Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Carcinogenesis ; 43(4): 360-370, 2022 05 19.
Article in English | MEDLINE | ID: mdl-34965300

ABSTRACT

Development of cancer, including renal cancer, is a major problem in immunosuppressed patients. The mTOR inhibitor Rapamycin (RAPA) is used as an immunosuppressive agent in patients with organ transplants and other immunological disorders; and it also has antitumorigenic potential. However, long-term use of RAPA causes reactivation of Akt, and ultimately leads to enhanced tumor growth. Honokiol (HNK) is a natural compound, which possesses both anti-inflammatory and antitumorigenic properties. In this study, we investigated the effect of a novel combination therapy using RAPA + HNK on allograft survival and post-transplantation renal tumor growth. We observed that it effectively modulated the expression of some key regulatory molecules (like Carabin, an endogenous Ras inhibitor; and Rubicon, a negative regulator of autophagy) that play important roles in tumor cell growth and survival. This combination induced toxic autophagy and apoptosis to promote cancer cell death; and was associated with a reduced expression of the tumor-promoting receptor tyrosine kinase AXL. Finally, we utilized a novel murine model to examine the effect of RAPA + HNK on post-transplantation renal tumor growth. The combination treatment prolonged the allograft survival and significantly inhibited post-transplantation tumor growth. It was associated with reduced tumor expression of Rubicon and the cytoprotective/antioxidant heme oxygenase-1 to overcome therapeutic resistance. It also downregulated the coinhibitory programmed death-1 ligand, which plays major role(s) in the immune escape of tumor cells. Together, this combination treatment has a great potential to restrict renal tumor growth in transplant recipients as well as other immunosuppressed patients.


Subject(s)
Kidney Neoplasms , Organ Transplantation , Animals , Apoptosis , Autophagy , Biphenyl Compounds , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins , Kidney Neoplasms/pathology , Lignans , Mice , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
3.
PLoS Pathog ; 12(4): e1005600, 2016 04.
Article in English | MEDLINE | ID: mdl-27128797

ABSTRACT

Type I (IFN-α/ß) and type III (IFN-λ) interferons (IFNs) exert shared antiviral activities through distinct receptors. However, their relative importance for antiviral protection of different organ systems against specific viruses remains to be fully explored. We used mouse strains deficient in type-specific IFN signaling, STAT1 and Rag2 to dissect distinct and overlapping contributions of type I and type III IFNs to protection against homologous murine (EW-RV strain) and heterologous (non-murine) simian (RRV strain) rotavirus infections in suckling mice. Experiments demonstrated that murine EW-RV is insensitive to the action of both types of IFNs, and that timely viral clearance depends upon adaptive immune responses. In contrast, both type I and type III IFNs can control replication of the heterologous simian RRV in the gastrointestinal (GI) tract, and they cooperate to limit extra-intestinal simian RRV replication. Surprisingly, intestinal epithelial cells were sensitive to both IFN types in neonatal mice, although their responsiveness to type I, but not type III IFNs, diminished in adult mice, revealing an unexpected age-dependent change in specific contribution of type I versus type III IFNs to antiviral defenses in the GI tract. Transcriptional analysis revealed that intestinal antiviral responses to RV are triggered through either type of IFN receptor, and are greatly diminished when receptors for both IFN types are lacking. These results also demonstrate a murine host-specific resistance to IFN-mediated antiviral effects by murine EW-RV, but the retention of host efficacy through the cooperative action by type I and type III IFNs in restricting heterologous simian RRV growth and systemic replication in suckling mice. Collectively, our findings revealed a well-orchestrated spatial and temporal tuning of innate antiviral responses in the intestinal tract where two types of IFNs through distinct patterns of their expression and distinct but overlapping sets of target cells coordinately regulate antiviral defenses against heterologous or homologous rotaviruses with substantially different effectiveness.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Intestines/immunology , Rotavirus Infections/immunology , Animals , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Rotavirus
5.
J Biol Chem ; 290(13): 8110-20, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25645920

ABSTRACT

The receptor tyrosine kinase c-Met is overexpressed in renal cancer cells and can play major role in the growth and survival of tumor. We investigated how the c-Met-mediated signaling through binding to its ligand hepatocyte growth factor (HGF) can modulate the apoptosis and immune escape mechanism(s) of renal cancer cells by the regulations of novel molecules heme oxygenase-1 (HO-1) and programmed death-1 ligand 1 (PD-L1). We found that HGF/c-Met-mediated signaling activated the Ras/Raf pathway and down-regulated cancer cell apoptosis; and it was associated with the overexpression of cytoprotective HO-1 and anti-apoptotic Bcl-2/Bcl-xL. c-Met-induced HO-1 overexpression was regulated at the transcriptional level. Next, we observed that c-Met induction markedly up-regulated the expression of the negative co-stimulatory molecule PD-L1, and this can be prevented following treatment of the cells with pharmacological inhibitors of c-Met. Interestingly, HGF/c-Met-mediated signaling could not induce PD-L1 at the optimum level when either Ras or HO-1 was knocked down. To study the functional significance of c-Met-induced PD-L1 expression, we performed a co-culture assay using mouse splenocytes (expressing PD-L1 receptor PD-1) and murine renal cancer cells (RENCA, expressing high PD-L1). We observed that the splenocyte-mediated apoptosis of cancer cells during co-culture was markedly increased in the presence of either c-Met inhibitor or PD-L1 neutralizing antibody. Finally, we found that both c-Met and PD-L1 are significantly up-regulated and co-localized in human renal cancer tissues. Together, our study suggests a novel mechanism(s) by which c-Met can promote increased survival of renal cancer cells through the regulation of HO-1 and PD-L1.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Renal Cell/enzymology , Heme Oxygenase-1/metabolism , Kidney Neoplasms/enzymology , Proto-Oncogene Proteins c-met/physiology , Animals , Apoptosis , B7-H1 Antigen/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Survival , Coculture Techniques , Gene Expression , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Heme Oxygenase-1/genetics , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Signal Transduction , Tumor Cells, Cultured , Tumor Escape , ras Proteins/metabolism
6.
J Biol Chem ; 289(6): 3126-37, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24366869

ABSTRACT

Chemokines and their receptors play diverse roles in regulating cancer growth and progression. The receptor CXCR3 can have two splice variants with opposite functions. CXCR3-A promotes cell growth, whereas CXCR3-B mediates growth-inhibitory signals. However, the negative signals through CXCR3-B in cancer cells are not well characterized. In this study, we found that CXCR3-B-mediated signaling in MCF-7 and T47D breast cancer cells induced apoptotic cell death. Signals through CXCR3-B decreased the levels of the antiapoptotic proteins Bcl-2 and Bcl-xL and increased the expression of apoptotic cleaved poly(ADP-ribose) polymerase. Along with up-regulation in apoptosis, CXCR3-B signals were associated with a decrease in cellular autophagy with reduced levels of the autophagic markers Beclin-1 and LC3B. Notably, CXCR3-B down-regulated the expression of the cytoprotective and antiapoptotic molecule heme oxygenase-1 (HO-1) at the transcriptional level. There was an increased nuclear localization of Bach-1 and nuclear export of Nrf2, which are important negative and positive transcription factors, respectively, for HO-1 expression. We also observed that CXCR3-B promoted the activation of p38 MAPK and the inhibition of ERK-1/2. CXCR3-B could not induce cancer cell apoptosis at the optimal level when we either inhibited p38 activity or knocked down Bach-1. Further, CXCR3-B-induced apoptosis was down-regulated when we overexpressed HO-1. Together, our data suggest that CXCR3-B mediates a growth-inhibitory signal in breast cancer cells through the modulations of nuclear translocation of Bach-1 and Nrf2 and down-regulation of HO-1. We suggest that the induction of CXCR3-B-mediated signaling can serve as a novel therapeutic approach where the goal is to promote tumor cell apoptosis.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , MAP Kinase Signaling System , NF-E2-Related Factor 2/metabolism , Receptors, CXCR3/metabolism , Active Transport, Cell Nucleus/genetics , Alternative Splicing/genetics , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Beclin-1 , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/pathology , Fanconi Anemia Complementation Group Proteins/genetics , Female , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-E2-Related Factor 2/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, CXCR3/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Redox Biol ; 68: 102945, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37898101

ABSTRACT

Receptor tyrosine kinase (RTK), c-Met, is overexpressed and hyper active in renal cell carcinoma (RCC). Most of the therapeutic agents mediate cancer cell death through increased oxidative stress. Induction of c-Met in renal cancer cells promotes the activation of redox-sensitive transcription factor Nrf2 and cytoprotective heme oxygenase-1 (HO-1), which can mediate therapeutic resistance against oxidative stress. c-Met/RTK inhibitor, Cabozantinib, has been approved for the treatment of advanced RCC. However, acquired drug resistance is a major hurdle in the clinical use of cabozantinib. Honokiol, a naturally occurring phenolic compound, has a great potential to downregulate c-Met-induced pathways. In this study, we found that a novel combination treatment with cabozantinib + Honokiol inhibits the growth of renal cancer cells in a synergistic manner through increased production of reactive oxygen species (ROS); and it significantly facilitates apoptosis-and autophagy-mediated cancer cell death. Activation of c-Met can induce Rubicon (a negative regulator of autophagy) and p62 (an autophagy adaptor protein), which can stabilize Nrf2. By utilizing OncoDB online database, we found a positive correlation among c-Met, Rubicon, p62 and Nrf2 in renal cancer. Interestingly, the combination treatment significantly downregulated Rubicon, p62 and Nrf2 in RCC cells. In a tumor xenograft model, this combination treatment markedly inhibited renal tumor growth in vivo; and it is associated with decreased expression of Rubicon, p62, HO-1 and vessel density in the tumor tissues. Together, cabozantinib + Honokiol combination can significantly inhibit c-Met-induced and Nrf2-mediated anti-oxidant pathway in renal cancer cells to promote increased oxidative stress and tumor cell death.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , NF-E2-Related Factor 2/metabolism , Carcinoma, Renal Cell/drug therapy , Signal Transduction , Oxidative Stress , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Reactive Oxygen Species/metabolism , Oxidation-Reduction , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism
8.
Clin Dev Immunol ; 2011: 349575, 2011.
Article in English | MEDLINE | ID: mdl-22190970

ABSTRACT

The discovery of the interferon-lambda (IFN-λ) family has considerably contributed to our understanding of the role of interferon not only in viral infections but also in cancer. IFN-λ proteins belong to the new type III IFN group. Type III IFN is structurally similar to type II IFN (IFN-γ) but functionally identical to type I IFN (IFN-α/ß). However, in contrast to type I or type II IFNs, the response to type III IFN is highly cell-type specific. Only epithelial-like cells and to a lesser extent some immune cells respond to IFN-λ. This particular pattern of response is controlled by the differential expression of the IFN-λ receptor, which, in contrast to IFN-α, should result in limited side effects in patients. Recently, we and other groups have shown in several animal models a potent antitumor role of IFN-λ that will open a new challenging era for the current IFN therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Interferons/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Drug Synergism , Humans , Immunotherapy , Interferon-alpha/metabolism , Interferon-alpha/therapeutic use , Interferons/metabolism , Neoplasms/therapy
9.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188559, 2021 08.
Article in English | MEDLINE | ID: mdl-33965513

ABSTRACT

Recent studies have established that tumors can reprogram the pathways involved in nutrient uptake and metabolism to withstand the altered biosynthetic, bioenergetics and redox requirements of cancer cells. This phenomenon is called metabolic reprogramming, which is promoted by the loss of tumor suppressor genes and activation of oncogenes. Because of alterations and perturbations in multiple metabolic pathways, renal cell carcinoma (RCC) is sometimes termed as a "metabolic disease". The majority of metabolic reprogramming in renal cancer is caused by the inactivation of von Hippel-Lindau (VHL) gene and activation of the Ras-PI3K-AKT-mTOR pathway. Hypoxia-inducible factor (HIF) and Myc are other important players in the metabolic reprogramming of RCC. All types of RCCs are associated with reprogramming of glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle. Metabolism of glutamine, tryptophan and arginine is also reprogrammed in renal cancer to favor tumor growth and oncogenesis. Together, understanding these modifications or reprogramming of the metabolic pathways in detail offer ample opportunities for the development of new therapeutic targets and strategies, discovery of biomarkers and identification of effective tumor detection methods.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cellular Reprogramming , Energy Metabolism , Kidney Neoplasms/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Signal Transduction
10.
Cancer Immunol Immunother ; 59(7): 1059-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20217081

ABSTRACT

Hepatocellular carcinoma (HCC) occurs most commonly secondary to cirrhosis due to chronic hepatitis C or B virus (HCV/HBV) infections. Type I interferon (IFN-alpha) treatment of chronic HCV/HBV infections reduces the incidence of HCC in cirrhotic patients. However, IFN-alpha toxicity limits its tolerability and efficacy highlighting a need for better therapeutic treatments. A recently discovered type III IFN (IFN-lambda) has been shown to possess antiviral properties against HCV and HBV in vitro. In phase I clinical trials, IFN-lambda treatment did not cause significant adverse reactions. Using a gene therapy approach, we compared the antitumor properties of IFN-alpha and IFN-lambda in a transplantable hepatoma model of HCC. BALB/c mice were inoculated with syngeneic BNL hepatoma cells, or BNL cells expressing IFN-lambda (BNL.IFN-lambda cells) or IFN-alpha (BNL.IFN-alpha cells). Despite the lack of antiproliferative activity of IFNs on BNL cells, both BNL.IFN-lambda and BNL.IFN-alpha cells displayed retarded growth kinetics in vivo. Depletion of NK cells from splenocytes inhibited splenocyte-mediated cytotoxicity, demonstrating that NK cells play a role in IFN-induced antitumor responses. However, isolated NK cells did not respond directly to IFN-lambda. There was also a marked NK cell infiltration in IFN-lambda producing tumors. In addition, IFN-lambda and, to a lesser extent, IFN-alpha enhanced immunocytotoxicity of splenocytes primed with irradiated BNL cells. Splenocyte cytotoxicity against BNL cells was dependent on IL-12 and IFN-gamma, and mediated by dendritic cells. In contrast to NK cells, isolated from spleen CD11c+ and mPDCA+ dendritic cells responded directly to IFN-lambda. The antitumor activities of IFN-lambda against hepatoma, in combination with HCV and HBV antiviral activities warrant further investigation into the clinical use of IFN-lambda to prevent HCC in HCV/HBV-infected cirrhotic patients, as well as to treat liver cancer.


Subject(s)
Cell Proliferation/drug effects , Interferon Type I/pharmacology , Interferons/pharmacology , Liver Neoplasms, Experimental/prevention & control , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Flow Cytometry , Immunohistochemistry , Interferon Type I/genetics , Interferons/classification , Interferons/genetics , Interleukin-12/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transfection
11.
PLoS One ; 15(12): e0243837, 2020.
Article in English | MEDLINE | ID: mdl-33332399

ABSTRACT

Renal Cell Carcinoma (RCC) often becomes resistant to targeted therapies, and in addition, dose-dependent toxicities limit the effectiveness of therapeutic agents. Therefore, identifying novel drug delivery approaches to achieve optimal dosing of therapeutic agents can be beneficial in managing toxicities and to attain optimal therapeutic effects. Previously, we have demonstrated that Honokiol, a natural compound with potent anti-tumorigenic and anti-inflammatory effects, can induce cancer cell apoptosis and inhibit the growth of renal tumors in vivo. In cancer treatment, implant-based drug delivery systems can be used for gradual and sustained delivery of therapeutic agents like Honokiol to minimize systemic toxicity. Electrospun polymeric fibrous scaffolds are ideal candidates to be used as drug implants due to their favorable morphological properties such as high surface to volume ratio, flexibility and ease of fabrication. In this study, we fabricated Honokiol-loaded Poly(lactide-co-glycolide) (PLGA) electrospun scaffolds; and evaluated their structural characterization and biological activity. Proton nuclear magnetic resonance data proved the existence of Honokiol in the drug loaded polymeric scaffolds. The release kinetics showed that only 24% of the loaded Honokiol were released in 24hr, suggesting that sustained delivery of Honokiol is feasible. We calculated the cumulative concentration of the Honokiol released from the scaffold in 24hr; and the extent of renal cancer cell apoptosis induced with the released Honokiol is similar to an equivalent concentration of direct application of Honokiol. Also, Honokiol-loaded scaffolds placed directly in renal cell culture inhibited renal cancer cell proliferation and migration. Together, we demonstrate that Honokiol delivered through electrospun PLGA-based scaffolds is effective in inhibiting the growth of renal cancer cells; and our data necessitates further in vivo studies to explore the potential of sustained release of therapeutic agents-loaded electrospun scaffolds in the treatment of RCC and other cancer types.


Subject(s)
Biphenyl Compounds/pharmacology , Kidney Neoplasms/pathology , Lignans/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Scaffolds/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Liberation , Humans
12.
Cancers (Basel) ; 12(7)2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32635337

ABSTRACT

The mTOR inhibitor Rapamycin has tumor inhibitory properties; and it is also used as an immunosuppressive agent after organ transplantation. However, prolonged Rapamycin treatment re-activates Akt and can promote cancer growth. Honokiol is a natural compound with both anti-tumorigenic and anti-inflammatory properties. Here, we assessed the anti-tumor effects of Rapamycin and Honokiol combination in renal cell carcinoma (RCC). Receptor tyrosine kinase c-Met-mediated signaling plays a major role in RCC growth. We observed that compared with Rapamycin alone, Rapamycin + Honokiol combination can effectively down-regulate c-Met-induced Akt phosphorylation in renal cancer cells; and it markedly inhibited Ras activation and cell proliferation and promoted G1 phase cell cycle arrest. The combination treatment significantly induced ROS generation and cancer cell apoptosis even when c-Met is activated. Importantly, Honokiol, but not Rapamycin, decreased c-Met-induced expression of the co-inhibitory molecule PD-L1, implied in the immune escape of renal cancer cells. In mouse renal cancer cells and Balb/c splenocytes co-culture assay, Rapamycin + Honokiol markedly potentiated immune-cell-mediated killing of cancer cells, possibly through the down-regulation of PD-L1. Together, Honokiol can effectively overcome the limitation of Rapamycin treatment alone; and the combination treatment can markedly restrict the growth of RCC, with particular importance to post-transplantation renal cancer.

13.
Clin Lab Med ; 39(1): 171-183, 2019 03.
Article in English | MEDLINE | ID: mdl-30709505

ABSTRACT

Immunosuppression is essential to prevent graft rejection. However, immunosuppression impairs the ability of the host immune system to control viral infection and decreases tumor immunosurveillance. Therefore, immunosuppression after organ transplantation is a major risk factor for posttransplantation cancer. Notably, recent reports suggest that immunosuppressive agents can activate tumorigenic pathways independent of the involvement of the host immune system. In this review, we focus on cell-intrinsic tumorigenic pathways directly activated by immunosuppressive agents and discuss the much-described infection- and immune-mediated mechanisms of cancer development in organ transplant recipients.


Subject(s)
Immunosuppression Therapy/adverse effects , Neoplasms/immunology , Organ Transplantation/adverse effects , Signal Transduction , Cytokines/metabolism , Cytokines/physiology , Humans , Neoplasms/metabolism , Receptors, Chemokine/metabolism , Receptors, Chemokine/physiology
14.
Oncogenesis ; 8(2): 7, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30647407

ABSTRACT

Any imbalance between reactive oxygen species (ROS) generation and the anti-oxidant capacity lead to cellular oxidative stress. Many chemotherapeutic agents mediate their cytotoxic functions through the generation of ROS. c-Met, a receptor tyrosine kinase, is over-expressed in renal cancer and plays very crucial role(s) in its growth and survival. Here, we show that c-Met activation protected renal cancer cells from ROS, oxidative stress and cytotoxicity induced by the anti-cancer agent sorafenib (used for renal cancer treatment); and it markedly attenuated sorafenib-induced DNA damage. Activated c-Met promoted the anti-apoptotic proteins (Bcl-2 and Bcl-xL) and inhibited apoptotic cleaved caspase-3. We found that the cytoprotective function of c-Met against sorafenib-induced ROS generation and apoptosis was mediated primarily through the activation of anti-oxidant Nrf2-HO-1. c-Met promoted the nuclear localization of Nrf2 and hindered its binding with the inhibitory protein Keap1. Silencing of Nrf2 attenuated the protective action of c-Met against sorafenib-induced oxidative stress. To evaluate the physiological significance of our findings, in a tumor xenograft model, we observed that a combination treatment with pharmacological inhibitors of c-Met and it's anti-oxidant downstream effecter HO-1 markedly reduced the growth of renal tumor in vivo; it increased the oxidative stress, DNA damage and apoptotic markers in the tumor xenografts, along with reduced tumor vessel density. Our observations indicate that the c-Met-Nrf2-HO-1 pathway plays a vital role in relieving ROS-mediated oxidative stress of renal tumors. Targeting this pathway can significantly increase the oxidative stress to promote apoptotic death of cancer cells.

15.
Br J Haematol ; 139(5): 645-57, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17941951

ABSTRACT

Mouse models are valuable tools in the study of human chronic lymphocytic leukaemia (CLL). The New Zealand Black (NZB) strain is a naturally occurring model of late-onset CLL characterized by B-cell hyperproliferation and autoimmunity early in life, followed by progression to CLL. Other genetically engineered models of CLL that have been developed include (NZB x NZW) F1 mice engineered to express IL5, mice expressing human TCL1A, and mice overexpressing both BCL2 and a tumour necrosis factor receptor-associated factor. The applicability to human CLL varies with each model, suggesting that CLL is a multifactorial disease. Our work with the de novo NZB model has revealed many similarities to the human situation, particularly familial CLL. In NZB, the malignant clones express CD5, zap-70, and have chromosomal instability and germline Ig sequence. We also identified a point mutation in the 3'-flanking sequence of Mirn16-1, which resulted in decreased levels of the microRNA, miR-16 in lymphoid tissue. Exogenous restoration of miR-16 to an NZB malignant B-1 cell line resulted in cell cycle alterations, suggesting that the altered expression of Mirn15a/16-1 is an important molecular lesion in CLL. Future studies utilizing the NZB mouse could ascertain the role of environmental triggers, such as low dose radiation and organic chemicals in the augmentation of a pre-existing propensity to develop CLL.


Subject(s)
Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , MicroRNAs/genetics , RNA, Neoplasm/genetics , Animals , Base Sequence , Mice , Mice, Inbred NZB , Mice, Transgenic , Molecular Sequence Data , Point Mutation
16.
Sci Rep ; 7(1): 5900, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724911

ABSTRACT

Honokiol (HNK) is a small molecule with potent anti-inflammatory and anti-tumorigenic properties; yet the molecular targets of HNK are not well studied. Hyperactivation of the receptor tyrosine kinase c-Met and overexpression of the cytoprotective enzyme heme oxygenase-1 (HO-1) play a critical role in the growth and progression of renal cell carcinoma (RCC). Interestingly, the calcineurin inhibitor (CNI) cyclosporine A (CsA), an immunosuppressant used to prevent allograft rejection, can also increase the risk of RCC in transplant patients. We studied the potential role of c-Met signaling axis on CNI-induced renal tumor growth and tested the anti-tumor efficacy of HNK. Importantly, CNI treatment promoted c-Met induction and enhanced c-Met-induced Ras activation. We found that HNK treatment effectively down-regulated both c-Met phosphorylation and Ras activation in renal cancer cells. It inhibited the expression of both c-Met- and CNI-induced HO-1, and promoted cancer cell apoptosis. In vivo, HNK markedly inhibited CNI-induced renal tumor growth; and it decreased the expression of phospho-c-Met and HO-1 and reduced blood vessel density in tumor tissues. Our results suggest a novel mechanism(s) by which HNK exerts its anti-tumor activity through the inhibition of c-Met-Ras-HO-1 axis; and it can have significant therapeutic potential to prevent post-transplantation cancer in immunosuppressed patients.


Subject(s)
Biphenyl Compounds/pharmacology , Calcineurin Inhibitors/pharmacology , Heme Oxygenase-1/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lignans/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis/drug effects , Biphenyl Compounds/therapeutic use , Calcineurin Inhibitors/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Hepatocyte Growth Factor/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kidney Neoplasms/blood supply , Lignans/therapeutic use , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , ras Proteins/metabolism
17.
Mol Cancer Res ; 13(1): 50-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25185584

ABSTRACT

UNLABELLED: Tuberous sclerosis complex (TSC) is an autosomal dominant syndrome associated with tumors of the brain, heart, kidney, and lung. The TSC protein complex inhibits the mammalian or mechanistic target of rapamycin complex 1 (mTORC1). Inhibitors of mTORC1, including rapamycin, induce a cytostatic response in TSC tumors, resulting in temporary disease stabilization and prompt regrowth when treatment is stopped. The lack of TSC-specific cytotoxic therapies represents an important unmet clinical need. Using a high-throughput chemical screen in TSC2-deficient, patient-derived cells, we identified a series of molecules antagonized by rapamycin and therefore selective for cells with mTORC1 hyperactivity. In particular, the cell-permeable alkaloid chelerythrine induced reactive oxygen species (ROS) and depleted glutathione (GSH) selectively in TSC2-null cells based on metabolic profiling. N-acetylcysteine or GSH cotreatment protected TSC2-null cells from chelerythrine's effects, indicating that chelerythrine-induced cell death is ROS dependent. Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. In vivo, chelerythrine inhibited the growth of TSC2-null xenograft tumors with no evidence of systemic toxicity with daily treatment over an extended period of time. This study reports the results of a bioactive compound screen and the identification of a potential lead candidate that acts via a novel oxidative stress-dependent mechanism to selectively induce necroptosis in TSC2-deficient tumors. IMPLICATIONS: This study demonstrates that TSC2-deficient tumor cells are hypersensitive to oxidative stress-dependent cell death, and provide critical proof of concept that TSC2-deficient cells can be therapeutically targeted without the use of a rapalog to induce a cell death response.


Subject(s)
Benzophenanthridines/administration & dosage , Drug Screening Assays, Antitumor , Tuberous Sclerosis/drug therapy , Tumor Suppressor Proteins/genetics , Cell Death/drug effects , Cell Line, Tumor , Glutathione/genetics , Heme Oxygenase-1/genetics , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/complications , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/biosynthesis
19.
ASN Neuro ; 5(3): 183-93, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23763269

ABSTRACT

We recently established that the EGF-R (epidermal growth factor receptor) (EGF-R) is an essential regulator of the reactive expansion of SVZ (subventricular zone) NPs (neural precursors) that occurs during recovery from hypoxic-ischemic brain injury. The purpose of the current studies was to identify the conditions and the transcription factor (s) responsible for inducing the EGF-R. Here, we show that the increase in EGF-R expression and the more rapid division of the NPs can be recapitulated in in vitro by exposing SVZ NPs to hypoxia and hypoglycemia simultaneously, but not separately. The EGF-R promoter has binding sites for multiple transcription factors that includes the zinc finger transcription factor, Egr-1. We show that Egr-1 expression increases in NPs, but not astrocytes, following hypoxia and hypoglycemia where it accumulates in the nucleus. To determine whether Egr-1 is necessary for EGF-R expression, we used SiRNAs (small interfering RNA) specific for Egr-1 to decrease Egr-1 expression. Knocking-down Egr-1 decreased basal levels of EGF-R and it abolished the stress-induced increase in EGF-R expression. By contrast, HIF-1 accumulation did not contribute to EGF-R expression and FGF-2 only modestly induced EGF-R. These studies establish a new role for Egr-1 in regulating the expression of the mitogenic EGF-R. They also provide new information into mechanisms that promote NP expansion and provide insights into strategies for amplifying the numbers of stem cells for CNS (central nervous system) regeneration.


Subject(s)
Brain/pathology , Early Growth Response Protein 1/physiology , ErbB Receptors/physiology , Hypoglycemia/pathology , Hypoxia, Brain/pathology , Neural Stem Cells/physiology , Animals , Astrocytes/metabolism , Blotting, Western , Cell Nucleus/metabolism , Cell Nucleus/physiology , Immunohistochemistry , Polymerase Chain Reaction , Primary Cell Culture , RNA/biosynthesis , RNA/isolation & purification , Rats , Stress, Physiological , Transfection , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL