Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Circ Res ; 133(1): 71-85, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37232163

RESUMO

BACKGROUND: As a part of natural disease progression, acute kidney injury (AKI) can develop into chronic kidney disease via renal fibrosis and inflammation. LTBP4 (latent transforming growth factor beta binding protein 4) regulates transforming growth factor beta, which plays a role in renal fibrosis pathogenesis. We previously investigated the role of LTBP4 in chronic kidney disease. Here, we examined the role of LTBP4 in AKI. METHODS: LTBP4 expression was evaluated in human renal tissues, obtained from healthy individuals and patients with AKI, using immunohistochemistry. LTBP4 was knocked down in both C57BL/6 mice and human renal proximal tubular cell line HK-2. AKI was induced in mice and HK-2 cells using ischemia-reperfusion injury and hypoxia, respectively. Mitochondrial division inhibitor 1, an inhibitor of DRP1 (dynamin-related protein 1), was used to reduce mitochondrial fragmentation. Gene and protein expression were then examined to assess inflammation and fibrosis. The results of bioenergetic studies for mitochondrial function, oxidative stress, and angiogenesis were assessed. RESULTS: LTBP4 expression was upregulated in the renal tissues of patients with AKI. Ltbp4-knockdown mice showed increased renal tissue injury and mitochondrial fragmentation after ischemia-reperfusion injury, as well as increased inflammation, oxidative stress, and fibrosis, and decreased angiogenesis. in vitro studies using HK-2 cells revealed similar results. The energy profiles of Ltbp4-deficient mice and LTBP4-deficient HK-2 cells indicated decreased ATP production. LTBP4-deficient HK-2 cells exhibited decreased mitochondrial respiration and glycolysis. Human aortic endothelial cells and human umbilical vein endothelial cells exhibited decreased angiogenesis when treated with LTBP4-knockdown conditioned media. Mitochondrial division inhibitor 1 treatment ameliorated inflammation, oxidative stress, and fibrosis in mice and decreased inflammation and oxidative stress in HK-2 cells. CONCLUSIONS: Our study is the first to demonstrate that LTBP4 deficiency increases AKI severity, consequently leading to chronic kidney disease. Potential therapies focusing on LTBP4-associated angiogenesis and LTBP4-regulated DRP1-dependent mitochondrial division are relevant to renal injury.


Assuntos
Injúria Renal Aguda , Insuficiência Renal Crônica , Traumatismo por Reperfusão , Animais , Humanos , Camundongos , Injúria Renal Aguda/prevenção & controle , Células Endoteliais/metabolismo , Fibrose , Inflamação/metabolismo , Rim/metabolismo , Proteínas de Ligação a TGF-beta Latente , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Insuficiência Renal Crônica/complicações , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Fator de Crescimento Transformador beta/metabolismo
2.
Cancer Lett ; 563: 216192, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37088327

RESUMO

Immune checkpoint inhibitors are groundbreaking resources for cancer therapy. However, only a few patients with hepatocellular carcinoma (HCC) have shown positive responses to anti-PD-1 therapy. Neoantigens are sequence-altered proteins resulting from somatic mutations in cancer. This study identified the neoantigens of Hep-55.1C and Dt81 Hepa1-6 HCCs by comparing their whole exome sequences with those of a normal C57BL/6 mouse liver. Immunogenic long peptides were pooled as peptide vaccines. The vaccination elicited tumor-reactive immune responses in C57BL/6 mice, as demonstrated by IFN-γ ELISPOT and an in vitro killing assay of splenocytes. In the treatment of three mouse HCC models, combined neoantigen vaccination and anti-PD-1 resulted in more significant tumor regression than monotherapies. Flow cytometry of the tumor-infiltrating lymphocytes showed decreased Treg cells and monocytic myeloid-derived suppressor cells, increased CD8+ T cells, enhanced granzyme B expression, and reduced exhaustion-related markers PD-1 and Lag-3 on CD8+ T cells in the combination group. These findings provide a strong rationale for conducting clinical studies of using neoantigen vaccination in combination with anti-PD-1 to treat patients with HCC.


Assuntos
Vacinas Anticâncer , Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Linfócitos T CD8-Positivos , Camundongos Endogâmicos C57BL , Vacinas Anticâncer/farmacologia
3.
Colloids Surf B Biointerfaces ; 222: 113129, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36610364

RESUMO

A new drug delivery system (DDS) type complexing magnetic nanoparticles (MNP) along with boron nanosheets (BNN) coated with a pH-responsive polymer-polyethylene glycol (PEG) for the manageable loading/release of the anti-cancerous drug, doxorubicin (DOX), was created (MNP-BNN-PEG-DOX). The X-ray diffraction patterns of the nanocomposites displayed wide diffraction peaks for BNN at 25.1° and 42.3°, belonging to the (002) and (100) planes, correspondingly. Additionally, the characteristic peaks of Fe3O4 appeared at 30.5°, 35.9°, 43.6°, 54.1°, 57.5°, and 63.2°, belonging to the (220), (311), (400), (422), (511), and (440) crystal planes, correspondingly. Moreover, the magnetic properties of the nanocomposites revealed that the MNP-BNN remained magnetic after coating with PEG. The saturation magnetization (Ms) of the uncoated-MNP-BNN and MNP-BNN-PEG-1 were 49.4 and 42.3 emu g-1, respectively. Both in vitro and in vivo analyses shown that DDS might inhibit tumor growth, provoke cancer cell apoptosis, and reduce the cytotoxic effects of DOX. In vivo analysis demonstrated that after treatment with phosphate-buffered saline (PBS), MNP-BNN-PEG-1, free DOX, and MNP-BNN-PEG-1-DOX, the average tumor growth and weight were 1906, 1997, 1188, and 1043 nm and 0.17, 0.20, 0.13, and 0.07 g, respectively. The MNP-BNN-PEG-DOX nanoparticles could be an effective treatment and potential alternative for liver cancer therapy.


Assuntos
Antineoplásicos , Neoplasias Hepáticas , Humanos , Doxorrubicina , Antineoplásicos/química , Sistemas de Liberação de Medicamentos , Neoplasias Hepáticas/tratamento farmacológico , Polietilenoglicóis/química , Concentração de Íons de Hidrogênio , Fenômenos Magnéticos , Portadores de Fármacos/química
4.
Pharmacol Res ; 188: 106646, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36621619

RESUMO

The efficacy of treatment for advanced hepatocellular carcinoma (HCC) has remained limited. Polyinosinic-polycytidylic acid-poly-L-lysine carboxymethylcellulose (poly-ICLC) is a synthetic double-stranded RNA that serves as a viral mimic and induces an immune response. Intratumoral (IT) poly-ICLC injections can induce an autovaccination effect and prime the immune system, whereas intramuscular (IM) injection of poly-ICLC can attract and maintain tumor-specific cytotoxic T lymphocytes in tumors. We found that IT injection of poly-ICLC upregulated the expression of CD83 and CD86 on conventional type 1 dendritic cells in tumors. Combination therapy with IT followed by IM injections of poly-ICLC significantly inhibited tumor growth and increased the tumor-infiltrating CD8+ T cells in two syngeneic mouse models of HCC. Depletion of CD8+ T cells attenuated the antitumor effect. An IFN-γ enzyme-linked immunospot of purified tumoral CD8+ T cells revealed a significant proportion of tumor-specific T cells. Finally, the sequential poly-ICLC therapy induced abscopal effects in two dual-tumor models. This study provides evidence that the sequential poly-ICLC therapy significantly increased infiltration of tumor-specific CD8+ T cells in the tumors and induced CD8+ T cell-dependent inhibition of tumor growth, as well as abscopal effects.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/patologia , Carboximetilcelulose Sódica , Linfócitos T CD8-Positivos , Neoplasias Hepáticas/terapia , Poli I-C , Polilisina , Vacinação
5.
Colloids Surf B Biointerfaces ; 220: 112923, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36252536

RESUMO

In recent years, nanoscience has attracted considerable attention in the field of biomedicine. This involves the use of engineered nanomaterials as vital platforms for targeted drug delivery, diagnosis, imaging, and observation of therapeutic efficiency. This study explored the preparation, characterization, and applications of doxorubicin-loaded magnetic rice husk ash-derived SBA-15 (MIO@RHAS15-DOX nanocomposites) for drug delivery and in vitro/in vivo efficiency in the treatment of liver cancer. The small-angle XRD patterns of the MIO@RHAS15 nanocomposites demonstrated a core diffraction peak at 0.94°, with two noticeable peaks at 1.6° and 1.8°, representing (100), (110), and (200) crystalline planes, respectively, thereby indicating the existence of a well-defined mesostructure. A sharp melting endothermic peak (Tm) at 79 °C was observed for MIO@RHAS15 nanocomposites. The DOX release from MIO@RHAS15 followed the Higuchi model with the best correlation coefficient R2 value of 0.9799. The in vitro studies indicated a concentration dependent anticancer efficiency, with high cancer cells inhibition for MIO@RHAS15-DOX than free DOX. At the highest concentration of DOX (120 µg/mL), there was less than 25% and 15% cell viability after 24 h and 48 h, respectively. The in vivo studies demonstrated that the tumor sizes after treatment with PBS, MIO@RHAS15, free DOX, and MIO@RHS15-DOX were 1081, 904, 143, and 167 mm3, respectively. The in vivo animal test results depicted that the MIO@RHAS15-DOX nanocomposites were able to inhibit liver tumors in all tested mice. Therefore, the prepared nanocomposites possess a great potential for drug delivery application towards cancer treatment, thereby overcoming the limitations of traditional chemotherapy.


Assuntos
Antineoplásicos , Neoplasias Hepáticas , Nanocompostos , Oryza , Camundongos , Animais , Doxorrubicina , Nanocompostos/química , Neoplasias Hepáticas/tratamento farmacológico , Fenômenos Magnéticos
6.
J Pathol ; 250(1): 55-66, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31579932

RESUMO

Peritoneal fibrosis remains a problem in kidney failure patients treated with peritoneal dialysis. Severe peritoneal fibrosis with encapsulation or encapsulating peritoneal sclerosis is devastating and life-threatening. Although submesothelial fibroblasts as the major precursor of scar-producing myofibroblasts in animal models and M2 macrophage (Mϕ)-derived chemokines in peritoneal effluents of patients before diagnosis of encapsulating peritoneal sclerosis have been identified, attenuation of peritoneal fibrosis is an unmet medical need partly because the mechanism for cross talk between Mϕs and fibroblasts remains unclear. We use a sodium hypochlorite-induced mouse model akin to clinical encapsulated peritoneal sclerosis to study how the peritoneal Mϕs activate fibroblasts and fibrosis. Sodium hypochlorite induces the disappearance of CD11bhigh F4/80high resident Mϕs but accumulation of CD11bint F4/80int inflammatory Mϕs (InfMϕs) through recruiting blood monocytes and activating local cell proliferation. InfMϕs switch to express chemokine (C-C motif) ligand 17 (CCL17), CCL22, and arginase-1 from day 2 after hypochlorite injury. More than 75% of InfMϕs undergo genetic recombination by Csf1r-driven Cre recombinase, providing the possibility to reduce myofibroblasts and fibrosis by diphtheria toxin-induced Mϕ ablation from day 2 after injury. Furthermore, administration of antibody against CCL17 can reduce Mϕs, myofibroblasts, fibrosis, and improve peritoneal function after injury. Mechanistically, CCL17 stimulates migration and collagen production of submesothelial fibroblasts in culture. By breeding mice that are induced to express red fluorescent protein in Mϕs and green fluorescence protein (GFP) in Col1a1-expressing cells, we confirmed that Mϕs do not produce collagen in peritoneum before and after injury. However, small numbers of fibrocytes are found in fibrotic peritoneum of chimeric mice with bone marrow from Col1a1-GFP reporter mice, but they do not contribute to myofibroblasts. These data demonstrate that InfMϕs switch to pro-fibrotic phenotype and activate peritoneal fibroblasts through CCL17 after injury. CCL17 blockade in patients with peritoneal fibrosis may provide a novel therapy. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Quimiocina CCL17/metabolismo , Fibroblastos/metabolismo , Mediadores da Inflamação/metabolismo , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Comunicação Parácrina , Fibrose Peritoneal/metabolismo , Peritônio/metabolismo , Animais , Proliferação de Células , Quimiocina CCL17/genética , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Fibroblastos/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Macrófagos Peritoneais/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/genética , Fibrose Peritoneal/patologia , Peritônio/patologia , Fenótipo , Regiões Promotoras Genéticas , Transdução de Sinais , Hipoclorito de Sódio
7.
Am J Cancer Res ; 8(9): 1739-1751, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30323967

RESUMO

Aberrant glycosylation affects the malignant progression of cancers. Here, we report that N-acetyl-galactosaminyltransferase 2 (GALNT2), an enzyme that initiates the mucin type-O glycosylation, suppresses malignant phenotypes in gastric adenocarcinoma (GCA) cells by modifying epidermal growth factor receptor (EGFR) activity. GALNT2 was knocked down using siRNA in AGS and MKN28 cells. The expression of phosphorylated EGFR (pEGFR), phosphorylated Akt (pAkt) and Tn antigen were detected by western blotting. Proliferation, migration and invasion of cells with/without GLANT2-knockdown were assessed. Expression of pEGFR in the resected gastric cancer tissue was analyzed by Immunohistochemical staining, and was correlated with clinicopathological factors. The results showed that GALNT2 knockdown enhanced phosphorylation of EGFR and decreased expression of the Tn antigen on EGFR. Inhibiting EGFR activity with Gefitinib decreased the migration/invasion abilities and reversed the increase pAkt caused by GALNT2 knockdown in GCA cells. The addition of MK2206 (Akt inhibitor) mitigated the migration and invasion abilities of the GALNT2-knockdown cells. Patients with increased expressions of pEGFR in their cancer tissues were associated more metastasis, advanced stage and recurrence after surgical resection. Our results indicate that GALNT2 suppresses the malignant potential of GCA cells through the EGFR-Akt signaling pathway. The significance of O-glycosylation in receptor tyrosine kinases activities and GCA progression deserve further studies.

8.
Antioxid Redox Signal ; 26(11): 583-597, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-27580417

RESUMO

AIMS: Ischemic postconditioning (iPoC) is known to mitigate ischemia-reperfusion (IR) injury of the liver, the mechanisms of which remain to be elucidated. This study explored the role of microRNA-183 (miR-183) in the protective mechanism of iPoC. RESULTS: Microarray analysis showed miR-183 was robustly expressed in rats' livers with iPoC. miR-183 repressed the mRNA expression of Apaf-1, which is an apoptosis promoting factor. Using an oxygen-glucose deprivation (OGD) injury model in Clone 9 cells, hypoxic postconditioning (HPoC) and an miR-183 mimetic significantly decreased cell death after OGD, but miR-183 inhibitors eliminated the protection of HPoC. The increased expression of Apaf-1 and the downstream activation of capsase-3/9 after OGD were mitigated by HPoC or the addition of miR-183 mimetics, whereas miR-183 inhibitor diminished the effect of HPoC on Apaf-1-caspase signaling. In the in vivo experiment, iPoC and agomiR-183 decreased the expression of serum ALT after liver IR in the mice, but antagomiR-183 mitigated the effect of iPoC. The results of hematoxylin and eosin and TUNEL staining were compatible with the biochemical assay. Moreover, iPoC and agomiR-183 decreased the expression of Apaf-1 and 4-HNE after IR injury in mouse livers, whereas the antagomiR-mediated prevention of miR-183 expression led to increased protein expression of Apaf-1 and 4-HNE in the postischemic livers. INNOVATION: Our experiment showed the first time that miR-183 was induced in protective postconditioning and reduced reperfusion injury of the livers via the targeting of apoptotic signaling. CONCLUSION: miR-183 mediated the tolerance induced by iPoC in livers via Apaf-1 repressing. Antioxid. Redox Signal. 26, 583-597.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/genética , Pós-Condicionamento Isquêmico , Fígado/irrigação sanguínea , Fígado/metabolismo , MicroRNAs/genética , Interferência de RNA , Animais , Apoptose/genética , Expressão Gênica , Regulação da Expressão Gênica , Glucose/metabolismo , Fígado/patologia , Masculino , Camundongos , Estresse Oxidativo , Oxigênio/metabolismo , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais
9.
Oncotarget ; 7(10): 11251-62, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26848976

RESUMO

Glycosylation affects malignancy in cancer. Here, we report that N- acetylgalactosaminyltransferase 2 (GALNT2), an enzyme that mediates the initial step of mucin type-O glycosylation, suppresses malignant phenotypes in gastric adenocarcinoma (GCA) by modifying MET (Hepatocyte growth factor receptor) activity. GALNT2 mRNA and protein were downregulated in GCAs, and this reduction was associated with more advanced disease stage and shorter recurrence-free survival. Suppressing GALNT2 expression in GCA cells increased cell growth, migration, and invasion in vitro, and tumor metastasis in vivo. GALNT2 knockdown enhanced phosphorylation of MET and decreased expression of the Tn antigen on MET. Inhibiting MET activity with PHA665752 decreased the malignant phenotypes caused by GALNT2 knockdown in GCA cells. Our results indicate that GALNT2 suppresses the malignant potential of GCA cells and provide novel insights into the significance of O-glycosylation in MET activity and GCA progression.


Assuntos
Adenocarcinoma/patologia , N-Acetilgalactosaminiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Neoplasias Gástricas/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Idoso , Animais , Feminino , Glicosilação , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Mucinas/metabolismo , Fosforilação , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidade , Polipeptídeo N-Acetilgalactosaminiltransferase
10.
Eur Surg Res ; 55(1-2): 24-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25766749

RESUMO

BACKGROUND: Peritoneal lavage after cancer surgery is performed to reduce microscopic residual tumors in the peritoneum. This study evaluated the effects and mechanism of dialysate lavage in reducing the peritoneal implantation of gastric cancer cells. METHODS: Gastric cancer cells (MKN45 or AGS) were cultured with 1.5% peritoneal dialysate (PD) or normal saline (NS) for 30 min. The in vitro cell susceptibility to dialysate, including cell proliferation, cell death, cleaved PARP expression, and mitochondrial membrane potential, was evaluated. A murine model for gastric cancer cell peritoneal seeding was established to test the effects of PD and NS lavage on animal survival and tumor growth. RESULTS: A significant decrease in cell proliferation in PD and NS (75.2 ± 0.1 vs. 12.4 ± 0.2% in MKN45, p = 0.009; 58.2 ± 0.01 vs. 28.0 ± 0.01% in AGS, p = 0.008), an increase in mitochondrial permeability transition (93.0 ± 2.6 vs. 18.0 ± 2.9% in MKN45, p = 0.021; 86.8 ± 4.6 vs. 47.7 ± 10.2% in AGS, p < 0.001), and an increase in the expression of cleaved PARP and increased death (25.6 ± 9.4 vs. 16.9 ± 5.3% in MKN45, p = 0.031; 39.5 ± 5.1 vs. 20.9 ± 3.9% in AGS, p = 0.008) were recorded for gastric cancer cells separately exposed to PD and NS. Twenty-four days after inoculating MKN45 cells (5 × 10(6)/0.1 ml) in the peritoneal cavity, the average number of seeded tumors was 67.3 ± 10.8, 92.3 ± 6.0, and 29.2 ± 16.7 (p = 0.032), and the total weight of tumors was 0.98 ± 0.21, 0.58 ± 0.12, and 0.31 ± 0.17 g (p = 0.008), respectively, for mice receiving sham operation, NS lavage, and PD lavage. The 45-day survival rate for the PD lavage group was 22% compared to 0% for the sham injection and NS lavage groups (p = 0.034). CONCLUSION: PD induced significant cytotoxicity in gastric cancer cells that was related to mitochondrial perturbation. The use of PD lavage was effective in reducing the peritoneal implantation of gastric cancers in a murine model.


Assuntos
Inoculação de Neoplasia , Neoplasias Experimentais/terapia , Lavagem Peritoneal , Neoplasias Peritoneais/prevenção & controle , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Potencial da Membrana Mitocondrial , Camundongos Endogâmicos BALB C , Camundongos Nus , Peritônio/patologia , Neoplasias Gástricas/patologia
11.
J Neurosci Res ; 93(1): 140-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25082329

RESUMO

Postconditioning mitigates ischemia-induced cellular damage via a modified reperfusion procedure. Mitochondrial permeability transition (MPT) is an important pathophysiological change in reperfusion injury. This study explores the role of MPT modulation underlying hypoxic postconditioning (HPoC) in PC12 cells and studies the neuroprotective effects of ischemic postconditioning (IPoC) on rats. Oxygen-glucose deprivation (OGD) was performed for 10 hr on PC12 cells. HPoC was induced by three cycles of 10-min reoxygenation/10-min rehypoxia after OGD. The MPT inhibitor N-methyl-4-isoleucine cyclosporine (NIM811) and the MPT inducer carboxyatractyloside (CATR) were administered to selective groups before OGD. Cellular death was evaluated by flow cytometry and Western blot analysis. JC-1 fluorescence signal was used to estimate the mitochondrial membrane potential (△Ψm ). Transient global cerebral ischemia (tGCI) was induced via the two-vessel occlusion and hypotension method in male Sprague Dawley rats. IPoC was induced by three cycles of 10-sec reperfusion/10-sec reocclusion after index ischemia. HPoC and NIM811 administration attenuated cell death, cytochrome c release, and caspase-3 activity and maintained △Ψm of PC12 cells after OGD. The addition of CATR negated the protection conferred by HPoC. IPoC reduced neuronal degeneration and cytochrome c release and cleaved caspase-9 expression of hippocampal CA1 neurons in rats after tGCI. HPoC protected PC12 cells against OGD by modulating the MPT. IPoC attenuated degeneration of hippocampal neurons after cerebral ischemia.


Assuntos
Glucose/metabolismo , Pós-Condicionamento Isquêmico , Oxigênio/metabolismo , Traumatismo por Reperfusão/patologia , Animais , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Citocromos c/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Fluoresceínas , Formazans , Hipocampo/patologia , Masculino , Potencial da Membrana Mitocondrial , Células PC12 , Ratos , Sais de Tetrazólio
12.
Oxid Med Cell Longev ; 2014: 642793, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24891925

RESUMO

Human peritoneal mesothelial cells (HPMCs) are a critical component of the peritoneal membrane and play a pivotal role in dialysis adequacy. Loss of HPMCs can contribute to complications in peritoneal dialysis. Compelling evidence has shown that high-dialysate glucose is a key factor causing functional changes and cell death in HPMCs. We investigated the mechanism of HPMC apoptosis induced by high-dialysate glucose, particularly the role of mitochondria in the maintenance of HPMCs. HPMCs were incubated at glucose concentrations of 5 mM, 84 mM, 138 mM, and 236 mM. Additionally, N-acetylcysteine (NAC) was used as an antioxidant to clarify the mechanism of high-dialysate-glucose-induced apoptosis. Exposing HPMCs to high-dialysate glucose resulted in substantial apoptosis with cytochrome c release, followed by caspase activation and poly(ADP-ribose) polymerase cleavage. High-dialysate glucose induced excessive reactive oxygen species production and lipid peroxidation as well as oxidative damage to DNA. Mitochondrial fragmentation, multiple mitochondrial DNA deletions, and dissipation of the mitochondrial membrane potential were also observed. The mitochondrial dysfunction and cell death were suppressed using NAC. These results indicated that mitochondrial dysfunction is one of the main causes of high-dialysate-glucose-induced HPMC apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/metabolismo , Glucose/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Acetilcisteína/farmacologia , Caspases/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Dano ao DNA/efeitos dos fármacos , Soluções para Diálise/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Peritônio/citologia , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo
13.
Shock ; 39(3): 304-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23364428

RESUMO

A recent study showed that the injection of mitochondria isolated from a nonischemic region mitigated myocardial injury. We tested the protective effects of infusing isolated mitochondria on the reperfusion injury in the liver of rats. A partial liver ischemia-reperfusion (I/R) model in male Wistar rats was used. At the 45th minute of liver ischemia, the recipient's spleen was infused with vehicle (I/R-vehicle group) or vehicle containing isolated mitochondria (7.7 × 10 ± 1.5 × 10/mL, I/R-mito group). After a 240-min reperfusion, the serum and livers were collected to assess tissue injury. Our results show that the elevation of serum alanine aminotransferase (414.3 ± 67.1 vs. 208.8 ± 30.2 U/L), the necrosis of hepatocytes on hematoxylin-eosin staining, increase in positive counts in TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining (59.5% ± 4.4% vs. 24.6% ± 9.1%), the expression of cytosolic cytochrome c, cleaved caspase 9, and 4-hydroxynonenal were all reduced in the I/R-mito group, compared with the I/R-vehicle group. The membrane potential of the isolated mitochondria measured by JC-1 fluorescence remained high, and the infused mitochondria were distributed in the liver parenchyma at 240 min after reperfusion. These results demonstrate that an intrasplenic infusion of viable mitochondria isolated from the donor before reperfusion significantly reduced I/R injury in the liver.


Assuntos
Fígado/irrigação sanguínea , Mitocôndrias Hepáticas/transplante , Traumatismo por Reperfusão/prevenção & controle , Alanina Transaminase/sangue , Animais , Apoptose/fisiologia , Caspase 9/metabolismo , Citocromos c/metabolismo , Hepatócitos/patologia , Fígado/patologia , Masculino , Necrose , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Baço
14.
ACS Appl Mater Interfaces ; 4(12): 6720-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23151216

RESUMO

A new core-shell nanostructure consisting of inorganic hydroxyapatite (HAP) nanoparticles as the core and organic alginate as the shell (denoted as HAP@Alg) was successfully synthesized by a pre-gel method and applied to pH-responsive drug delivery systems (DDS). HAP@Alg nanoparticles have the advantages of hydroxyapatite and alginate, where hydroxyapatite provides pH-responsive degradability, and alginate provides excellent biocompatibility and COOH functionality. Through the subsequent addition of CaCl(2) and phosphate solutions to the alginate solution, HAP@Alg nanoparticles with controllable particle sizes (ranging from 160 to 650 nm) were obtained, and their core-shell structure was confirmed through transmission electron microscopy (TEM) observation. Rhodamine 6G (R6G), a positively charged dye, was selected as a model drug for pH-sensitive DDS. R6G was encapsulated in the HAP/Alg nanoparticles upon synthesis, and its loading efficiency could reach up to approximately 63.0%. The in vitro release behavior of the loaded R6G at different pH values was systematically studied, and the results indicated that more R6G molecules were released at lower pH conditions. For example, after releasing for 8 h, the release amount of R6G at pH 2.0 was 2.53-fold the amount at pH 7.4. We attributed this pH-sensitive release behavior to the dissolution of the HAP core in acidic conditions. The results of the MTT assay and confocal laser scanning microscopy indicated that the HAP@Alg were successfully uptaken by liver cancer cells (HepG2) without apparent cytotoxicity. The synthesized HAP@Alg nanoparticles show great potential as drug nanovehicles with high biocompatibility, enhanced drug loading, and pH-responsive features for future intracellular DDS.

15.
Am J Nephrol ; 29(3): 192-202, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18791325

RESUMO

BACKGROUND/AIMS: High-glucose (HG)-induced mesangial apoptosis and fibrogenesis possibly involves reactive oxygen species (ROS) formation and activated mitochondrial stress. We investigated the therapeutic effect of the antioxidant N-acetylcysteine (NAC) on cellular apoptosis and matrix accumulation in HG-treated rat mesangial cells (RMCs). METHODS: RMCs were cultured in media containing 5 (control) or 35 mM (HG) glucose. Cellular apoptosis was assayed by TdT-mediated dUTP nick-end labeling staining. Collagen and transforming growth factor-1 gene expression were measured by reverse transcriptase-polymerase chain reaction or Northern blotting. Mitochondrial capacity and intracellular ROS generation was assayed by fluorescence microscopy and flow cytometry, respectively. Cellular ATP production and malondialdehyde (MDA) formation were determined by a luciferin-luciferase reaction and high-performance liquid chromatography, respectively. Cytochrome c release, caspase activation and poly(ADP)ribose polymerase cleavage were assayed by Western blotting. RESULTS: HG-treated RMCs displayed enhanced cellular apoptosis (65%) and collagen gene expression (1.8-fold increase); these reactions could be significantly suppressed by 1 mM NAC (p < 0.05). Intracellular ROS generation, production of ATP and MDA, and caspase-3, -8 and -9 activities were significantly increased in HG-treated RMCs, and were effectively attenuated by addition of NAC. CONCLUSION: It is concluded that NAC prevents HG-induced mesangial apoptosis and fibrogenesis pathways by the reduction of oxidative stress.


Assuntos
Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Colágeno/biossíntese , Sequestradores de Radicais Livres/farmacologia , Hiperglicemia/tratamento farmacológico , Células Mesangiais/efeitos dos fármacos , Acetilcisteína/uso terapêutico , Trifosfato de Adenosina/biossíntese , Animais , Caspases/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Sequestradores de Radicais Livres/uso terapêutico , Expressão Gênica/efeitos dos fármacos , Malondialdeído/metabolismo , Células Mesangiais/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA