Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 11(1)2023 01.
Article in English | MEDLINE | ID: mdl-36653071

ABSTRACT

BACKGROUND: As a major driver of lymphocyte proliferation and activation interleukin 2 (IL-2) is a crucial mediator for antitumor responses. Despite promising activity in a subset of patients, wider therapeutic utility of IL-2 (aldesleukin) has been hampered by severe dose-limiting toxicities, the expansion of immunosuppressive regulatory T cells and a poor pharmacokinetic (PK) profile. Recent engineering efforts, including non-α IL-2 variants, have lowered the toxicity profile, but have yet to induce meaningful antitumor activity in a wider patient population. METHODS: We engineered INBRX-120, a CD8α-targeted Cisleukin™ molecule consisting of an affinity tuned IL-2 (IL2-x) connected to two high affinity CD8α-specific single domain antibodies via an effector-silenced Fc domain. To show that this large affinity differential enables directed IL-2 cis-signaling exclusively on CD8α-expressing tumoricidal effector cell populations, INBRX-120 effects on target cell expansion, activation and antitumor activity were tested in vitro. In vivo antitumor efficacy was evaluated in syngeneic mouse models alone or in combination with programmed cell death protein-1 (PD-1) blockade. Preclinical safety, as well as pharmacodynamic (PD) and PK profiling was carried out in non-human primates. RESULTS: INBRX-120 effectively expanded and enhanced the cytotoxic capacity of CD8 T cells and natural killer cells towards tumor cells without affecting regulatory T cells in vitro and in vivo. In syngeneic mouse models, INBRX-120 surrogate showed safe, potent, and durable antitumor efficacy alone and in combination with PD-1 blockade. In non-human primates, INBRX-120 expanded and activated CD8α-expressing effector cells, showed a favorable PK profile, and was well tolerated up to a dose of 1 mg/kg. CONCLUSIONS: Through its unique cis-signaling activity on CD8α-expressing effector cells, INBRX-120 overcomes the major limitations of IL-2-based therapy and effectively harnesses IL-2's potent intrinsic antitumor activity. This novel therapeutic strategy promises safer clinical activity that could induce meaningful antitumor efficacy in a wider set of patients with various cancer indications.


Subject(s)
Interleukin-2 , Neoplasms , Animals , Mice , Humans , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Programmed Cell Death 1 Receptor , Cytotoxicity, Immunologic , CD8-Positive T-Lymphocytes
2.
Elife ; 82019 09 03.
Article in English | MEDLINE | ID: mdl-31478830

ABSTRACT

Gene copy number alterations, tumor cell stemness, and the development of platinum chemotherapy resistance contribute to high-grade serous ovarian cancer (HGSOC) recurrence. Stem phenotypes involving Wnt-ß-catenin, aldehyde dehydrogenase activities, intrinsic platinum resistance, and tumorsphere formation are here associated with spontaneous gains in Kras, Myc and FAK (KMF) genes in a new aggressive murine model of ovarian cancer. Adhesion-independent FAK signaling sustained KMF and human tumorsphere proliferation as well as resistance to cisplatin cytotoxicity. Platinum-resistant tumorspheres can acquire a dependence on FAK for growth. Accordingly, increased FAK tyrosine phosphorylation was observed within HGSOC patient tumors surviving neo-adjuvant chemotherapy. Combining a FAK inhibitor with platinum overcame chemoresistance and triggered cell apoptosis. FAK transcriptomic analyses across knockout and reconstituted cells identified 135 targets, elevated in HGSOC, that were regulated by FAK activity and ß-catenin including Myc, pluripotency and DNA repair genes. These studies reveal an oncogenic FAK signaling role supporting chemoresistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Focal Adhesion Kinase 1/metabolism , Ovarian Neoplasms/drug therapy , Platinum/pharmacology , Animals , Cisplatin/pharmacology , Disease Models, Animal , Female , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Stem Cells
3.
Oncotarget ; 7(48): 79869-79884, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27829215

ABSTRACT

In glioblastoma (GBM), infiltration of primary tumor cells into the normal tissue and dispersal throughout the brain is a central challenge to successful treatment that remains unmet. Indeed, patients respond poorly to the current therapies of tumor resection followed by chemotherapy with radiotherapy and have only a 16-month median survival. It is therefore imperative to develop novel therapies. RSK2 is a kinase that regulates proliferation and adhesion and can promote metastasis. We demonstrate that active RSK2 regulates GBM cell adhesion and is essential for cell motility and invasion of patient-derived GBM neurospheres. RSK2 control of adhesion and migration is mediated in part by its effects on integrin-Filamin A complexes. Importantly, inhibition of RSK2 by either RSK inhibitors or shRNA silencing impairs invasion and combining RSK2 inhibitors with temozolomide improves efficacy in vitro. In agreement with the in vitro data, using public datasets, we find that RSK2 is significantly upregulated in vivo in human GBM patient tumors, and that high RSK2 expression significantly correlates with advanced tumor stage and poor patient survival. Together, our data provide strong evidence that RSK inhibitors could enhance the effectiveness of existing GBM treatment, and support RSK2 targeting as a promising approach for novel GBM therapy.


Subject(s)
Brain Neoplasms/pathology , Cell Adhesion/genetics , Cell Movement/genetics , Glioblastoma/pathology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Adult , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Glioblastoma/genetics , HEK293 Cells , Humans , Mice , Molecular Targeted Therapy , Neoplasm Invasiveness
5.
J Cell Sci ; 129(8): 1580-91, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26906414

ABSTRACT

Oxidized low-density lipoprotein (oxLDL) accumulates early in atherosclerosis and promotes endothelial nuclear factor κB (NF-κB) activation, proinflammatory gene expression and monocyte adhesion. Like for other atherogenic factors, oxLDL-induced proinflammatory responses requires integrin-dependent focal adhesion kinase (FAK, also known as PTK2) signaling; however, the mechanism by which FAK mediates oxLDL-dependent NF-κB signaling has yet to be revealed. We now show that oxLDL induces NF-κB activation and VCAM-1 expression through FAK-dependent IκB kinase ß (IKKß, also known as IKBKB) activation. We further identify FAK-dependent activation of p90 ribosomal S6 kinase family proteins (RSK) as a crucial mediator of oxLDL-dependent IKKß and NF-κB signaling, as inhibiting RSK blocks oxLDL-induced IKKß and NF-κB activation, VCAM-1 expression and monocyte adhesion. Finally, transgenic mice containing a kinase-dead mutation in FAK specifically in the endothelial cells show reduced RSK activity, decreased VCAM-1 expression and reduced macrophage accumulation in regions of early atherosclerosis. Taken together, our data elucidates a new mechanism whereby oxLDL-induced endothelial FAK signaling drives an ERK-RSK pathway to activate IKKß and NF-κB signaling and proinflammatory gene expression.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/physiology , Focal Adhesion Kinase 1/metabolism , I-kappa B Kinase/metabolism , Lipoproteins, LDL/metabolism , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Cell Adhesion , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/genetics
6.
J Nat Prod ; 78(3): 543-7, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25585025

ABSTRACT

Two new polycyclic alkaloids, neopetrocyclamines A and B (1 and 2), along with the known metabolites papuamine (3) and haliclonadiamine (4), were isolated from the Indonesian sponge Neopetrosia cf exigua. Neopetrocyclamine A contains a formamidinium moiety, a rare functional group. While these compounds share the same basic biosynthetic building blocks, the size of the ring system differs in 1 and 2 because of the formamidinium moiety. Biological evaluations of 1-4 revealed that papuamine is cytotoxic against glioblastoma SF-295 cells (GI50 = 0.8 µM).


Subject(s)
Alkaloids/isolation & purification , Antineoplastic Agents/isolation & purification , Diamines/isolation & purification , Polycyclic Compounds/isolation & purification , Porifera/chemistry , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Diamines/chemistry , Diamines/pharmacology , Drug Screening Assays, Antitumor , Humans , Indonesia , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Polycyclic Compounds/chemistry , Polycyclic Compounds/pharmacology
7.
Organometallics ; 33(22): 6669-6681, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25435644

ABSTRACT

Early-late transition metal TiAu2 compounds [(η-C5H5)2Ti{OC(O)CH2PPh2AuCl}2] (3) and new [(η-C5H5)2Ti{OC(O)-4-C6H4PPh2AuCl}2] (5) were evaluated as potential anticancer agents in vitro against renal and prostate cancer cell lines. The compounds were significantly more effective than monometallic titanocene dichloride and gold(I) [{HOC(O)RPPh2}AuCl] (R = -CH2- 6, -4-C6H4- 7) derivatives in renal cancer cell lines, indicating a synergistic effect of the resulting heterometallic species. The activity on renal cancer cell lines (for 5 in the nanomolar range) was considerably higher than that of cisplatin and highly active titanocene Y. Initial mechanistic studies in Caki-1 cells in vitro coupled with studies of their inhibitory properties on a panel of 35 kinases of oncological interest indicate that these compounds inhibit protein kinases of the AKT and MAPKAPK families with a higher selectivity toward MAPKAPK3 (IC503 = 91 nM, IC505 = 117 nM). The selectivity of the compounds in vitro against renal cancer cell lines when compared to a nontumorigenic human embryonic kidney cell line (HEK-293T) and the favorable preliminary toxicity profile on C57black6 mice indicate that these compounds (especially 5) are excellent candidates for further development as potential renal cancer chemotherapeutics.

8.
Nat Rev Cancer ; 14(9): 598-610, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25098269

ABSTRACT

Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. FAK promotes tumour progression and metastasis through effects on cancer cells, as well as stromal cells of the tumour microenvironment. The kinase-dependent and kinase-independent functions of FAK control cell movement, invasion, survival, gene expression and cancer stem cell self-renewal. Small molecule FAK inhibitors decrease tumour growth and metastasis in several preclinical models and have initial clinical activity in patients with limited adverse events. In this Review, we discuss FAK signalling effects on both tumour and stromal cell biology that provide rationale and support for future therapeutic opportunities.


Subject(s)
Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Stromal Cells/enzymology , Animals , Humans , Molecular Targeted Therapy/methods , Neoplasm Invasiveness , Neoplasms/pathology , Signal Transduction , Small Molecule Libraries/pharmacology , Tumor Microenvironment
9.
Mol Cancer Ther ; 13(8): 2050-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899686

ABSTRACT

Ovarian cancer ascites fluid contains matrix proteins that can impact tumor growth via integrin receptor binding. In human ovarian tumor tissue arrays, we find that activation of the cytoplasmic focal adhesion (FAK) tyrosine kinase parallels increased tumor stage, ß5 integrin, and osteopontin matrix staining. Elevated osteopontin, ß5 integrin, and FAK mRNA levels are associated with decreased serous ovarian cancer patient survival. FAK remains active within ovarian cancer cells grown as spheroids, and anchorage-independent growth analyses of seven ovarian carcinoma cell lines identified sensitive (HEY, OVCAR8) and resistant (SKOV3-IP, OVCAR10) cells to 0.1 µmol/L FAK inhibitor (VS-4718, formerly PND-1186) treatment. VS-4718 promoted HEY and OVCAR8 G0-G1 cell-cycle arrest followed by cell death, whereas growth of SKOV3-IP and OVCAR10 cells was resistant to 1.0 µmol/L VS-4718. In HEY cells, genetic or pharmacological FAK inhibition prevented tumor growth in mice with corresponding reductions in ß5 integrin and osteopontin expression. ß5 knockdown reduced HEY cell growth in soft agar, tumor growth in mice, and both FAK Y397 phosphorylation and osteopontin expression in spheroids. FAK inhibitor-resistant (SKOV3-IP, OVCAR10) cells exhibited anchorage-independent Akt S473 phosphorylation, and expression of membrane-targeted and active Akt in sensitive cells (HEY, OVCAR8) increased growth but did not create a FAK inhibitor-resistant phenotype. These results link osteopontin, ß5 integrin, and FAK in promoting ovarian tumor progression. ß5 integrin expression may serve as a biomarker for serous ovarian carcinoma cells that possess active FAK signaling.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Integrin beta Chains/metabolism , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Ovarian Neoplasms/metabolism , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Mice, Nude , Neoplasm Transplantation , Neoplasms, Cystic, Mucinous, and Serous/mortality , Osteopontin/metabolism , Ovarian Neoplasms/mortality , Signal Transduction
10.
Gynecol Oncol ; 134(1): 104-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24786638

ABSTRACT

OBJECTIVE: Focal adhesion kinase (FAK) is overexpressed in serous ovarian cancer. Loss of merlin, a product of the neurofibromatosis 2 tumor suppressor gene, is being evaluated as a biomarker for FAK inhibitor sensitivity in mesothelioma. Connections between merlin and FAK in ovarian cancer remain undefined. METHODS: Nine human and two murine ovarian cancer cell lines were analyzed for growth in the presence of a small molecule FAK inhibitor (PF-271, also termed VS-6062) from 0.1 to 1 µM for 72 h. Merlin was evaluated by immunoblotting and immunostaining of a human ovarian tumor tissue array. Growth of cells was analyzed in an orthotopic tumor model and evaluated in vitro after stable shRNA-mediated merlin knockdown. RESULTS: Greater than 50% inhibition of OVCAR8, HEY, and ID8-IP ovarian carcinoma cell growth occurred with 0.1 µM PF-271 in anchorage-independent (p<0.001) but not in adherent culture conditions. PF-271-mediated reduction in FAK Y397 phosphorylation occurred independently of growth inhibition. Suspended growth of OVCAR3, OVCAR10, IGROV1, IGROV1-IP, SKOV3, SKOV3-IP, A2780, and 5009-MOVCAR was not affected by 0.1 µM PF-271. Merlin expression did not correlate with serous ovarian tumor grade or stage. PF-271 (30 mg/kg, BID) did not inhibit 5009-MOVCAR tumor growth and merlin knockdown in SKOV3-IP and OVCAR10 cells did not alter suspended cell growth upon PF-271 addition. CONCLUSIONS: Differential responsiveness to FAK inhibitor treatment was observed. Intrinsic low merlin protein level correlated with PF-271-mediated anchorage-independent growth inhibition, but reduction in merlin expression did not induce sensitivity to FAK inhibition. Merlin levels may be useful for patient stratification in FAK inhibitor trials.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Neurofibromin 2/metabolism , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Humans , Mice , Neurofibromin 2/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/metabolism
11.
Cancer Res ; 73(20): 6099-105, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24097826

ABSTRACT

Metastasis, the spreading of cancer cells from a primary tumor to secondary sites throughout the body, is the primary cause of death for patients with cancer. New therapies that prevent invasion and metastasis in combination with current treatments could therefore significantly reduce cancer recurrence and morbidity. Metastasis is driven by altered signaling pathways that induce changes in cell-cell adhesion, the cytoskeleton, integrin function, protease expression, epithelial-to-mesenchymal transition and cell survival. The ribosomal S6 kinase (RSK) family of kinases is a group of extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) effectors that can regulate these steps of metastasis by phosphorylating both nuclear and cytoplasmic targets. However, our understanding of RSK function in metastasis remains incomplete and is complicated by the fact that the four RSK isoforms perform nonredundant, sometimes opposing functions. Although some isoforms promote cell motility and invasion by altering transcription and integrin activity, others impair cell motility and invasion through effects on the actin cytoskeleton. The mechanism of RSK action depends both on the isoform and the cancer type. However, despite the variance in RSK-mediated outcomes, chemical inhibition of this group of kinases has proven effective in blocking invasion and metastasis of several solid tumors in preclinical models. RSKs are therefore a promising drug target for antimetastatic cancer treatments that could supplement and improve current therapeutic approaches. This review highlights contradiction and agreement in the current data on the function of RSK isoforms in metastasis and suggests ways forward in developing RSK inhibitors as new antimetastasis drugs.


Subject(s)
Neoplasms/enzymology , Neoplasms/pathology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Humans , Isoenzymes , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction
12.
PLoS One ; 7(10): e48032, 2012.
Article in English | MEDLINE | ID: mdl-23144724

ABSTRACT

The number of renal cancers has increased over the last ten years and patient survival in advanced stages remains very poor. Therefore, new therapeutic approaches for renal cancer are essential. Englerin A is a natural product with a very potent and selective cytotoxicity against renal cancer cells. This makes it a promising drug candidate that may improve current treatment standards for patients with renal cancers in all stages. However, little is known about englerin A's mode of action in targeting specifically renal cancer cells. Our study is the first to investigate the biological mechanism of englerin A action in detail. We report that englerin A is specific for renal tumor cells and does not affect normal kidney cells. We find that englerin A treatment induces necrotic cell death in renal cancer cells but not in normal kidney cells. We further show that autophagic and pyroptotic proteins are unaffected by the compound and that necrotic signaling in these cells coincided with production of reactive oxygen species and calcium influx into the cytoplasm. As the first study to analyze the biological effects of englerin A, our work provides an important basis for the evaluation and validation of the compound's use as an anti-tumor drug. It also provides a context in which to identify the specific target or targets of englerin A in renal cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Necrosis , Sesquiterpenes, Guaiane/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Autophagy/drug effects , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chloroquine/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Molecular Structure , Reactive Oxygen Species/metabolism , Sesquiterpenes, Guaiane/chemistry , Staurosporine/pharmacology
13.
J Biol Chem ; 287(52): 43424-37, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23118220

ABSTRACT

Modulation of integrin activation is important in many cellular functions including adhesion, migration, and assembly of the extracellular matrix. RSK2 functions downstream of Ras/Raf and promotes tumor cell motility and metastasis. We therefore investigated whether RSK2 affects integrin function. We report that RSK2 mediates Ras/Raf inactivation of integrins. As a result, we find that RSK2 impairs cell adhesion and integrin-mediated matrix assembly and promotes cell motility. Active RSK2 appears to affect integrins by reducing actin stress fibers and disrupting focal adhesions. Moreover, RSK2 co-localizes with the integrin activator talin and is present at integrin cytoplasmic tails. It is thereby in a position to modulate integrin activation and integrin-mediated migration. Activation of RSK2 promotes filamin phosphorylation and binding to integrins. We also find that RSK2 is activated in response to integrin ligation to fibronectin. Thus, RSK2 could participate in a feedback loop controlling integrin function. These results reveal RSK2 as a key regulator of integrin activity and provide a novel mechanism by which it may promote cell migration and cancer metastasis.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , CHO Cells , Cell Adhesion/physiology , Contractile Proteins/genetics , Contractile Proteins/metabolism , Cricetinae , Cricetulus , Enzyme Activation/physiology , Filamins , HeLa Cells , Humans , Integrins/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/physiology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Talin/genetics , Talin/metabolism , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
14.
Int J Cancer ; 131(7): 1556-68, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22213050

ABSTRACT

ERK and RSK2 drive proliferation and invasion of many cancers. Phosphoprotein enriched in astrocytes 15 (PEA15) binds ERK and RSK2 and high PEA15 levels can impair ERK- and RSK2-dependent transcription. PEA15 expression also inversely correlates with cell motility and invasiveness. We therefore tested PEA15 effects on neuroblastoma cells in vitro. We further analyzed PEA15 expression in the context of clinical and genetic features of neuroblastoma in tumor samples to determine its correlation with disease progression. Affymetrix microarray analysis was performed using 24 different neuroblastoma cell lines. Cell lines expressing low to intermediate levels of PEA15 were chosen for in vitro functional studies. The cell line results were verified by Affymetrix analysis of three different neuroblastic tumor types (total of 110 samples) PEA15 overexpression inhibited neuroblastoma migration in vitro. We verified that inhibition of motility required PEA15 interaction with its binding partners ERK and RSK2. Additionally, synthetic inhibitors of RSK2 suppressed integrin-dependent migration. PEA15 expression correlates with clinical parameters and a 25% increase in patient survival rate. The highest PEA15 levels were found in low stage, more differentiated and less metastatic neuroblastic tumors, and correlated with lack of MYCN amplification. PEA15 blocks neuroblastoma migration through inhibition of ERK/RSK2 signaling. PEA15 expression levels correlate with favorable clinical features suggesting that PEA15 limits metastatic progression of neuroblastoma. Thus, PEA15 and its partners ERK and RSK2 are potential targets for the development of new therapeutics to impede progression of minimal residual disease in patients with high-risk neuroblastoma.


Subject(s)
Cell Movement/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neuroblastoma/diagnosis , Neuroblastoma/genetics , Phosphoproteins/genetics , Animals , Apoptosis Regulatory Proteins , COS Cells , Cell Adhesion/genetics , Cell Line, Tumor , Chlorocebus aethiops , Chromosome Aberrations , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Amplification , Humans , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , N-Myc Proto-Oncogene Protein , Neuroblastoma/mortality , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Phosphoproteins/metabolism , Prognosis , Protein Binding , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
15.
Mol Nutr Food Res ; 55(8): 1197-206, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21732533

ABSTRACT

SCOPE: trans-Resveratrol has been shown to improve insulin sensitivity and to enhance cellular glucose uptake. Evidence from recent studies indicates that these effects depend on SIRT1-pathways. METHODS AND RESULTS: Since ingestion of resveratrol leads to the presence of resveratrol and resveratrol metabolites in the body, we aimed at investigating (i) whether a daily dose of 300 mg resveratrol/kg body weight in healthy male Wistar rats for a period of 8 wk affects the selected parameters of glucose and lipid metabolism and (ii) whether the resulting plasma concentrations of resveratrol metabolites were effective in modulating SIRT1 expression. The dietary dose was based on the results from preceding toxicity studies. The results from the feeding experiment revealed plasma concentrations of resveratrol and its metabolites below 1 µmol/L and showed that fasting glucose and insulin levels were decreased by 35 and 41%, respectively, in the resveratrol group compared with controls. Insulin sensitivity was enhanced by 70%, whereas liver SIRT1 protein expression was not affected. Treatment of HepG2 cells with 10 µM resveratrol (1.49-fold) or its diglucuronides (1.21-fold) increased SIRT1 expression. CONCLUSION: These results suggest that the improved insulin sensitivity after dietary administration of 300 mg resveratrol/kg body weight does not involve increased protein expression of SIRT1.


Subject(s)
Insulin Resistance , Sirtuin 1/genetics , Stilbenes/pharmacology , Animals , Blood Glucose/analysis , Cholesterol/metabolism , Fatty Acids, Nonesterified/metabolism , Female , Glycated Hemoglobin/analysis , Hep G2 Cells , Humans , Insulin/blood , Liver/metabolism , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Resveratrol , Sirtuin 1/analysis , Stilbenes/metabolism , Stilbenes/toxicity , Triglycerides/metabolism
16.
J Org Chem ; 76(10): 3635-43, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21500817

ABSTRACT

An extensive study of the secondary metabolites produced by a new Sticta sp. of lichen has led to the isolation of three new compounds containing the 4-amino-3-hydroxy-5-phenylpentanoic acid residue (Ahppa). The structures of stictamides A-C (1-3) were assigned by 2D NMR spectroscopic and chemical methods. Due to extensive epimerization of the Ahppa residue observed after acid hydrolysis, the configuration of this unit was deduced through conversion of 1 to an appropriate derivative and application of our recently developed statine NMR database. Evaluation of stictamide A against a panel of disease-relevant proteases showed that it inhibited MMP12 at 2.3 µM and significantly reduced invasion in the human glioma cell line U87MG. Docking studies suggest that stictamide A inhibits MMP12 by a non-zinc-binding mechanism.


Subject(s)
Amino Acids/chemistry , Matrix Metalloproteinase Inhibitors , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Matrix Metalloproteinase 12/chemistry , Models, Molecular , Neoplasm Invasiveness , Protein Conformation
17.
J Biol Chem ; 286(16): 14713-23, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21383007

ABSTRACT

Loss of properly regulated cell death and cell survival pathways can contribute to the development of cancer and cancer metastasis. Cell survival signals are modulated by many different receptors, including integrins. Bit-1 is an effector of anoikis (cell death due to loss of attachment) in suspended cells. The anoikis function of Bit-1 can be counteracted by integrin-mediated cell attachment. Here, we explored integrin regulation of Bit-1 in adherent cells. We show that knockdown of endogenous Bit-1 in adherent cells decreased cell survival and re-expression of Bit-1 abrogated this effect. Furthermore, reduction of Bit-1 promoted both staurosporine and serum-deprivation induced apoptosis. Indeed knockdown of Bit-1 in these cells led to increased apoptosis as determined by caspase-3 activation and positive TUNEL staining. Bit-1 expression protected cells from apoptosis by increasing phospho-IκB levels and subsequently bcl-2 gene transcription. Protection from apoptosis under serum-free conditions correlated with bcl-2 transcription and Bcl-2 protein expression. Finally, Bit-1-mediated regulation of bcl-2 was dependent on focal adhesion kinase, PI3K, and AKT. Thus, we have elucidated an integrin-controlled pathway in which Bit-1 is, in part, responsible for the survival effects of cell-ECM interactions.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Gene Expression Regulation , Mitochondrial Proteins/metabolism , Animals , Apoptosis , CHO Cells , Caspase 3/metabolism , Cell Adhesion , Cell Survival , Cricetinae , Cricetulus , Culture Media, Serum-Free/pharmacology , Fibronectins/chemistry , Green Fluorescent Proteins/chemistry , Humans , Integrins/metabolism , Mice , Neoplasm Metastasis , Plasmids/metabolism , Transfection
18.
FASEB J ; 24(8): 2818-28, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20354143

ABSTRACT

PEA-15 is a death effector domain-containing phosphoprotein that binds ERK and restricts it to the cytoplasm. PEA-15 also binds to FADD and thereby blocks apoptosis induced by death receptors. Abnormal expression of PEA-15 is associated with type II diabetes and some cancers; however, its physiological function remains unclear. To determine the function of PEA-15 in vivo, we used C57BL/6 mice in which the PEA-15 coding region was deleted. We thereby found that PEA-15 regulates T-cell proliferation. PEA-15-null mice did not have altered thymic or splenic lymphocyte cellularity or differentiation. However, PEA-15 deficient T cells had increased CD3/CD28-induced nuclear translocation of ERK and increased activation of IL-2 transcription and secretion in comparison to control wild-type littermates. Indeed, activation of the T-cell receptor in wild-type mice caused PEA-15 release of ERK. In contrast, overexpression of PEA-15 in Jurkat T cells blocked nuclear translocation of ERK and IL-2 transcription. Finally, PEA-15-null T cells showed increased IL-2 dependent proliferation on stimulation. No differences in T cell susceptibility to apoptosis were found. Thus, PEA-15 is a novel player in T-cell homeostasis. As such this work may have far reaching implications in understanding how the immune response is controlled.


Subject(s)
Phosphoproteins/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , Active Transport, Cell Nucleus , Animals , Apoptosis Regulatory Proteins , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-2/genetics , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/immunology , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...