Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Med ; 27(1): 28, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33765907

ABSTRACT

BACKGROUND: Glioblastoma is the most common primary brain tumor and remains uniformly fatal, highlighting the dire need for developing effective therapeutics. Significant intra- and inter-tumor heterogeneity and inadequate delivery of therapeutics across blood-brain barrier continue to be significant impediments towards developing therapies which can significantly enhance survival. We hypothesize that microRNAs have the potential to serve as effective therapeutics for glioblastoma as they modulate the activity of multiple signaling pathways, and hence can counteract heterogeneity if successfully delivered. METHODS: Using a computational approach, we identified microRNA-34a as a microRNA that maximally reduces the activation status of the three core signaling networks (the receptor tyrosine kinase, p53 and Rb networks) that have been found to be deregulated in most glioblastoma tumors. Glioblastoma cultures were transfected with microRNA-34a or control microRNA to assess biological function and therapeutic potential in vitro. Nanocells were derived from genetically modified bacteria and loaded with microRNA-34a for intravenous administration to orthotopic patient-derived glioblastoma xenografts in mice. RESULTS: Overexpression of microRNA-34a strongly reduced the activation status of the three core signaling networks. microRNA-34a transfection also inhibited the survival of multiple established glioblastoma cell lines, as well as primary patient-derived xenograft cultures representing the proneural, mesenchymal and classical subtypes. Transfection of microRNA-34a enhanced temozolomide (TMZ) response in in vitro cultures of glioblastoma cells with primary TMZ sensitivity, primary TMZ resistance and acquired TMZ resistance. Mechanistically, microRNA-34a downregulated multiple therapeutic resistance genes which are associated with worse survival in glioblastoma patients and are enriched in specific tumor spatial compartments. Importantly, intravenous administration of nanocells carrying miR-34a and targeted to epidermal growth factor receptor (EGFR) strongly enhanced TMZ sensitivity in an orthotopic patient-derived xenograft mouse model of glioblastoma. CONCLUSIONS: Targeted bacterially-derived nanocells are an effective vehicle for the delivery of microRNA-34a to glioblastoma tumors. microRNA-34a inhibits survival and strongly sensitizes a wide range of glioblastoma cell cultures to TMZ, suggesting that combination therapy of TMZ with microRNA-34a loaded nanocells may serve as a novel therapeutic approach for the treatment of glioblastoma tumors.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , MicroRNAs/administration & dosage , Nanostructures/administration & dosage , Temozolomide/therapeutic use , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Mice, Nude
2.
Am J Pathol ; 190(10): 2165-2176, 2020 10.
Article in English | MEDLINE | ID: mdl-32693062

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain cancer in adults. A hallmark of GBM is aggressive invasion of tumor cells into the surrounding normal brain. Both the current standard of care and targeted therapies have largely failed to specifically address this issue. Therefore, identifying key regulators of GBM cell migration and invasion is important. The leukemia-associated Rho guanine nucleotide exchange factor (LARG) has previously been implicated in cell invasion in other tumor types; however, its role in GBM pathobiology remains undefined. Herein, we report that the expression levels of LARG and ras homolog family members C (RhoC), and A (RhoA) increase with glial tumor grade and are highest in GBM. LARG and RhoC protein expression is more prominent in invading cells, whereas RhoA expression is largely restricted to cells in the tumor core. Knockdown of LARG by siRNA inhibits GBM cell migration in vitro and invasion ex vivo in organotypic brain slices. Moreover, siRNA-mediated silencing of RhoC suppresses GBM cell migration in vitro and invasion ex vivo, whereas depletion of RhoA enhances GBM cell migration and invasion, supporting a role for LARG and RhoC in GBM cell migration and invasion. Depletion of LARG increases the sensitivity of GBM cells to temozolomide treatment. Collectively, these results suggest that LARG and RhoC may represent unappreciated targets to inhibit glioma invasion.


Subject(s)
Cell Movement/physiology , Glioblastoma/metabolism , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Humans , Signal Transduction/physiology
3.
Neoplasia ; 20(10): 1045-1058, 2018 10.
Article in English | MEDLINE | ID: mdl-30219706

ABSTRACT

Glioblastoma multiforme (GBM) is the most common type of malignant brain tumors in adults and has a dismal prognosis. The highly aggressive invasion of malignant cells into the normal brain parenchyma renders complete surgical resection of GBM tumors impossible, increases resistance to therapeutic treatment, and leads to near-universal tumor recurrence. We have previously demonstrated that TROY (TNFRSF19) plays an important role in glioblastoma cell invasion and therapeutic resistance. However, the potential downstream effectors of TROY signaling have not been fully characterized. Here, we identified PDZ-RhoGEF as a binding partner for TROY that potentiated TROY-induced nuclear factor kappa B activation which is necessary for both cell invasion and survival. In addition, PDZ-RhoGEF also interacts with Pyk2, indicating that PDZ-RhoGEF is a component of a signalsome that includes TROY and Pyk2. PDZ-RhoGEF is overexpressed in glioblastoma tumors and stimulates glioma cell invasion via Rho activation. Increased PDZ-RhoGEF expression enhanced TROY-induced glioma cell migration. Conversely, silencing PDZ-RhoGEF expression inhibited TROY-induced glioma cell migration, increased sensitivity to temozolomide treatment, and extended survival of orthotopic xenograft mice. Furthermore, depletion of RhoC or RhoA inhibited TROY- and PDZ-RhoGEF-induced cell migration. Mechanistically, increased TROY expression stimulated Rho activation, and depletion of PDZ-RhoGEF expression reduced this activation. Taken together, these data suggest that PDZ-RhoGEF plays an important role in TROY signaling and provides insights into a potential node of vulnerability to limit GBM cell invasion and decrease therapeutic resistance.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Receptors, Tumor Necrosis Factor/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Female , Focal Adhesion Kinase 2/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Mice, Nude , Receptors, Tumor Necrosis Factor/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Temozolomide/pharmacology , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein/genetics , rhoC GTP-Binding Protein/metabolism
4.
Onco Targets Ther ; 10: 5633-5642, 2017.
Article in English | MEDLINE | ID: mdl-29200877

ABSTRACT

BACKGROUND: The cytotoxic effects of microtubule-targeting agents (MTAs) are often attributed to targeted effects on mitotic cells. In clinical practice, MTAs are combined with DNA-damaging agents such as ionizing radiation (IR) with the rationale that mitotic cells are highly sensitive to DNA damage. In contrast, recent studies suggest that MTAs synergize with IR by interfering with the trafficking of DNA damage response (DDR) proteins during interphase. These studies, however, have yet to demonstrate the functional consequences of interfering with interphase microtubules in the presence of IR. To address this, we combined IR with an established MTA, mebendazole (MBZ), to treat glioma cells exclusively during interphase. MATERIALS AND METHODS: To test whether MTAs can sensitize interphase cells to IR, we treated GL261 and GBM14 glioma cells with MBZ during 3-9 hours post IR (when the mitotic index was 0%). Cell viability was measured using a WST-1 assay, and radiosensitization was quantified using the dose enhancement factor (DEF). The effect of MBZ on the DDR was studied via Western blot analysis of H2AX phosphorylation. To examine the effects of MTAs on intracellular transport of DDR proteins, Nbs1 and Chk2, cytoplasmic and nuclear fractionation studies were conducted following treatment of glioma cells with MBZ. RESULTS: Treatment with MBZ sensitized interphase cells to the effects of IR, with a maximal DEF of 1.34 in GL261 cells and 1.69 in GBM14 cells. Treatment of interphase cells with MBZ led to more sustained γH2AX levels post IR, indicating a delay in the DDR. Exposure of glioma cells to MBZ resulted in a dose-dependent sequestration of Chk2 and Nbs1 in the cytoplasm. CONCLUSION: This study demonstrates that MBZ can sensitize cancer cells to IR independently of the induction of mitotic arrest. In addition, evidence is provided supporting the hypothesis that MTA-induced radiosensitization is mediated by inhibiting DDR protein accumulation into the nucleus.

6.
Mol Med ; 23: 50-56, 2017 04.
Article in English | MEDLINE | ID: mdl-28386621

ABSTRACT

The microtubule inhibitor vincristine is currently used to treat a variety of brain tumors, including low-grade glioma and anaplastic oligodendroglioma. Vincristine, however, does not penetrate well into brain tumor tissue, and moreover, it displays dose-limiting toxicities, including peripheral neuropathy. Mebendazole, a Food and Drug Administration-approved anthelmintic drug with a favorable safety profile, has recently been shown to display strong therapeutic efficacy in animal models of both glioma and medulloblastoma. Importantly, appropriate formulations of mebendazole yield therapeutically effective concentrations in the brain. Mebendazole has been shown to inhibit microtubule formation, but it is not known whether its potency against tumor cells is mediated by this inhibitory effect. To investigate this, we examined the effects of mebendazole on GL261 glioblastoma cell viability, microtubule polymerization and metaphase arrest, and found that the effective concentrations to inhibit these functions are very similar. In addition, using mebendazole as a seed for the National Cancer Institute (NCI) COMPARE program revealed that the top-scoring drugs were highly enriched in microtubule-targeting drugs. Taken together, these results indicate that the cell toxicity of mebendazole is indeed caused by inhibiting microtubule formation. We also compared the therapeutic efficacy of mebendazole and vincristine against GL261 orthotopic tumors. We found that mebendazole showed a significant increase in animal survival time, whereas vincristine, even at a dose close to its maximum tolerated dose, failed to show any efficacy. In conclusion, our results strongly support the clinical use of mebendazole as a replacement for vincristine for the treatment of brain tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Mebendazole/therapeutic use , Tubulin Modulators/therapeutic use , Vincristine/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Repositioning , Female , Humans , Hyperalgesia/chemically induced , Mebendazole/pharmacology , Mice, Inbred C57BL , Neurotoxicity Syndromes/etiology , Tubulin Modulators/pharmacology , Vincristine/pharmacology
7.
Int J Cancer ; 139(12): 2838-2849, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27543754

ABSTRACT

Glioblastoma (GBM) is one of the most pernicious forms of cancer and currently chances of survival from this malady are extremely low. We have used the noninvasive strategy of intranasal (IN) delivery of a glioblastoma-directed adduct of curcumin (CC), CC-CD68Ab, into the brain of mouse GBM GL261-implanted mice to study the effect of CC on tumor remission and on the phenotype of the tumor-associated microglial cells (TAMs). The treatment caused tumor remission in 50% of GL261-implanted GBM mice. A similar rescue rate was also achieved through intraperitoneal infusion of a lipid-encapsulated formulation of CC, Curcumin Phytosome, into the GL261-implanted GBM mice. Most strikingly, both forms of CC elicited a dramatic change in the tumor-associated Iba1+ TAMs, suppressing the tumor-promoting Arginase1high , iNOSlow M2-type TAM population while inducing the Arginase1low , iNOShigh M1-type tumoricidal microglia. Concomitantly, we observed a marked induction and activation of microglial NF-kB and STAT1, which are known to function in coordination to cause induction of iNOS. Therefore, our novel findings indicate that appropriately delivered CC can directly kill GBM cells and also repolarize the TAMs to the tumoricidal M1 state.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Glioblastoma/pathology , Microglia/drug effects , Microglia/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antineoplastic Agents/administration & dosage , Arginase/metabolism , Biomarkers , Calcium-Binding Proteins , Cell Line, Tumor , Curcumin/administration & dosage , DNA-Binding Proteins/metabolism , Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Immunophenotyping , Inhibitory Concentration 50 , Male , Mice , Microfilament Proteins , Microglia/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , STAT1 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 15(8): 1799-808, 2016 08.
Article in English | MEDLINE | ID: mdl-27207779

ABSTRACT

Medulloblastoma is a cerebellar tumor and the most common pediatric brain malignancy. Radiotherapy is part of the standard care for this tumor, but its effectiveness is accompanied by significant neurocognitive sequelae due to the deleterious effects of radiation on the developing brain. We have previously shown that the protein kinase MRK/ZAK protects tumor cells from radiation-induced cell death by regulating cell-cycle arrest after ionizing radiation. Here, we show that siRNA-mediated MRK depletion sensitizes medulloblastoma primary cells to radiation. We have, therefore, designed and tested a specific small molecule inhibitor of MRK, M443, which binds to MRK in an irreversible fashion and inhibits its activity. We found that M443 strongly radiosensitizes UW228 medulloblastoma cells as well as UI226 patient-derived primary cells, whereas it does not affect the response to radiation of normal brain cells. M443 also inhibits radiation-induced activation of both p38 and Chk2, two proteins that act downstream of MRK and are involved in DNA damage-induced cell-cycle arrest. Importantly, in an animal model of medulloblastoma that employs orthotopic implantation of primary patient-derived UI226 cells in nude mice, M443 in combination with radiation achieved a synergistic increase in survival. We hypothesize that combining radiotherapy with M443 will allow us to lower the radiation dose while maintaining therapeutic efficacy, thereby minimizing radiation-induced side effects. Mol Cancer Ther; 15(8); 1799-808. ©2016 AACR.


Subject(s)
Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Protein Kinases/metabolism , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/radiotherapy , DNA Damage , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , MAP Kinase Kinase Kinases , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Medulloblastoma/radiotherapy , Mice , Models, Molecular , Molecular Conformation , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protein Kinases/genetics , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Radiation, Ionizing , Radiation-Sensitizing Agents/chemistry , Signal Transduction , Xenograft Model Antitumor Assays
9.
Cancers (Basel) ; 8(4)2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27043632

ABSTRACT

Radiation therapy (RT) is frequently used as part of the standard of care treatment of the majority of brain tumors. The efficacy of RT is limited by radioresistance and by normal tissue radiation tolerance. This is highlighted in pediatric brain tumors where the use of radiation is limited by the excessive toxicity to the developing brain. For these reasons, radiosensitization of tumor cells would be beneficial. In this review, we focus on radioresistance mechanisms intrinsic to tumor cells. We also evaluate existing approaches to induce radiosensitization and explore future avenues of investigation.

10.
J Neurooncol ; 128(1): 175-182, 2016 05.
Article in English | MEDLINE | ID: mdl-26897014

ABSTRACT

The Brain Tumor Biotech Center at the Feinstein Institute for Medical Research, in collaboration with Voices Against Brain Cancer hosted The Brain Tumor Biotech Summit at in New York City in June 2015. The focus was once again on fostering collaboration between neuro-oncologist, neurosurgeons, scientists, leaders from biotechnology and pharmaceutical industries, and members of the financial community. The summit highlighted the recent advances in the treatment of brain tumor, and specifically focused on targeting of stem cells and EGFR, use of prophage and immunostimulatory vaccines, retroviral vectors for drug delivery, biologic prodrug, Cesium brachytherapy, and use of electric field to disrupt tumor cell proliferation. This article summarizes the current progress in brain tumor research as presented at 2015 The Brain Tumor Biotech Summit.


Subject(s)
Biotechnology , Brain Neoplasms/therapy , Animals , Congresses as Topic , Drug Industry , Humans
11.
PLoS One ; 9(5): e95885, 2014.
Article in English | MEDLINE | ID: mdl-24816734

ABSTRACT

Glioblastoma is the most malignant and lethal form of astrocytoma, with patients having a median survival time of approximately 15 months with current therapeutic modalities. It is therefore important to identify novel therapeutics. There is mounting evidence that microglia (specialized brain-resident macrophages) play a significant role in the development and progression of glioblastoma tumors. In this paper we show that microglia, in addition to stimulating glioblastoma cell invasion, also promote glioblastoma cell proliferation and resistance to ionizing radiation in vitro. We found that semapimod, a drug that selectively interferes with the function of macrophages and microglia, potently inhibits microglia-stimulated GL261 invasion, without affecting serum-stimulated glioblastoma cell invasion. Semapimod also inhibits microglia-stimulated resistance of glioblastoma cells to radiation, but has no significant effect on microglia-stimulated glioblastoma cell proliferation. We also found that intracranially administered semapimod strongly increases the survival of GL261 tumor-bearing animals in combination with radiation, but has no significant benefit in the absence of radiation. In conclusion, our observations indicate that semapimod sensitizes glioblastoma tumors to ionizing radiation by targeting microglia and/or infiltrating macrophages.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Hydrazones/pharmacology , Microglia/drug effects , Radiation, Ionizing , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Chemoradiotherapy , Glioblastoma/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Radiation-Sensitizing Agents/pharmacology , Survival Analysis , Tumor Burden/drug effects , Tumor Burden/radiation effects
12.
Mol Cancer Ther ; 13(1): 221-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24170769

ABSTRACT

Sorafenib is U.S. Food and Drug Adminstration-approved for the treatment of renal cell carcinoma and hepatocellular carcinoma and has been combined with numerous other targeted therapies and chemotherapies in the treatment of many cancers. Unfortunately, as with other RAF inhibitors, patients treated with sorafenib have a 5% to 10% rate of developing cutaneous squamous cell carcinoma (cSCC)/keratoacanthomas. Paradoxical activation of extracellular signal-regulated kinase (ERK) in BRAF wild-type cells has been implicated in RAF inhibitor-induced cSCC. Here, we report that sorafenib suppresses UV-induced apoptosis specifically by inhibiting c-jun-NH(2)-kinase (JNK) activation through the off-target inhibition of leucine zipper and sterile alpha motif-containing kinase (ZAK). Our results implicate suppression of JNK signaling, independent of the ERK pathway, as an additional mechanism of adverse effects of sorafenib. This has broad implications for combination therapies using sorafenib with other modalities that induce apoptosis.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/adverse effects , Protein Kinases/metabolism , Skin Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinases , MAP Kinase Signaling System/drug effects , Niacinamide/administration & dosage , Niacinamide/adverse effects , Phenylurea Compounds/administration & dosage , Protein Kinases/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sorafenib , raf Kinases/genetics , raf Kinases/metabolism
13.
Elife ; 2: e00969, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24192036

ABSTRACT

Vemurafenib and dabrafenib selectively inhibit the v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) kinase, resulting in high response rates and increased survival in melanoma. Approximately 22% of individuals treated with vemurafenib develop cutaneous squamous cell carcinoma (cSCC) during therapy. The prevailing explanation for this is drug-induced paradoxical ERK activation, resulting in hyperproliferation. Here we show an unexpected and novel effect of vemurafenib/PLX4720 in suppressing apoptosis through the inhibition of multiple off-target kinases upstream of c-Jun N-terminal kinase (JNK), principally ZAK. JNK signaling is suppressed in multiple contexts, including in cSCC of vemurafenib-treated patients, as well as in mice. Expression of a mutant ZAK that cannot be inhibited reverses the suppression of JNK activation and apoptosis. Our results implicate suppression of JNK-dependent apoptosis as a significant, independent mechanism that cooperates with paradoxical ERK activation to induce cSCC, suggesting broad implications for understanding toxicities associated with BRAF inhibitors and for their use in combination therapies. DOI: http://dx.doi.org/10.7554/eLife.00969.001.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Indoles/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects , Sulfonamides/pharmacology , Animals , Humans , MAP Kinase Kinase 4/metabolism , Mice , Mice, Hairless , Vemurafenib
14.
J Biol Chem ; 288(8): 5364-73, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23319595

ABSTRACT

The small GTPase RhoC is overexpressed in many invasive tumors and is essential for metastasis. Despite its high structural homology to RhoA, RhoC appears to perform functions that are different from those controlled by RhoA. The identity of the signaling components that are differentially regulated by these two GTPases is only beginning to emerge. Here, we show that the MAP3K protein MRK directly binds to the GTP-bound forms of both RhoA and RhoC in vitro. However, siRNA-mediated depletion of MRK in cells phenocopies depletion of RhoC, rather than that of RhoA. MRK depletion, like that of RhoC, inhibits LPA-stimulated cell invasion, while depletion of RhoA increases invasion. We also show that active MRK enhances LPA-stimulated invasion, further supporting a role for MRK in the regulation of invasion. Depletion of either RhoC or MRK causes sustained myosin light chain phosphorylation after LPA stimulation. In addition, activation of MRK causes a reduction in myosin light chain phosphorylation. In contrast, as expected, depletion of RhoA inhibits myosin light chain phosphorylation. We also present evidence that both RhoC and MRK are required for LPA-induced stimulation of the p38 and ERK MAP kinases. In conclusion, we have identified MRK as a novel RhoC effector that controls LPA-stimulated cell invasion at least in part by regulating myosin dynamics, ERK and p38.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lysophospholipids/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/physiology , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement , Collagen/chemistry , Drug Combinations , Female , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Humans , Laminin/chemistry , Models, Biological , Myosins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/chemistry , Proteoglycans/chemistry , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
15.
Cell Mol Neurobiol ; 32(7): 1199-208, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22552889

ABSTRACT

Ionizing radiation (IR) induces a DNA damage response that includes activation of cell cycle checkpoints, leading to cell cycle arrest. In addition, IR enhances cell invasiveness of glioblastoma cells, among other tumor cell types. Using RNA interference, we found that the protein kinase MRK, previously implicated in the DNA damage response to IR, also inhibits IR-induced cell migration and invasion of glioblastoma cells. We showed that MRK activation by IR requires the checkpoint protein Nbs1 and that Nbs1 is also required for IR-stimulated migration. In addition, we show that MRK acts upstream of Chk2 and that Chk2 is also required for IR-stimulated migration and invasion. Thus, we have identified Nbs1, MRK, and Chk2 as elements of a novel signaling pathway that mediates IR-stimulated cell migration and invasion. Interestingly, we found that inhibition of cell cycle progression, either with the CDK1/2 inhibitor CGP74514A or by downregulation of the CDC25A protein phosphatase, restores IR-induced migration and invasion in cells depleted of MRK or Chk2. These data indicate that cell cycle progression, at least in the context of IR, exerts a negative control on the invasive properties of glioblastoma cells and that checkpoint proteins mediate IR-induced invasive behavior by controlling cell cycle arrest.


Subject(s)
Cell Movement/radiation effects , DNA Damage/physiology , DNA Damage/radiation effects , Glioblastoma/pathology , Neoplasm Invasiveness/pathology , Radiation, Ionizing , Cell Line, Tumor , Cell Movement/physiology , Glioblastoma/genetics , Humans
16.
J Biol Chem ; 279(46): 47652-60, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15342622

ABSTRACT

DNA damage induced by ionizing radiation (IR) activates a complex cellular response that includes checkpoints leading to cell cycle arrest. The stress-activated mitogen-activated protein kinase (MAPK) p38gamma has been implicated in the G(2) phase checkpoint induced by IR. We recently discovered MRK as a member of the MAPK kinase kinase family that activates p38gamma. Here we investigated the role of MRK in the checkpoint response to IR. We identified autophosphorylation sites on MRK that are important for its kinase activity. A phosphospecific antibody that recognizes these sites showed that MRK is activated upon IR in a rapid and sustained manner. MRK depletion by RNA interference resulted in defective S and G(2) checkpoints induced by IR that were accompanied by reduced Chk2 phosphorylation and delayed Cdc25A degradation. We also showed that Chk2 is a substrate for MRK in vitro and is phosphorylated at Thr(68) by active MRK in cells. MRK depletion also increased sensitivity to the killing effects of IR. In addition, MRK depletion reduced IR-induced activation of p38gamma but had no effect on p38alpha activation, indicating that MRK is a specific activator of p38gamma after IR. Inhibition of p38gamma by RNA interference, however, did not impair IR-induced checkpoints. Thus, in response to IR MRK controls two independent pathways: the Chk2-Cdc25A pathway leading to cell cycle arrest and the p38gamma MAPK pathway.


Subject(s)
Cell Cycle/physiology , DNA Damage , Mitogen-Activated Protein Kinase 12/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , Checkpoint Kinase 2 , DNA-Binding Proteins , Enzyme Activation , Humans , MAP Kinase Kinase Kinases , Molecular Sequence Data , Phosphorylation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Radiation, Ionizing , Sequence Alignment , Tumor Suppressor Proteins , cdc25 Phosphatases/metabolism
17.
J Biol Chem ; 277(16): 13873-82, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11836244

ABSTRACT

Mitogen-activated protein (MAP) kinase pathways are three-kinase modules that mediate diverse cellular processes and have been highly conserved among eukaryotes. By using a functional complementation screen in yeast, we have identified a human MAP kinase kinase kinase (MAPKKK) that shares homology with members of the mixed lineage kinase (MLK) family and therefore was called MRK (MLK-related kinase). We report the structure of the MRK gene, from which are generated two splice forms of MRK, MRK-alpha and MRK-beta, encoding for proteins of 800 and 456 amino acids, respectively. By using a combination of solid phase protein kinase assays, transient transfections in cells, and analysis of endogenous proteins in stably transfected Madin-Darby canine kidney cells, we found that MRK-beta preferentially activates ERK6/p38gamma via MKK3/MKK6 and JNK through MKK4/MKK7. We also show that expression of wild type MRK increases the cell population in the G(2)/M phase of the cell cycle, whereas dominant negative MRK attenuates the G(2) arrest caused by gamma-radiation. In addition, exposure of cells to gamma-radiation induces MRK activity. These data suggest that MRK may mediate gamma-radiation signaling leading to cell cycle arrest and that MRK activity is necessary for the cell cycle checkpoint regulation in cells.


Subject(s)
Cell Cycle/radiation effects , Protein Serine-Threonine Kinases/physiology , Amino Acid Sequence , Animals , Blotting, Northern , COS Cells , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Dogs , Gamma Rays , Gene Library , Genes, Dominant , Humans , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 12 , Mitogen-Activated Protein Kinases/metabolism , Models, Genetic , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...