Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731829

ABSTRACT

Kidney ischemia and reperfusion injury (IRI) is a significant contributor to acute kidney injury (AKI), characterized by tubular injury and kidney dysfunction. Salvador family WW domain containing protein 1 (SAV1) is a key component of the Hippo pathway and plays a crucial role in the regulation of organ size and tissue regeneration. However, whether SAV1 plays a role in kidney IRI is not investigated. In this study, we investigated the role of SAV1 in kidney injury and regeneration following IRI. A proximal tubule-specific knockout of SAV1 in kidneys (SAV1ptKO) was generated, and wild-type and SAV1ptKO mice underwent kidney IRI or sham operation. Plasma creatinine and blood urea nitrogen were measured to assess kidney function. Histological studies, including periodic acid-Schiff staining and immunohistochemistry, were conducted to assess tubular injury, SAV1 expression, and cell proliferation. Western blot analysis was employed to assess the Hippo pathway-related and proliferation-related proteins. SAV1 exhibited faint expression in the proximal tubules and was predominantly expressed in the connecting tubule to the collecting duct. At 48 h after IRI, SAV1ptKO mice continued to exhibit severe kidney dysfunction, compared to attenuated kidney dysfunction in wild-type mice. Consistent with the functional data, severe tubular damage induced by kidney IRI in the cortex was significantly decreased in wild-type mice at 48 h after IRI but not in SAV1ptKO mice. Furthermore, 48 h after IRI, the number of Ki67-positive cells in the cortex was significantly higher in wild-type mice than SAV1ptKO mice. After IRI, activation and expression of Hippo pathway-related proteins were enhanced, with no significant differences observed between wild-type and SAV1ptKO mice. Notably, at 48 h after IRI, protein kinase B activation (AKT) was significantly enhanced in SAV1ptKO mice compared to wild-type mice. This study demonstrates that SAV1 deficiency in the kidney proximal tubule worsens the injury and delays kidney regeneration after IRI, potentially through the overactivation of AKT.


Subject(s)
Acute Kidney Injury , Cell Cycle Proteins , Kidney Tubules, Proximal , Mice, Knockout , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Male , Cell Proliferation , Signal Transduction , Hippo Signaling Pathway , Mice, Inbred C57BL , Disease Models, Animal
2.
Pharmacol Rep ; 74(5): 1041-1053, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35989399

ABSTRACT

BACKGROUND: Kidney ischemia reperfusion injury (IRI) is characterized by tubular cell death. DNA double-strand breaks is one of the major sources of tubular cell death induced by IRI. 2-Mercaptoethanol (2-ME) is protective against DNA double-strand breaks derived from calf thymus and bovine embryo. Here, we sought to determine whether treatment with 2-ME attenuated DNA double-strand breaks, resulting in reduced kidney dysfunction and structural damage in IRI. METHODS: Kidney IRI or sham-operation in mice was carried out. The mice were treated with 2-ME, Ras-selective lethal 3, or vehicle. Kidney function, tubular injury, DNA damage, antioxidant enzyme expression, and DNA damage response (DDR) kinases activation were assessed. RESULTS: Treatment with 2-ME significantly attenuated kidney dysfunction, tubular injury, and DNA double-strand breaks after IRI. Among DDR kinases, IRI induced phosphorylation of ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3 related (ATR), but IRI reduced phosphorylation of other DDR kinases including ataxia telangiectasia and Rad3 related, checkpoint kinase 1 (Chk1), Chk2, and Chinese hamster cells 1 (XRCC1). Treatment with 2-ME enhanced phosphorylation of ATM and ATM-mediated effector kinases in IRI-subjected kidneys, suggesting that 2-ME activates ATM-mediated DDR signaling pathway. Furthermore, 2-ME dramatically upregulated glutathione peroxidase 4 (GPX4) in IRI-subjected kidneys. Inhibition of GPX4 augmented adverse IRI consequences including kidney dysfunction, tubular injury, DNA double-strand breaks, and inactivation of ATM-mediated DDR signaling pathway after IRI in 2-ME-treated kidneys. CONCLUSIONS: We have demonstrated that exogenous 2-ME protects against DNA double-strand breaks after kidney IRI through GPX4 upregulation and ATM activation.


Subject(s)
Ataxia Telangiectasia , Reperfusion Injury , Cattle , Animals , Mice , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Mercaptoethanol/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation , Ataxia Telangiectasia/metabolism , Antioxidants/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , DNA Damage , Phosphorylation , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Kidney/metabolism , DNA/metabolism , Ischemia/metabolism , Cell Cycle Proteins/genetics
3.
Technol Cancer Res Treat ; 20: 15330338211038487, 2021.
Article in English | MEDLINE | ID: mdl-34490820

ABSTRACT

Objective: To investigate a feasible candidate for an appropriate cell line for the orthotopic renal cell carcinoma (RCC) model. Methods: Normal human proximal tubule cells (HK-2) and RCC cells were used for MTT assay, Western blotting, sphere-forming assay, and orthotopic injection of BALB/c-nude mice. Immunohistochemistry was adopted in tissue arrays and orthotopic tumors. Results: Primary RCC cells showed resistance to a GPX4 inhibitor compared to HK-2 and to metastatic RCC cells, Caki-1. Caki-2 and SNU-333 cells showed resistance to ferroptosis via increased GPX4 and FTH1, respectively. RCC cells showed increased αSMA, in which Caki-2 and SNU-333 cells exhibited different epithelial-mesenchymal transition and cancer stem cell markers. Caki-1 and SNU-333 cells formed spheres in vitro and orthotopic tumor masses in vivo. The injected SNU-333 tumor only showed high intensities of CD10 and PAX8, markers of renal origin. Conclusion: SNU-333 cell line exhibited resistance via iron metabolism and stemness, and had tumor-initiating capacities in vitro and in vivo. These results suggest that among the cells tested, SNU-333 cells were the most promising for the establishment of an orthotopic RCC model for further researches.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Kidney Neoplasms/pathology , Animals , Biomarkers, Tumor , Carcinoma, Renal Cell/drug therapy , Cell Survival , Ferroptosis/genetics , Humans , Immunohistochemistry , Kidney Neoplasms/drug therapy , Male , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Xenograft Model Antitumor Assays
4.
J Ethnopharmacol ; 280: 114412, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34265383

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Orostachys japonica A. Berger, also known as Wa-song in Korea, has traditionally been used as a folk medicine, but the potential anti-cancer effects of aqueous extract of Orostachys japonica (OJe) have not yet been thoroughly investigated. AIM OF THE STUDY: To evaluate the anti-cancer effects of OJe, its possible mechanisms of action were investigated in 5-fluorouracil (5-FU) resistant SNU-C5/5-FUR colorectal cancer cells. MATERIALS AND METHODS: The functional compounds of OJe were identified with high performance liquid chromatography. The anti-cancer effects of OJe in SNU-C5/5-FUR cells were investigated by a cell viability assays, flow cytometry analysis, and a subcutaneous xenograft model employing BALB/c-nude mice. Possible signalling pathways were assayed with Western blotting. RESULTS: OJe (250 µg/ml) showed anti-cancer effects in SNU-C5/5-FUR cells, that were mediated via apoptosis as well as cell cycle arrest at the G0/G1 phase. Gallic acid and (-)-epicatechin, the major functional components of OJe, induced cell cycle arrest. OJe treatment (250 mg/kg, p.o.) produced a significant anti-proliferative effect in the xenograft model via decreased ß-catenin/GSK3ß and increased p27 expression. OJe treatment significantly activated ERK and p38 both in vitro and in vivo. CONCLUSIONS: These results suggest that OJe has anti-proliferative effects on 5-FU-resistant colorectal cancer cells via regulation of MAPK signalling pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Crassulaceae/chemistry , Fluorouracil/pharmacology , MAP Kinase Signaling System/drug effects , Plant Extracts/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Mice , Mice, Nude , Plant Extracts/chemistry , Random Allocation , Xenograft Model Antitumor Assays
5.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R843-R854, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186196

ABSTRACT

Cisplatin is a well-known chemotherapy medication used to treat numerous cancers. However, treatment with cisplatin in cancer therapy has major side effects, such as nephrotoxic acute kidney injury. Adult vertebrate kidneys are commonly used as models of cisplatin-induced nephrotoxic acute kidney injury. Embryonic zebrafish kidney is more simplified and is composed simply of two nephrons and thus is an excellent model for the investigation of cisplatin nephrotoxicity. Here, we developed a novel model to induce cisplatin nephrotoxicity in adult zebrafish and demonstrated that intraperitoneal injection of cisplatin caused a decline in kidney proximal tubular function based on fluorescein-labeled dextran uptake and alkaline phosphatase staining. We also showed that cisplatin induced histological injury of the kidney tubules, quantified by tubular injury scores on the periodic acid-Schiff-stained kidney sections. As shown in a mouse model of cisplatin-induced nephrotoxicity, the activation of poly(ADP-ribose) polymerase (PARP), an enzyme implicated in cisplatin-induced cell death, was markedly increased after cisplatin injection in adult zebrafish. Furthermore, pharmacological inhibition of PARP using a specific PARP inhibitor PJ 34 hydrochloride (PJ34) or 3-aminobenzamide ameliorated kidney proximal tubular functional and histological damages in cisplatin-injected adult zebrafish kidneys. Administration of a combination of PARP inhibitors PJ34 and 3-aminobenzamide additively protected renal function and histology in zebrafish and mouse models of cisplatin nephrotoxicity. In conclusion, these data suggest that adult zebrafish are not only suitable for drug screening and genetic manipulation but also useful as a simplified but powerful model to study the pathophysiology of cisplatin nephrotoxicity and establish new therapies for treating human kidney diseases.


Subject(s)
Cisplatin , Kidney Diseases/enzymology , Kidney Tubules/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Zebrafish Proteins/metabolism , Animals , Benzamides/pharmacology , DNA Damage , Disease Models, Animal , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Signal Transduction , Zebrafish
6.
Ann Clin Lab Sci ; 50(1): 31-44, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32161010

ABSTRACT

The microbiome has recently become a key interest for cancer research. Anti-tumor effects of reinforced clostridium media (RCM) were investigated for all ingredients of RCM, which showed that yeast extract could be a candidate for this phenomenon. MTT assay, cell counting, cell death analysis, cell cycle analysis, and Western blotting were done on colorectal cancer cells with or without 5-fluorouracil resistance (SNU-C5 and SNU-C5/5-FUR). Yeast extract treatment showed dose- and time-dependent anti-tumor effects on SNU-C5 and SNU-C5/5-FUR. Anti-tumor effects were related to G0/G1 phase arrest with increased p21, reactive oxygen species scavenger activities, and decreased free iron. Yeast extract treatment significantly increased apoptosis, which was effectively blocked with the PARP inhibitor. Anti-tumor effects of yeast extract were correlated with the increased phosphorylation of p38 and p53. These results suggest that yeast extract might inhibit the proliferation of colorectal cancer cells via the activation of the p38-p53-p21 cascade.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Checkpoints , Colorectal Neoplasms/drug therapy , Complex Mixtures/pharmacology , Yeasts/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Humans , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Yeasts/physiology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Anat Cell Biol ; 52(3): 312-323, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31598361

ABSTRACT

Cyclosporin A (CsA) does not only exert a toxic effect on kidney parenchymal cells, but also protects them against necrotic cell death by inhibiting opening of mitochondrial permeability transition pore. However, whether CsA plays a role in hydrogen peroxide-induced kidney proximal tubular cell death is currently unclear. In the present study, treatment with CsA further increased apoptosis and necrosis in HK-2 human kidney proximal tubule epithelial cells during exposure to hydrogen peroxide. In addition, hydrogen peroxide-induced p53 activation and BH3 interacting-domain death agonist (BID) expression were higher in CsA-treated cells than those in non-treated cells, whereas hydrogen peroxide-induced activation of mitogen-activated protein kinases including p38, c-Jun N-terminal kinase, and extracellular signal-regulated kinase and activation of protein kinase B were not significantly altered by treatment with CsA. In oxidant-antioxidant system, reactive oxygen species (ROS) production induced by hydrogen peroxide was further enhanced by treatment with CsA. However, expression levels of antioxidant enzymes including manganese superoxide dismutase, copper/zinc superoxide dismutase, and catalase were not altered by treatment with hydrogen peroxide or CsA. Treatment with CsA further enhanced mitochondrial membrane potential induced by exposure to hydrogen peroxide, although it did not alter endoplasmic reticulum stress based on expression of glucose-regulated protein 78 and 94. Taken together, these data suggest that CsA can aggravate hydrogen peroxide-induced cell death through p53 activation, BID expression, and ROS production.

8.
Mol Med Rep ; 20(4): 3933-3941, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432187

ABSTRACT

The microbiome has recently attracted research interest in a variety of subjects, including cancer. In the present study, it was determined that reinforced clostridium media (RCM) for microbiome culture, exerts antitumor effects on renal cell carcinoma cells when compared to the microbiome 'X'. The antitumor effects of RCM were investigated for all ingredients of RCM, and the results revealed that yeast extract could be a candidate for the ingredient driving this phenomenon. Further experiments including MTT assay, cell counting, cell death analysis, cell cycle analysis and western blotting were conducted with yeast extract on renal cell carcinoma cells (Caki­1 and Caki­2) and normal human proximal tubular cells (HK­2). As a result, yeast extract exhibited dose­dependent antitumor effects on Caki­1 and Caki­2, but only slight effects on HK­2. In addition, yeast extract only exhibited slight effects on necrosis, autophagy, or apoptosis of Caki­1 and Caki­2. Yeast extract produced cell cycle arrest with an increased G0/G1 fraction and a decreased S fraction, and this was considered to be related to the decreased cyclin D1. Although yeast extract treatment increased anti­oxidant activities, the antitumor effects of yeast extract were also related to iron metabolism, based on the decreased transferrin receptor and increased ferritin. In addition, decreased GPX4 may be related to iron­dependent cell death, particularly in Caki­2. These results revealed that yeast extract may inhibit proliferation of renal cell carcinoma cells by regulating iron metabolism. Since an increased iron requirement is a classic phenomenon of cancer cells, yeast extract may be a candidate for adjuvant treatment of renal cell carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/therapy , Culture Media/pharmacology , Iron/metabolism , Kidney Neoplasms/therapy , Yeasts/chemistry , Antineoplastic Agents/chemistry , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/microbiology , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media/chemistry , Gastrointestinal Microbiome , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/microbiology , Yeasts/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...