4304, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31949826

RESUMO

Diabetic nephropathy is a common complication of type 2 diabetes and is related to the epithelial to mesenchymal transition. In this study, we aimed to find whether the RhoA/ROCK pathway affects the development of diabetic nephropathy caused by the epithelial to mesenchymal transition both in vivo and in vitro. The results show that inhibition of the RhoA/ROCK signaling pathway improved the pathology and degree of fibrosis in diabetic nephropathy as determined by hematoxylin and eosin staining and Masson staining. We also found, using immunohistochemistry and quantitative reverse transcription polymerase chain reaction, that a RhoA/Rock inhibitor regulated relative protein and gene expression levels in a dose-dependent manner. Furthermore, the inhibitor improved fibrosis induced by high levels of glucose in HK-2 cells by suppressing E-cadherin, α-SMA, and FSP-1 expression. In conclusion, the RhoA/ROCK signaling pathway plays an important role in the development of diabetic nephropathy and could be a potential therapeutic target for type 2 diabetes.

20.
Kidney Blood Press Res ; 42(2): 209-219, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28478435

RESUMO

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.


Assuntos
Adipócitos/fisiologia , Inflamação/etiologia , Neovascularização Patológica/etiologia , Diálise Peritoneal/efeitos adversos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA