Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Lab Invest ; 102(8): 846-858, 2022 08.
Article in English | MEDLINE | ID: mdl-35288653

ABSTRACT

Myeloid cell mediated mechanisms regulate synovial joint inflammation. IL-34, a macrophage (Mø) growth and differentiation molecule, is markedly expressed in neutrophil and Mø-rich arthritic synovium. IL-34 engages a newly identified independent receptor, protein-tyrosine phosphatase, receptor-type, zeta (PTPRZ), that we find is expressed by Mø. As IL-34 is prominent in rheumatoid arthritis, we probed for the IL-34 and PTPRZ-dependent myeloid cell mediated mechanisms central to arthritis using genetic deficient mice in K/BxN serum-transfer arthritis. Unanticipatedly, we now report that IL-34 and PTPRZ limited arthritis as intra-synovial pathology and bone erosion were more severe in IL-34 and PTPRZ KO mice during induced arthritis. We found that IL-34 and PTPRZ: (i) were elevated, bind, and induce downstream signaling within the synovium in arthritic mice and (ii) were upregulated in the serum and track with disease activity in rheumatoid arthritis patients. Mechanistically, IL-34 and PTPRZ skewed Mø toward a reparative phenotype, and enhanced Mø clearance of apoptotic neutrophils, thereby decreasing neutrophil recruitment and intra-synovial neutrophil extracellular traps. With fewer neutrophils and neutrophil extracellular traps in the synovium, destructive inflammation was restricted, and joint pathology and bone erosion diminished. These novel findings suggest that IL-34 and PTPRZ-dependent mechanisms in the inflamed synovium limit, rather than promote, inflammatory arthritis.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Interleukins , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Animals , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Carrier Proteins , Inflammation , Interleukins/metabolism , Mice , Mice, Knockout , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Synovial Membrane/metabolism
2.
J Am Soc Nephrol ; 27(4): 1015-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26260164

ABSTRACT

Nephrotoxicity due to drugs and environmental chemicals accounts for significant patient mortality and morbidity, but there is no high throughput in vitro method for predictive nephrotoxicity assessment. We show that primary human proximal tubular epithelial cells (HPTECs) possess characteristics of differentiated epithelial cells rendering them desirable to use in such in vitro systems. To identify a reliable biomarker of nephrotoxicity, we conducted multiplexed gene expression profiling of HPTECs after exposure to six different concentrations of nine human nephrotoxicants. Only overexpression of the gene encoding heme oxygenase-1 (HO-1) significantly correlated with increasing dose for six of the compounds, and significant HO-1 protein deregulation was confirmed with each of the nine nephrotoxicants. Translatability of HO-1 increase across species and platforms was demonstrated by computationally mining two large rat toxicogenomic databases for kidney tubular toxicity and by observing a significant increase in HO-1 after toxicity using an ex vivo three-dimensional microphysiologic system (kidney-on-a-chip). The predictive potential of HO-1 was tested using an additional panel of 39 mechanistically distinct nephrotoxic compounds. Although HO-1 performed better (area under the curve receiver-operator characteristic curve [AUC-ROC]=0.89) than traditional endpoints of cell viability (AUC-ROC for ATP=0.78; AUC-ROC for cell count=0.88), the combination of HO-1 and cell count further improved the predictive ability (AUC-ROC=0.92). We also developed and optimized a homogenous time-resolved fluorescence assay to allow high throughput quantitative screening of nephrotoxic compounds using HO-1 as a sensitive biomarker. This cell-based approach may facilitate rapid assessment of potential nephrotoxic therapeutics and environmental chemicals.


Subject(s)
Heme Oxygenase-1/analysis , Kidney Diseases/chemically induced , Toxicity Tests , Biomarkers/analysis , Cells, Cultured , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Kidney Diseases/enzymology , Kidney Diseases/genetics , Kidney Tubules, Proximal/cytology , Toxicity Tests/methods
3.
J Am Soc Nephrol ; 25(1): 105-18, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24158981

ABSTRACT

Kidney injury molecule-1 (KIM-1)/T cell Ig and mucin domain-containing protein-1 (TIM-1) is upregulated more than other proteins after AKI, and it is highly expressed in renal damage of various etiologies. In this capacity, KIM-1/TIM-1 acts as a phosphatidylserine receptor on the surface of injured proximal tubular epithelial cells, mediating phagocytosis of apoptotic cells, and it may also act as a costimulatory molecule for immune cells. Despite recognition of KIM-1 as an important therapeutic target for kidney disease, the regulators of KIM-1 transcription in the kidney remain unknown. Using a bioinformatics approach, we identified upstream regulators of KIM-1 after AKI. In response to tubular injury in rat and human kidneys or oxidant stress in human proximal tubular epithelial cells (HPTECs), KIM-1 expression increased significantly in a manner that corresponded temporally and regionally with increased phosphorylation of checkpoint kinase 1 (Chk1) and STAT3. Both ischemic and oxidant stress resulted in a dramatic increase in reactive oxygen species that phosphorylated and activated Chk1, which subsequently bound to STAT3, phosphorylating it at S727. Furthermore, STAT3 bound to the KIM-1 promoter after ischemic and oxidant stress, and pharmacological or genetic induction of STAT3 in HPTECs increased KIM-1 mRNA and protein levels. Conversely, inhibition of STAT3 using siRNAs or dominant negative mutants reduced KIM-1 expression in a kidney cancer cell line (769-P) that expresses high basal levels of KIM-1. These observations highlight Chk1 and STAT3 as critical upstream regulators of KIM-1 expression after AKI and may suggest novel approaches for therapeutic intervention.


Subject(s)
Acute Kidney Injury/metabolism , Cell Adhesion Molecules/metabolism , Membrane Glycoproteins/metabolism , Protein Kinases/metabolism , Receptors, Virus/metabolism , STAT3 Transcription Factor/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/therapy , Animals , Cell Adhesion Molecules/genetics , Cell Line , Checkpoint Kinase 1 , Computational Biology , DNA Damage , Gene Expression Regulation , Hepatitis A Virus Cellular Receptor 1 , Humans , Kidney/metabolism , Male , Membrane Glycoproteins/genetics , Oxidative Stress , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Receptors, Virus/genetics , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics
4.
Blood ; 118(7): 1934-42, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21685370

ABSTRACT

Ischemia/reperfusion (I/R) injury in the kidney is a major cause of acute kidney injury (AKI) in humans and is associated with significantly high mortality. To identify genes that modulate kidney injury and repair, we conducted genome-wide expression analysis in the rat kidneys after I/R and found that the mRNA levels of fibrinogen (Fg)α, Fgß, and Fgγ chains significantly increase in the kidney and remain elevated throughout the regeneration process. Cellular characterization of Fgα and Fgγ chain immunoreactive proteins shows a predominant expression in renal tubular cells and the localization of immunoreactive Fgß chain protein is primarily in the renal interstitium in healthy and regenerating kidney. We also show that urinary excretion of Fg is massively increased after kidney damage and is capable of distinguishing human patients with acute or chronic kidney injury (n = 25) from healthy volunteers (n = 25) with high sensitivity and specificity (area under the receiver operating characteristic of 0.98). Furthermore, we demonstrate that Fgß-derived Bß(15-42) peptide administration protects mice from I/R-induced kidney injury by aiding in epithelial cell proliferation and tissue repair. Given that kidney regeneration is a major determinant of outcome for patients with kidney damage, these results provide new opportunities for the use of Fg in diagnosis, prevention, and therapeutic interventions in kidney disease.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Fibrin Fibrinogen Degradation Products/therapeutic use , Peptide Fragments/therapeutic use , Reperfusion Injury/complications , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Aged , Amino Acid Sequence , Animals , Apoptosis/drug effects , Female , Fibrinogen/genetics , Fibrinogen/immunology , Fibrinogen/urine , Humans , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Rats, Wistar , Up-Regulation
5.
Int J Cancer ; 130(3): 497-508, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-21387314

ABSTRACT

Recent population-based epidemiological studies strongly hint towards a link between obesity and its occurrence as well as progression of several cancers including melanoma. Although effects of obesity on breast, colon and liver cancers have been extensively investigated, the links between obesity and melanoma remain largely unexplored. Present study aimed to understand the effect of high fat diet-induced weight gain on susceptibility of C57BL/6J mice to melanoma. For this, mice routinely were fed on high fat diet for 6 months (HFD mice). Subsequently, mouse melanoma cells were injected subcutaneously in control as well as HFD mice and followed for tumor initiation and progression. We provide strong evidence that diet-induced obesity leads to increased melanoma progression in male C57BL/6J mice. We observed that increased melanoma progression is associated with enhanced Cav-1 and FASN expression in tumors from HFD mice. Cav-1 and FASN are co-ordinately regulated and Cav-1 interacts with FASN in melanoma cells. Enhanced levels of Cav-1, FASN and pAkt control melanoma cell proliferation. Our study establishes a causative relationship between diet-induced obesity and melanoma progression as well as demonstrates that obesity affects important tumorigenic pathways in melanoma.


Subject(s)
Caveolin 1/metabolism , Diet, High-Fat , Disease Progression , Fatty Acid Synthases/metabolism , Melanoma/metabolism , Obesity/metabolism , Adipokines/blood , Animals , Caveolin 1/genetics , Cell Line, Tumor , Cell Proliferation , Fatty Acid Synthases/genetics , Gene Expression Regulation, Neoplastic , Lipids/blood , Male , Melanoma/genetics , Melanoma/mortality , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , Oncogene Protein v-akt/metabolism , Protein Binding , Weight Gain
6.
Genes Cancer ; 11(1-2): 66-82, 2020.
Article in English | MEDLINE | ID: mdl-32577158

ABSTRACT

BACKGROUNDS AND AIMS: Hepatocellular Carcinoma (HCC) is the leading cause of cancer-related mortality across the world. Non-viral etiological factors including obesity and metabolic syndrome have now become prevalent cause of hepatocellular carcinoma. Sonic Hedgehog (SHH) pathway is activated in hepatocellular carcinoma but its role in regulation of lipogenic molecules during the hepatocarcinogenesis is not known. The aim of present study is to explore the role of SHH pathway in fatty changes associated with hepatocarcinogenesis at different stages and to further correlate the expression of SHH with lipogenic pathways. RESULTS: Our results demonstrated significant increase in lipidosis and fibrosis in DEN+CCl4 treated animals. It was simultaneously associated with the enhanced expression level of SHH, E2F1, adiponectin, and lipogenic molecules in DEN+CCl4 treated animals. These results were also corroborated with the similar findings in higher stage patients' biospecimens. METHODS: N-Nitrosodiethylamine (DEN) and Carbon TetraChloride (CCl4) induced hepatocellular acrcinoma model in male Wistar rats were established to study the expression level of SHH pathway and associated fatty changes during different stages of hepatocarcinogenesis. The expression levels of SHH, E2F1, and lipogenic molecules were checked at different stages of hepatocellular carcinoma. These results were further compared with biospecimens of hepatocellular carcinoma patients of different stages. CONCLUSIONS: Our results revealed an unknown aspect of SHH pathway in hepatocarcinogenesis via its control over lipogenesis. It gives insight into the lipogenic properties of DEN+CCl4 induced rodent hepatocarcinogenesis model and how SHH pathway operate to arbitrate this response.

7.
Mol Cancer ; 8: 87, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19845939

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide that is particularly refractory to chemotherapy. Several studies have proposed combination chemotherapy regimen for HCC treatment. However, these therapies are not effective in regressing tumor and prolonging survival of patient's suffering from HCC. Therefore, the development of more effective therapeutic tools and new strategies for the treatment of HCC are urgently needed. Over the last decade much attention has been focused on "bystander effect" as a possible therapeutic strategy for the treatment of certain human tumors. Interest in this therapeutic approach originated from numerous reports describing the radiation induced bystander effect. However, the knowledge about chemotherapy induced bystander effect is still limited. Hence, chemotherapy induced bystander phenomenon in hepatoma cells was explored by utilizing Mitomycin C (MMC). RESULTS: MMC induced bystander killing was observed only in hepatoma cells and it did not occur in cervical cancer cells. MMC induced bystander killing was transferable via medium. It occurred in co-cultured cells indicating the involvement of secreted as well as membrane bound factors. FasL and TRAIL were detected in the conditioned medium from treated cells. In medium transfer experiment, pre-treatment with EDTA (a broad range protease inhibitor) diminished MMC induced bystander killing. Following drug exposure, expression of Fas and TRAIL receptors increased and treatment with neutralizing antibodies against FasL and TRAIL inhibited bystander killing. CONCLUSION: Our results highlight the therapeutic importance of MMC in the treatment of HCC and implicate role of membrane bound and secreted forms of FasL and TRAIL in MMC induced bystander killing.


Subject(s)
Bystander Effect/drug effects , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mitomycin/pharmacology , Models, Biological , Cell Count , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Coculture Techniques , Culture Media , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Edetic Acid/pharmacology , Female , Green Fluorescent Proteins/metabolism , Humans , Ligands , Receptors, Death Domain/metabolism , Tumor Stem Cell Assay , Uterine Cervical Neoplasms/pathology
8.
PLoS One ; 13(12): e0208194, 2018.
Article in English | MEDLINE | ID: mdl-30513115

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is leading cause of cancer-related mortality and is categorized among the most common malignancies around the world. It is a heterogeneous tumor, which shows significant degree of histopathological heterogeneity. Despite the apparent histopathological diversity there has been very little distinct correlation between histopathological features and molecular aberrations particularly when it comes to the expression level of Wnt and Hh pathway molecules. The role of Wnt and Hh pathways in relation to HCC behavior viz. histopathological heterogeneity and aggressiveness is not known. Determining the sequential molecular changes and associated histopathological characteristic during HCC initiation, promotion, and progression would probably lead to a better treatment and prognosis. METHODS: N-Nitrosodiethylamine (DEN) induced HCC model in male Wistar rats were established to study the expression level of Wnt and Hh pathway molecules during different stages of hepatocarcinogenesis. Their expression levels were checked at mRNA and protein levels at initiation, promotion, and progression stages of HCC. The expression levels of Wnt and Hh pathway molecules were correlated with biospecimens of HCC patients of different stages. RESULTS: In the present study we identified the comprehensive change in the expression pattern of Wnt and Hh pathway molecules in DEN induced rodent hepatocarcinogenesis model. Our results demonstrate that ß-catenin /CTNNB1 plays important role in tumor initiation and promotion by stimulating tumor cell proliferation. The activated Wnt signaling in early stage of HCC is associated with well-differentiated histological pattern. The Hh activity although activated during the initiation stage but is significantly increased during the early promotion stage of hepatocarcinogenesis. The increased activity of both Wnt & Hh pathways during promotion stage is associated with moderately-differentiated histological pattern and was simultaneously linked with an increased expression of MMP9. Furthermore, our data demonstrated that during the progression stage Wnt pathway is modestly down-regulated but the Hh pathway activity sustained which in turn is associated with aggressive and invasive phenotype and poorly-differentiated histopathology. CONCLUSION: Our data uncovers the grade related expression of Wnt and Hh pathway molecules and the potential utility of these molecular signatures in daily clinical practice is to decide best therapy according to patients characteristic. Additionally, our data offer insight into the interaction between Wnt and Hh pathways which triggers HCC development and progression.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hedgehog Proteins/metabolism , Liver Neoplasms, Experimental/pathology , Liver Neoplasms/pathology , Wnt Signaling Pathway , Adult , Aged , Aged, 80 and over , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/chemically induced , Diethylnitrosamine/toxicity , Female , Hedgehog Proteins/genetics , Humans , Liver Neoplasms, Experimental/chemically induced , Male , Middle Aged , Neoplasm Grading , RNA, Messenger/metabolism , Rats , Rats, Wistar , beta Catenin/genetics , beta Catenin/metabolism
9.
Nat Biomed Eng ; 2(8): 589-599, 2018 08.
Article in English | MEDLINE | ID: mdl-30956894

ABSTRACT

Effectively activating macrophages that can 'eat' cancer cells is challenging. In particular, cancer cells secrete macrophage colony stimulating factor (MCSF), which polarizes tumour-associated macrophages from an antitumour M1 phenotype to a pro-tumourigenic M2 phenotype. Also, cancer cells can express CD47, an 'eat me not' signal that ligates with the signal regulatory protein alpha (SIRPα) receptor on macrophages to prevent phagocytosis. Here, we show that a supramolecular assembly consisting of amphiphiles inhibiting the colony stimulating factor 1 receptor (CSF-1R) and displaying SIRPα-blocking antibodies with a drug-to-antibody ratio of 17,000 can disable both mechanisms. The supramolecule homes onto SIRPα on macrophages, blocking the CD47-SIRPα signalling axis while sustainedly inhibiting CSF-1R. The supramolecule enhances the M2-to-M1 repolarization within the tumour microenvironment, and significantly improves antitumour and antimetastatic efficacies in two aggressive animal models of melanoma and breast cancer, with respect to clinically available small-molecule and biologic inhibitors of CSF-1R signalling. Simultaneously blocking the CD47-SIRPα and MCSF-CSF-1R signalling axes may constitute a promising immunotherapy.


Subject(s)
Antineoplastic Agents , Macrophages/drug effects , Neoplasms/therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cells, Cultured , Drug Design , Female , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism
10.
FEBS Lett ; 581(2): 289-95, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17208232

ABSTRACT

In general, the activation of extracellular recognition kinase (ERK) cascade is implicated in exerting tumorigenic effects. Conversely, recent studies suggest that ERK activation may also have role in DNA-damage induced apoptosis [Wang, X., Martindale, J.L. and Holbrook, N.J. (2000) Requirement for ERK activation in cisplatin-induced apoptosis. J. Biol. Chem. 275, 39435-39443; Schweyer S., Soruri A., Meschter O., Heintze A., Zschunke F., Miosge N., Thelen P., Schlott T., Radzun H.J. and Fayyazi, A. (2004) Cisplatin-induced apoptosis in human malignant testicular germ cell lines depends on MEK/ERK activation. Br. J. Cancer 91, 589-598]. Here we observed an essential requirement of ERK activation in carboplatin (Carb) induced apoptosis in SiHa and CaSki cells. Under similar treatment conditions p53 was also involved in Carb induced apoptosis in these cells. Therefore, we investigated the relation between p53 and ERK in Carb induced apoptosis in these cells. Abrogation of p53 transactivation activity by pifithrin alpha or dominant-negative mutant of p53 resulted in decrease in activation of ERK in Carb treated cells. The present study for the first time proposes that p53 may act as one of the upstream regulators of ERK activation for the induction of apoptosis in Carb treated cervical cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carboplatin/pharmacology , Carcinoma/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/enzymology , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Transcriptional Activation
11.
J Biosci ; 42(1): 31-41, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28229963

ABSTRACT

The tumour suppressor gene p53 is mutated in approximately 50% of the human cancers. p53 is involved in genotoxic stress-induced cellular responses. The role of EGFR and ERK in DNA-damage-induced apoptosis is well known. We investigated the involvement of activation of ERK signalling as a consequence of non-functional p53, in sensitivity of cells to doxorubicin. We performed cell survival assays in cancer cell lines with varying p53 status: MCF-7 (wild-type p53, WTp53), MDA MB-468 (mutant p53, MUTp53), H1299 (absence of p53, NULLp53) and an isogenic cell line MCF-7As (WTp53 abrogated). Our results indicate that enhanced chemosensitivity of cells lacking wild-type p53 function is because of elevated levels of EGFR which activates ERK. Additionally, we noted that independent of p53 status, pERK contributes to doxorubicin-induced cell death.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Neoplasms/drug therapy , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Doxorubicin/administration & dosage , Humans , MAP Kinase Signaling System/genetics , MCF-7 Cells , Neoplasms/genetics , Neoplasms/pathology
12.
Stem Cell Res Ther ; 6: 233, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620570

ABSTRACT

The potential use of stem cells for cell-based tissue repair and regeneration offers alternative therapeutic strategies for various diseases. Adipose-derived stem cells (ADSCs) have emerged as a promising source of stem cells suitable for transplantation in regenerative medicine and wound repair. A recent publication in Stem Cell Research & Therapy by Zhang and colleagues reports a new finding about the anti-fibrosis role of ADSCs and conditioned media derived from them on hypertrophic scar formation in vivo.


Subject(s)
Adipocytes/cytology , Cicatrix, Hypertrophic/therapy , Ear/pathology , Stem Cells/cytology , Stem Cells/physiology , Animals
13.
PLoS One ; 7(9): e45628, 2012.
Article in English | MEDLINE | ID: mdl-23029147

ABSTRACT

Fibrinogen (Fg) has been recognized to play a central role in coagulation, inflammation and tissue regeneration. Several studies have used Fg deficient mice (Fg(-/-)) in comparison with heterozygous mice (Fg(+/-)) to point the proinflammatory role of Fg in diverse pathological conditions and disease states. Although Fg(+/-) mice are considered 'normal', plasma Fg is reduced to ~75% of the normal circulating levels present in wild type mice (Fg(+/+)). We report that this reduction in Fg protein production in the Fg(+/-) mice is enough to protect them from kidney ischemia reperfusion injury (IRI) as assessed by tubular injury, kidney dysfunction, necrosis, apoptosis and inflammatory immune cell infiltration. Mechanistically, we observed binding of Fg to ICAM-1 in kidney tissues of Fg(+/+) mice at 24 h following IRI as compared to a complete absence of binding observed in the Fg(+/-) and Fg(-/-) mice. Raf-1 and ERK were highly activated as evident by significantly higher phosphorylation in the Fg(+/+) kidneys at 24 h following IRI as compared to Fg(+/-) and Fg(-/-) mice kidneys. On the other hand Cyclin D1 and pRb, indicating higher cell proliferation, were significantly increased in the Fg(+/-) and Fg(-/-) as compared to Fg(+/+) kidneys. These data suggest that Fg heterozygosity allows maintenance of a critical balance of Fg that enables regression of initial injury and promotes faster resolution of kidney damage.


Subject(s)
Fibrinogen/genetics , Heterozygote , Kidney Diseases/genetics , Reperfusion Injury/genetics , Animals , Base Sequence , Blotting, Western , DNA Primers , Fluorescent Antibody Technique , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Knockout , Real-Time Polymerase Chain Reaction
14.
Dalton Trans ; 40(41): 10865-72, 2011 Nov 07.
Article in English | MEDLINE | ID: mdl-21709916

ABSTRACT

Metallo-organic compounds are interesting to study for their antitumor activity and related applications. This paper deals with the syntheses, characterization, structure determination of a copper complex of anthracenyl terpyridine (1) and its plasmid cleavage and cytotoxicity towards different cancer cell lines. The complex binds CT-DNA through partial intercalation mode. The plasmid cleavage studies carried out using pBR322 and pUC18 resulted in the formation of all the three forms of the plasmid DNA. Plasmid cleavage studies carried out with a non-redoxable Zn(2+) complex (2) supported the role of the redox activity of copper in 1. The complex 1 showed remarkable antiproliferative activity against cancer cell lines, viz., cervical (HeLa, SiHa, CaSki), breast (MCF-7), liver (HepG2) and lung (H1299). A considerable lowering was observed in the IC(50) values of HPV-infected (viz., HeLa, SiHa, CaSki) vs. non-HPV-infected cell lines (MCF-7, HepG2, H1299). Antiproliferative activity of 1 was found to be much higher than the carboplatin when treated with the same cell lines. Incubation of the cells with 1 results in granular structures only with the HPV-infected cells and not with others as studied by phase contrast and fluorescence microscopy. The lower IC(50) value observed in case of 1 with HPV-infected cell lines may be correlated with the involvement of HPV oncoprotein. The role of HPV has been further augmented by transfecting the MCF-7 cells (originally not possessing HPV copy) with e6 oncoprotein cDNA. To our knowledge this is the first copper complex that causes cell death by interacting with HPV oncoprotein followed by exhibition of remarkable antiproliferative activity.


Subject(s)
Coordination Complexes/chemical synthesis , Copper/chemistry , Plasmids/metabolism , Pyridines/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , DNA/metabolism , DNA Cleavage , Electrophoresis, Agar Gel , Humans , Microscopy, Fluorescence , Molecular Conformation , Neoplasms , Papillomaviridae/genetics , Transfection
15.
J Biosci ; 35(4): 525-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21289434

ABSTRACT

Insulin-stimulated translocation of glucose transporter 4 (GLUT4) to cell membrane leading to glucose uptake is the rate-limiting step in diabetes. It is also a defined target of antidiabetic drug research. Existing GLUT4 translocation assays are based on time-consuming immunoassays and are hampered by assay variability and low sensitivity. We describe a real-time, visual, cell-based qualitative GLUT4 translocation assay using CHO-HIRc-myc-GLUT4eGFP cells that stably express myc- and eGFP-tagged GLUT4 in addition to human insulin receptor (HIRc). GLUT4 translocation is visualized by live cell imaging based on GFP fluorescence by employing a cooled charge-coupled device camera attached to a fluorescent microscope. This video imaging method and further quantitative analysis of GLUT4 on the cell membrane provide rapid and foolproof visual evidence that this method is suitable for screening GLUT4 translocation modulators.


Subject(s)
Antigens, CD/metabolism , Glucose Transporter Type 4/metabolism , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis/methods , Androstadienes/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Cytochalasin B/pharmacology , Drug Evaluation, Preclinical/methods , Genistein/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Antagonists/pharmacology , Microscopy, Confocal/methods , Microscopy, Video/methods , Plant Extracts/pharmacology , Protein Transport , Trigonella , Wortmannin
16.
Curr Cancer Drug Targets ; 8(8): 741-52, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19075597

ABSTRACT

Neuronal cell specific cyclin-dependent kinase 5 (Cdk5) is a known regulator of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. We report that Cdk5 also plays an important role in the proliferation of breast cancer cells MCF-7 and MDA MB-231 and is functionally involved in chemosensitivity as well as in cell death pathways induced by anti-cancer drug carboplatin (Carb). Here, we demonstrate that carboplatin induced Cdk5 activation under positive regulation of ERK, promotes cell death in MCF-7 and MDA MB-231 cells. DNA-damage stress enhanced ERK activity utilizes Cdk5 as one of its downstream targets for the execution of death signal in carboplatin induced death in MCF-7 and MDA MB-231 cells. Additionally, present data clearly indicates that activated Cdk5 modulates p53 transactivation in MCF-7 cells. However, in p53 mutant MDA MB-231 cells, Cdk5 mediated cell death is likely to be p53 independent. Collectively, our findings not only draw attention to the extra-neuronal functions of Cdk5 but also propose Cdk5 as a novel and potential therapeutic target of chemotherapeutic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Carboplatin/pharmacology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation , Chloramphenicol O-Acetyltransferase/genetics , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoprecipitation , RNA, Small Interfering
17.
J Cell Biochem ; 100(5): 1191-9, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17063488

ABSTRACT

Gadd45alpha is shown to be induced by a wide spectrum of DNA-damaging agents and implicated in negative regulation of cell growth by causing G2-M arrest or induction of apoptosis. In the present study, we explored the involvement of p53 in the promoter activation of Gadd45alpha as well as the role of Gadd45alpha in carboplatin (Carb) or 5-fluorouracil (5-FU)-induced apoptosis in human papillomavirus virus (HPV)-positive HEp-2 and HeLa cells. We report that Carb or 5-FU upregulate Gadd45alpha and p53 in both these cells. Transient transfection of chloramphenicol acetyl transferase (CAT)-reporter construct driven by Gadd45alpha promoter clearly indicated that Gadd45alpha upregulation was mediated through activation of its promoter. Inhibition of p53 function by dominant-negative-p53 expression partially suppressed the activation of Gadd45alpha promoter. Further, the induction of apoptosis was assessed by detection of poly (ADP-ribose) polymerase (PARP) cleavage by Western blot analysis. Inhibition of upregulated Gadd45alpha expression by antisense expression vector did not modulate the Carb or 5-FU-induced apoptosis. Overall, we conclude that Gadd45alpha promoter activation partially depends on p53 function in HPV-positive cells. Moreover, Gadd45alpha protein does not modulate Carb or 5-FU-induced apoptosis in these cells.


Subject(s)
Apoptosis/drug effects , Carboplatin/pharmacology , Cell Cycle Proteins/metabolism , Fluorouracil/pharmacology , Nuclear Proteins/metabolism , Papillomaviridae , Papillomavirus Infections/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2 , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
18.
Toxicol Appl Pharmacol ; 216(2): 177-85, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16806341

ABSTRACT

The response rates of extensively used chemotherapeutic drugs, carboplatin (Carb) or 5-fluorouracil (5-FU) are relatively disappointing because of considerable side effects associated with their high-dose regimen. In the present study, we determined whether treatment with a cholesterol depleting agent, methyl-beta-cyclodextrin (MCD), enhances the weak efficacy of low doses of Carb or 5-FU in human breast cancer cells. Data demonstrate that pretreatment with MCD significantly potentiates the cytotoxic activity of Carb and 5-FU in both MCF-7 and MDA-MB-231. Furthermore, we explored the molecular basis of enhanced cytotoxicity, and our data revealed that low-dose treatment with these drugs in MCD pretreated cells exhibited significantly decreased Akt phosphorylation, NF-kappaB activity and down-regulation in expression of anti-apoptotic protein Bcl-2. In addition, MCD pretreated cells demonstrated an increased intracellular drug accumulation as compared to cells treated with drugs alone. Taken together, our data provide the basis for potential therapeutic application of MCD in combination with other conventional cytotoxic drugs to facilitate reduction of drug dosage that offers a better chemotherapeutic approach with low toxicity.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Carboplatin/pharmacology , Fluorouracil/pharmacology , beta-Cyclodextrins/pharmacology , Adjuvants, Pharmaceutic/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carboplatin/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Combinations , Drug Screening Assays, Antitumor , Female , Fluorouracil/pharmacokinetics , Humans , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , beta-Cyclodextrins/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL