Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 14(1): 1459, 2023 03 16.
Article de Anglais | MEDLINE | ID: mdl-36927729

RÉSUMÉ

There has been considerable scientific effort dedicated to understanding the biologic consequence and therapeutic implications of aberrant tryptophan metabolism in brain tumors and neurodegenerative diseases. A majority of this work has focused on the upstream metabolism of tryptophan; however, this has resulted in limited clinical application. Using global metabolomic profiling of patient-derived brain tumors, we identify the downstream metabolism of tryptophan and accumulation of quinolinate (QA) as a metabolic node in glioblastoma and demonstrate its critical role in promoting immune tolerance. QA acts as a metabolic checkpoint in glioblastoma by inducing NMDA receptor activation and Foxo1/PPARγ signaling in macrophages, resulting in a tumor supportive phenotype. Using a genetically-engineered mouse model designed to inhibit production of QA, we identify kynureninase as a promising therapeutic target to revert the potent immune suppressive microenvironment in glioblastoma. These findings offer an opportunity to revisit the biologic consequence of this pathway as it relates to oncogenesis and neurodegenerative disease and a framework for developing immune modulatory agents to further clinical gains in these otherwise incurable diseases.


Sujet(s)
Produits biologiques , Tumeurs du cerveau , Glioblastome , Maladies neurodégénératives , Souris , Animaux , Glioblastome/génétique , Tryptophane/métabolisme , Acide quinolinique/métabolisme , Récepteur PPAR gamma/métabolisme , Maladies neurodégénératives/métabolisme , Récepteurs du N-méthyl-D-aspartate/métabolisme , Macrophages/métabolisme , Tumeurs du cerveau/anatomopathologie , Tolérance immunitaire , Produits biologiques/métabolisme , Microenvironnement tumoral
2.
Cancers (Basel) ; 14(22)2022 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-36428642

RÉSUMÉ

Glioblastoma (GBM) represents an aggressive and immune-resistant cancer. Preclinical investigations have identified anti-tumor activity of a ketogenic diet (KD) potentially being used to target GBM's glycolytic phenotype. Since immune cells in the microenvironment have a similar reliance upon nutrients to perform their individual functions, we sought to determine if KD influenced the immune landscape of GBM. Consistent with previous publications, KD improved survival in GBM in an immune-competent murine model. Immunophenotyping of tumors identified KD-influenced macrophage polarization, with a paradoxical 50% increase in immune-suppressive M2-like-macrophages and a decrease in pro-inflammatory M1-like-macrophages. We recapitulated KD in vitro using a modified cell culture based on metabolomic profiling of serum in KD-fed mice, mechanistically linking the observed changes in macrophage polarization to PPARγ-activation. We hypothesized that parallel increases in M2-macrophage polarization tempered the therapeutic benefit of KD in GBM. To test this, we performed investigations combining KD with the CSF-1R inhibitor (BLZ945), which influences macrophage polarization. The combination demonstrated a striking improvement in survival and correlative studies confirmed BLZ945 normalized KD-induced changes in macrophage polarization. Overall, KD demonstrates antitumor activity in GBM; however, its efficacy is attenuated by promoting an immunosuppressive phenotype in macrophages. Combinatorial strategies designed to modulate macrophage polarization represent a rational approach to improve the anti-tumor activity of KD in GBM.

3.
In Vivo ; 35(1): 119-129, 2021.
Article de Anglais | MEDLINE | ID: mdl-33402457

RÉSUMÉ

AIM: To use inhibition of colony-stimulating factor-1 receptor (CSF-1R) to target tumor-associated macrophages (TAMs) and improve the efficacy of radiotherapy in glioblastoma (GBM). MATERIALS AND METHODS: The CSF-1R inhibitor BLZ-945 was used to examine the impact of CSF-1R inhibition on M2 polarization in vitro. Using an orthotopic, immunocompetent GBM model, mice were treated with vehicle, RT, BLZ-945, or RT plus BLZ-945. RESULTS: BLZ-945 reduced M2 polarization in vitro. BLZ-945 alone did not improve median overall survival (mOS=29 days) compared to control mice (mOS=27 days). RT improved survival (mOS=45 days; p=0.02), while RT plus BLZ-945 led to the longest survival (mOS=not reached; p=0.005). Resected tumors had a relatively large population of M2 TAMs in GBM at baseline, which was increased in response to RT. BLZ-945 reduced RT-induced M2 infiltration. CONCLUSION: Inhibition of CSF-1R improved response to RT in the treatment of GBM and may represent a promising strategy to improve RT-induced antitumor immune responses.


Sujet(s)
Glioblastome , Animaux , Facteurs de stimulation des colonies , Glioblastome/traitement médicamenteux , Glioblastome/radiothérapie , Macrophages , Souris , Récepteurs à activité tyrosine kinase
4.
Mol Cancer Ther ; 19(7): 1415-1422, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32430486

RÉSUMÉ

Glioblastoma is the most common primary malignant brain tumor in adults. Despite aggressive treatment, outcomes remain poor with few long-term survivors. Therefore, considerable effort is being made to identify novel therapies for this malignancy. Targeting tumor metabolism represents a promising therapeutic strategy and activation of fatty acid oxidation (FAO) has been identified as a central metabolic node contributing toward gliomagenesis. Perhexiline is a compound with a long clinical track record in angina treatment and commonly described as an FAO inhibitor. We therefore sought to determine whether this compound might be repurposed to serve as a novel therapy in glioblastoma. Perhexiline demonstrated potent in vitro cytotoxicity, induction of redox stress and apoptosis in a panel of glioblastoma cell lines. However, the antitumor activity of perhexiline was distinct when compared with the established FAO inhibitor etomoxir. By evaluating mitochondrial respiration and lipid dynamics in glioblastoma cells following treatment with perhexiline, we confirmed this compound did not inhibit FAO in our models. Using in silico approaches, we identified FYN as a probable target of perhexiline and validated the role of this protein in perhexiline sensitivity. We extended studies to patient samples, validating the potential of FYN to serve as therapeutic target in glioma. When evaluated in vivo, perhexiline demonstrated the capacity to cross the blood-brain barrier and antitumor activity in both flank and orthotopic glioblastoma models. Collectively, we identified potent FYN-dependent antitumor activity of perhexiline in glioblastoma, thereby, representing a promising agent to be repurposed for the treatment of this devastating malignancy.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Inhibiteurs des canaux calciques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Glioblastome/traitement médicamenteux , Perhexiline/pharmacologie , Protéines proto-oncogènes c-fyn/métabolisme , Animaux , Apoptose , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Prolifération cellulaire , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , Souris , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Oxydoréduction , Protéines proto-oncogènes c-fyn/génétique , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Cell Death Dis ; 11(4): 253, 2020 04 20.
Article de Anglais | MEDLINE | ID: mdl-32312953

RÉSUMÉ

Despite advances in molecularly characterizing glioblastoma (GBM), metabolic alterations driving its aggressive phenotype are only beginning to be recognized. Integrative cross-platform analysis coupling global metabolomic and gene expression profiling on patient-derived glioma identified fatty acid ß-oxidation (FAO) as a metabolic node in GBM. We determined that the biologic consequence of enhanced FAO is directly dependent upon tumor microenvironment. FAO serves as a metabolic cue to drive proliferation in a ß-HB/GPR109A dependent autocrine manner in nutrient favorable conditions, while providing an efficient, alternate source of ATP only in nutrient unfavorable conditions. Rational combinatorial strategies designed to target these dynamic roles FAO plays in gliomagenesis resulted in necroptosis-mediated metabolic synthetic lethality in GBM. In summary, we identified FAO as a dominant metabolic node in GBM that provides metabolic plasticity, allowing these cells to adapt to their dynamic microenvironment. Combinatorial strategies designed to target these diverse roles FAO plays in gliomagenesis offers therapeutic potential in GBM.


Sujet(s)
Tumeurs du cerveau/métabolisme , Plasticité cellulaire/physiologie , Acides gras/métabolisme , Glioblastome/métabolisme , Microenvironnement tumoral/physiologie , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes/méthodes , Glioblastome/anatomopathologie , Humains , Métabolomique/méthodes , Oxydoréduction , Phénotype , Microenvironnement tumoral/génétique
6.
Cancer Immunol Immunother ; 68(7): 1107-1120, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31119318

RÉSUMÉ

Glioblastoma (GBM) is one of the most aggressive tumors. Numerous studies in the field of immunotherapy have focused their efforts on identifying various pathways linked with tumor-induced immunosuppression. Recent research has demonstrated that metabolic reprogramming in a tumor can contribute towards immune tolerance. To begin to understand the interface between metabolic remodeling and the immune-suppressive state in GBM, we performed a focused, integrative analysis coupling metabolomics with gene-expression profiling in patient-derived GBM (n = 80) and compared them to low-grade astrocytoma (LGA; n = 28). Metabolic intermediates of tryptophan, arginine, prostaglandin, and adenosine emerged as immuno-metabolic nodes in GBM specific to the mesenchymal and classical molecular subtypes of GBM. Integrative analyses emphasized the importance of downstream metabolism of several of these metabolic pathways in GBM. Using CIBERSORT to analyze immune components from the transcriptional profiles of individual tumors, we demonstrated that tryptophan and adenosine metabolism resulted in an accumulation of Tregs and M2 macrophages, respectively, and was recapitulated in mouse models. Furthermore, we extended these findings to preclinical models to determine their potential utility in defining the biologic and/or immunologic consequences of the identified metabolic programs. Collectively, through integrative analysis, we uncovered multifaceted ways by which metabolic reprogramming may contribute towards immune tolerance in GBM, providing the framework for further investigations designed to determine the specific immunologic consequence of these metabolic programs and their therapeutic potential.


Sujet(s)
Tumeurs du cerveau/immunologie , Glioblastome/immunologie , Voies et réseaux métaboliques/immunologie , Métabolome/immunologie , Adénosine/métabolisme , Adulte , Animaux , Encéphale/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , Macrophages/immunologie , Macrophages/métabolisme , Métabolomique/méthodes , Souris , Souris de lignée C57BL , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Tryptophane/métabolisme
7.
Neuro Oncol ; 21(3): 337-347, 2019 02 19.
Article de Anglais | MEDLINE | ID: mdl-30476237

RÉSUMÉ

BACKGROUND: Although considerable progress has been made in understanding molecular alterations driving gliomagenesis, the diverse metabolic programs contributing to the aggressive phenotype of glioblastoma remain unclear. The aim of this study was to define and provide molecular context to metabolic reprogramming driving gliomagenesis. METHODS: Integrative cross-platform analyses coupling global metabolomic profiling with genomics in patient-derived glioma (low-grade astrocytoma [LGA; n = 28] and glioblastoma [n = 80]) were performed. Identified programs were then metabolomically, genomically, and functionally evaluated in preclinical models. RESULTS: Clear metabolic programs were identified differentiating LGA from glioblastoma, with aberrant lipid, peptide, and amino acid metabolism representing dominant metabolic nodes associated with malignant transformation. Although the metabolomic profiles of glioblastoma and LGA appeared mutually exclusive, considerable metabolic heterogeneity was observed in glioblastoma. Surprisingly, integrative analyses demonstrated that O6-methylguanine-DNA methyltransferase methylation and isocitrate dehydrogenase mutation status were equally distributed among glioblastoma metabolic profiles. Transcriptional subtypes, on the other hand, tightly clustered by their metabolomic signature, with proneural and mesenchymal tumor profiles being mutually exclusive. Integrating these metabolic phenotypes with gene expression analyses uncovered tightly orchestrated and highly redundant transcriptional programs designed to support the observed metabolic programs by actively importing these biochemical substrates from the microenvironment, contributing to a state of enhanced metabolic heterotrophy. These findings were metabolomically, genomically, and functionally recapitulated in preclinical models. CONCLUSION: Despite disparate molecular pathways driving the progression of glioblastoma, metabolic programs designed to maintain its aggressive phenotype remain conserved. This contributes to a state of enhanced metabolic heterotrophy supporting survival in diverse microenvironments implicit in this malignancy.


Sujet(s)
Acides aminés/métabolisme , Astrocytome/métabolisme , Tumeurs du cerveau/métabolisme , Carcinogenèse , Glioblastome/métabolisme , Métabolisme lipidique , Métabolomique , Astrocytome/génétique , Astrocytome/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Reprogrammation cellulaire , Méthylation de l'ADN , DNA modification methylases/génétique , Enzymes de réparation de l'ADN/génétique , Analyse de profil d'expression de gènes , Glioblastome/génétique , Glioblastome/anatomopathologie , Humains , Isocitrate dehydrogenases/génétique , Mutation , Grading des tumeurs , Peptides/métabolisme , Protéines suppresseurs de tumeurs/génétique
8.
Clin Cancer Res ; 24(15): 3632-3643, 2018 08 01.
Article de Anglais | MEDLINE | ID: mdl-29691296

RÉSUMÉ

Purpose: Immune checkpoint inhibitors designed to revert tumor-induced immunosuppression have emerged as potent anticancer therapies. Tryptophan metabolism represents an immune checkpoint, and targeting this pathway's rate-limiting enzyme IDO1 is actively being investigated clinically. Here, we studied the intermediary metabolism of tryptophan metabolism in glioblastoma and evaluated the activity of the IDO1 inhibitor GDC-0919, both alone and in combination with radiation (RT).Experimental Design: LC/GC-MS and expression profiling was performed for metabolomic and genomic analyses of patient-derived glioma. Immunocompetent mice were injected orthotopically with genetically engineered murine glioma cells and treated with GDC-0919 alone or combined with RT. Flow cytometry was performed on isolated tumors to determine immune consequences of individual treatments.Results: Integrated cross-platform analyses coupling global metabolomic and gene expression profiling identified aberrant tryptophan metabolism as a metabolic node specific to the mesenchymal and classical subtypes of glioblastoma. GDC-0919 demonstrated potent inhibition of this node and effectively crossed the blood-brain barrier. Although GDC-0919 as a single agent did not demonstrate antitumor activity, it had a strong potential for enhancing RT response in glioblastoma, which was further augmented with a hypofractionated regimen. RT response in glioblastoma involves immune stimulation, reflected by increases in activated and cytotoxic T cells, which was balanced by immune checkpoint reactivation, reflected by an increase in IDO1 expression and regulatory T cells (Treg). GDC-0919 mitigated RT-induced Tregs and enhanced T-cell activation.Conclusions: Tryptophan metabolism represents a metabolic node in glioblastoma, and combining RT with IDO1 inhibition enhances therapeutic response by mitigating RT-induced immunosuppression. Clin Cancer Res; 24(15); 3632-43. ©2018 AACR.


Sujet(s)
Points de contrôle du cycle cellulaire/immunologie , Antienzymes/administration et posologie , Glioblastome/traitement médicamenteux , Imidazoles/pharmacologie , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Indoles/pharmacologie , Tryptophane/métabolisme , Animaux , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des radiations , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/immunologie , Glioblastome/immunologie , Glioblastome/anatomopathologie , Glioblastome/radiothérapie , Humains , Imidazoles/usage thérapeutique , Indoleamine-pyrrole 2,3,-dioxygenase/immunologie , Indoles/usage thérapeutique , Métabolomique , Souris , Radiothérapie/effets indésirables , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Tryptophane/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Neuro Oncol ; 19(12): 1599-1606, 2017 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-28541485

RÉSUMÉ

BACKGROUND: Glioblastoma represents an archetypal example of a heterogeneous malignancy. To understand the diverse molecular consequences of this complex tumor ecology, we analyzed RNA-seq data generated from commonly identified intratumoral structures in glioblastoma enriched using laser capture microdissection. METHODS: Raw gene-level values of fragments per kilobase of transcript per million reads mapped and the associated clinical data were acquired from the publicly available Ivy Glioblastoma Atlas Project database and analyzed using MetaboAnalyst (v3.0). The database includes gene expression data generated from multiple structural features commonly identified in glioblastoma enriched by laser capture microdissection. RESULTS: We uncovered a relationship between subtype heterogeneity in glioblastoma and its unique tumor microenvironment, with infiltrating cells harboring a proneural signature while the mesenchymal subtype was enriched in perinecrotic regions. When evaluating the tumors' transcriptional profiles in the context of their derived structural regions, there was a relatively small amount of intertumoral heterogeneity in glioblastoma, with individual regions from different tumors clustering tightly together. Analyzing the transcriptional profiles in the context of evolutionary progression identified unique cellular programs associated with specific phases of gliomagenesis. Mediators of cell signaling and cell cycle progression appear to be critical events driving proliferation in the tumor core, while in addition to a multiplex strategy for promoting angiogenesis and/or an immune-tolerant environment, transformation to perinecrotic zones involved global metabolic alterations. CONCLUSION: These findings suggest that intratumoral heterogeneity in glioblastoma is a conserved, predictable consequence to its complex microenvironment, and combinatorial approaches designed to target these unequivocally present tumor biomes may lead to therapeutic gains.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Tumeurs du cerveau/anatomopathologie , Évolution moléculaire , Régulation de l'expression des gènes tumoraux , Glioblastome/anatomopathologie , Séquençage nucléotidique à haut débit/méthodes , Microenvironnement tumoral/génétique , Tumeurs du cerveau/génétique , Analyse de profil d'expression de gènes , Glioblastome/génétique , Humains , Pronostic
10.
Neuro Oncol ; 19(10): 1308-1315, 2017 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-28541512

RÉSUMÉ

The fields of tumor metabolism and immune oncology have both independently received considerable attention over the last several years. The majority of research in tumor metabolism has largely focused on the Warburg effect and its resulting biologic consequences, including energy and macromolecule production. However, recent investigations have identified elegant, multifaceted strategies by which alterations in tumor metabolism can also contribute to a potent tolerogenic immune environment. One of the most notable is increased tryptophan metabolism through activation of indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO). However, this pathway represents one of numerous metabolic pathways that may modulate the immune system. For example, metabolites associated with aerobic glycolysis, adenosine, arginine, and prostaglandin metabolism have all been implicated in cancer-mediated immune tolerance and represent attractive therapeutic targets. In this review, we will provide an overview of the emerging interface between these 2 timely areas of cancer research and provide an overview of strategies currently being tested to target these next-generation metabolic immune checkpoints.


Sujet(s)
Glioblastome/immunologie , Transduction du signal/immunologie , Tryptophane 2,3-dioxygenase/métabolisme , Tryptophane/métabolisme , Glioblastome/métabolisme , Humains , Indoleamine-pyrrole 2,3,-dioxygenase/immunologie , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Cynurénine/immunologie , Cynurénine/métabolisme , Tryptophane/immunologie , Tryptophane 2,3-dioxygenase/immunologie
11.
Mol Cell Biochem ; 397(1-2): 167-78, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25123669

RÉSUMÉ

Bicarbonate transporter (BCT) plays a crucial role in maintaining pH homeostasis of tumor cells by import of HCO3(-). This helps the tumor cells in manifesting extracellular tumor acidosis, accompanied by a relative intracellular alkalinization, which in turn promotes tumor progression. Therefore, blocking BCT-mediated HCO3(-) transport is envisaged as a promising anticancer therapeutic approach. Thus, using a murine model of a T cell lymphoma, designated as Dalton's lymphoma (DL), in the present in vitro investigation the antitumor consequences of blocking BCT function by its inhibitor 4-acetamido-4-isothiocyanostilbene-2,2-disulfonate (SITS) were explored. Treatment of DL cells with SITS resulted in an increase in the extracellular pH, associated with a decline in DL cell survival and augmented induction of apoptosis. BCT inhibition also elevated the expression of cytochrome c, caspase-9, caspase-3, Bax, reactive oxygen species, and nitric oxide along with inhibition of HSP-70 and Bcl2, which regulate tumor cell survival and apoptosis. SITS-treated DL cells displayed upregulated production of IFN-γ and IL-6 along with a decline of IL-10. Treatment of DL cells with SITS also inhibited the expression of fatty acid synthase, which is crucial for membrane biogenesis in neoplastic cells. The expression of lactate transporter MCT-1 and multidrug resistance regulating protein MRP-1 got inhibited along with hampered uptake of glucose and lactate production in SITS-treated DL cells. Thus, the declined tumor cell survival following inhibition of BCT could be the consequence of interplay of several inter-connected regulatory molecular events. The outcome of this study indicates the potential of BCT inhibition as a novel therapeutic approach for treatment of hematological malignancies.


Sujet(s)
Acide 4-acétamido-4'-isothiocyanato-stilbène-2,2'-disulfonique/pharmacologie , Transporteurs d'anions/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Lymphome T/traitement médicamenteux , Protéines tumorales/métabolisme , Tumeurs expérimentales/traitement médicamenteux , Symporteurs/antagonistes et inhibiteurs , Animaux , Transporteurs d'anions/génétique , Transporteurs d'anions/métabolisme , Apoptose/génétique , Protéines régulatrices de l'apoptose/génétique , Protéines régulatrices de l'apoptose/métabolisme , Hydrogénocarbonates/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Homéostasie/génétique , Concentration en ions d'hydrogène , Lymphome T/génétique , Lymphome T/métabolisme , Lymphome T/anatomopathologie , Souris , Souris de lignée BALB C , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Tumeurs expérimentales/génétique , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Symporteurs/génétique , Symporteurs/métabolisme
12.
Biochim Biophys Acta ; 1840(1): 294-302, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24060750

RÉSUMÉ

BACKGROUND: Orlistat, a fatty acid synthase (FASN) inhibitor, has been demonstrated to inhibit tumor cell survival. However, the mechanism(s) of its tumor growth retarding action against malignancies of hematological origin remains unclear. It is also not understood if the antitumor action of orlistat implicates modulated susceptibility of tumor cell to anticancer drugs. Therefore, the present investigation focuses to study the antitumor and chemosensitizing action of orlistat in a murine host bearing a progressively growing T cell lymphoma. METHODS: Tumor-bearing mice were administered with vehicle alone or containing orlistat followed by administration of PBS with or without cisplatin. Tumor progression and survival of tumor-bearing host were monitored along with analysis of tumor cell survival and apoptosis. Tumor ascitic fluid was examined for pH, NO and cytokines. Expression of genes and proteins was investigated by RT-PCR and western blot respectively. ROS was analyzed by DCFDA staining and FASN activity by spectrophotometry. RESULTS: Orlistat administration to tumor-bearing mice resulted in tumor growth retardation, prolonged life span, declined tumor cell survival and chemosensitization to cisplatin. It was accompanied by increased osmotic fragility, modulated acidosis, expression of ROS, NO, cytokines, MCT-1 and VH(+) ATPase, Bcl2, Caspase-3, P53, inhibited FASN activity and declined expression of MDR and MRP-1 proteins. CONCLUSION: Orlistat manifests antitumor and chemosensitizing action implicating modulated regulation of cell survival, reconstituted-tumor microenvironment and altered MDR phenotype. GENERAL SIGNIFICANCE: These observations indicate that orlistat could be utilized as an adjunct regimen for improving antitumor efficacy of cisplatin.


Sujet(s)
Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Fatty acid synthases/antagonistes et inhibiteurs , Lactones/pharmacologie , Lymphome T/prévention et contrôle , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Agents antiobésité/pharmacologie , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Modèles animaux de maladie humaine , Association de médicaments , Test ELISA , Fatty acid synthases/génétique , Fatty acid synthases/métabolisme , Cytométrie en flux , Lymphome T/métabolisme , Lymphome T/anatomopathologie , Souris , Souris de lignée BALB C , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Nitrites/métabolisme , Orlistat , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR
13.
PLoS One ; 8(12): e82396, 2013.
Article de Anglais | MEDLINE | ID: mdl-24349275

RÉSUMÉ

Orlistat, an inhibitor of fatty acid synthase (FASN), acts as an antitumor agent by blocking de novo fatty acid synthesis of tumor cells. Although, myelopoiesis also depends on de novo fatty acid synthesis, the effect of orlistat on differentiation of macrophages, which play a central role in host's antitumor defence, remains unexplored in a tumor-bearing host. Therefore, the present investigation was undertaken to examine the effect of orlistat administration on macrophage differentiation in a T cell lymphoma bearing host. Administration of orlistat (240 mg/kg/day/mice) to tumor-bearing mice resulted in a decline of tumor load accompanied by an augmentation of bone marrow cellularity and survival of bone marrow cells (BMC). The expression of apoptosis regulatory caspase-3, Bax and Bcl2 was modulated in the BMC of orlistat-administered tumor-bearing mice. Orlistat administration also resulted in an increase in serum level of IFN-γ along with decreased TGF-ß and IL-10. BMC of orlistat-administered tumor-bearing mice showed augmented differentiation into macrophages accompanied by enhanced expression of macrophage colony stimulating factor (M-CSF) and its receptor (M-CSFR). The macrophages differentiated from BMC of orlistat-administered mice showed characteristic features of M1 macrophage phenotype confirmed by expression of CD11c, TLR-2, generation of reactive oxygen species, phagocytosis, tumor cell cytotoxicity, production of IL-1,TNF-α and nitric oxide. These novel findings indicate that orlistat could be useful to support myelopoesis in a tumor-bearing host.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Lymphome T/anatomopathologie , Activation des macrophages/effets des médicaments et des substances chimiques , Macrophages/anatomopathologie , Myélopoïèse/effets des médicaments et des substances chimiques , Animaux , Apoptose/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Numération cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Lactones/administration et posologie , Lactones/pharmacologie , Facteur de stimulation des colonies de macrophages/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Souris , Souris de lignée BALB C , Orlistat , Phénotype , Récepteur du facteur de stimulation des colonies de macrophages/métabolisme
14.
Toxicol Appl Pharmacol ; 273(1): 196-208, 2013 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-24051182

RÉSUMÉ

Targeting of tumor metabolism is emerging as a novel therapeutic strategy against cancer. Dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), has been shown to exert a potent tumoricidal action against a variety of tumor cells. The main mode of its antineoplastic action implicates a shift of glycolysis to oxidative metabolism of glucose, leading to generation of cytotoxic reactive oxygen intermediates. However, the effect of DCA on tumor microenvironment, which in turn regulates tumor cell survival; remains speculative to a large extent. It is also unclear if DCA can exert any modulatory effect on the process of hematopoiesis, which is in a compromised state in tumor-bearing hosts undergoing chemotherapy. In view of these lacunas, the present study was undertaken to investigate the so far unexplored aspects with respect to the molecular mechanisms of DCA-dependent tumor growth retardation and chemosensitization. BALB/c mice were transplanted with Dalton's lymphoma (DL) cells, a T cell lymphoma of spontaneous origin, followed by administration of DCA with or without cisplatin. DCA-dependent tumor regression and chemosensitization to cisplatin was found to be associated with altered repertoire of key cell survival regulatory molecules, modulated glucose metabolism, accompanying reconstituted tumor microenvironment with respect to pH homeostasis, cytokine balance and alternatively activated TAM. Moreover, DCA administration also led to an alteration in the MDR phenotype of tumor cells and myelopoietic differentiation of macrophages. The findings of this study shed a new light with respect to some of the novel mechanisms underlying the antitumor action of DCA and thus may have immense clinical applications.


Sujet(s)
Antinéoplasiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Acide dichloro-acétique/pharmacologie , Glucose/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Apoptose/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Glycolyse , Méthode TUNEL , Lymphome T/anatomopathologie , Macrophages/cytologie , Macrophages/métabolisme , Mâle , Souris , Souris de lignée BALB C , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase
15.
Apoptosis ; 18(12): 1574-85, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-23955790

RÉSUMÉ

The present investigation was undertaken to study the effect of in vitro exposure of Colo205, colonadenocarcinoma cells, to monocarboxylate transporter inhibitor α-cyano-4-hydroxycinnamate (αCHC) on cell survival and evolution of resistance to chemotherapeutic drug cisplatin. αCHC-treated Colo205 cells showed inhibition of survival accompanied by an augmented induction of apoptosis. Changes in cell survival properties were associated with alterations in lactate efflux, pH homeostasis, expression of glucose transporters, glucose uptake, HIF-1α, generation of nitric oxide, expression pattern of some key cell survival regulatory molecules: Bcl2, Bax, active caspase-3 and p53. Pretreatment of Colo205 cells with αCHC also altered their susceptibility to the cytotoxicity of cisplatin accompanied by altered expression of multidrug resistance regulating MDR1 and MRP1 genes. This study for the first time deciphers some of the key molecular events underlying modulation of cell survival of cancer cells of colorectal origin by αCHC and its contribution to chemosensitization against cisplatin. Thus these findings will be of immense help in further research for optimizing the use of αCHC for improving the chemotherapeutic efficacy of anticancer drugs like cisplatin.


Sujet(s)
Adénocarcinome/physiopathologie , Apoptose/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Tumeurs du côlon/physiopathologie , Acides coumariques/pharmacologie , Résistance aux médicaments antinéoplasiques , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Protéines du muscle/antagonistes et inhibiteurs , Adénocarcinome/traitement médicamenteux , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Caspase-3/génétique , Caspase-3/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/génétique , Tumeurs du côlon/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Transporteurs d'acides monocarboxyliques/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Protéines du muscle/génétique , Protéines du muscle/métabolisme
16.
Anticancer Drugs ; 24(2): 158-71, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-22932131

RÉSUMÉ

In the present investigation, we explored the molecular mechanisms of the tumoricidal action of α-cyano-4-hydroxycinnamate (αCHC) on the cells of Dalton's lymphoma (DL), which is a murine T cell lymphoma. In-vitro treatment of the DL cells with αCHC resulted in the modulation of the biophysical parameters of the tumor cell culture medium with respect to pH, nitric oxide, glucose, and lactate, accompanied by an alteration in the expression of cytokines (IL-10, IL-6, and IFN-γ) and cell survival-regulatory proteins such as Bcl2, p53, caspase-3, caspase-activated DNase, HSP70, and IL2R. The expression of the pH-regulatory proteins vacuolar ATPase and MCT1 was also found to be altered. The study discusses the possible role of the aforementioned alterations triggered by αCHC in the induction of tumor cell apoptosis and decreased cell survival. These findings will provide a new insight into the novel molecular mechanisms of the antitumor action of αCHC.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Acides coumariques/pharmacologie , Lymphome T/traitement médicamenteux , Animaux , Protéines du cycle cellulaire/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Glucose/métabolisme , Interféron gamma/métabolisme , Interleukine-10/métabolisme , Interleukine-6/métabolisme , Acide lactique/métabolisme , Lymphome T/anatomopathologie , Mâle , Souris , Souris de lignée BALB C , Monoxyde d'azote/métabolisme , Protéines oncogènes/métabolisme , Thymocytes/effets des médicaments et des substances chimiques , Thymocytes/métabolisme , Cellules cancéreuses en culture
17.
Biochim Biophys Acta ; 1820(11): 1764-73, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22877747

RÉSUMÉ

BACKGROUND: De novo fatty acid synthesis catalyzed by fatty acid synthase (FASN) is crucial for tumor cell survival. Thus therapeutic targeting of FASN is considered as a novel antineoplastic strategy. However, little is understood in this respect regarding malignancies of hematological origin. The present investigation was therefore, undertaken to study the molecular mechanisms of the antitumor action of FASN inhibitor orlistat (tetrahydrolipstatin) using a murine model of a T cell lymphoma. METHODS: The antitumor efficacy of orlistat was investigated in vitro by estimating cell survival by MTT assay and apoptosis by Wright Giemsa, TUNEL, Annexin-V/PI staining and % DNA fragmentation. Generation of reactive oxygen species (ROS) in tumor cells was studied using fluorescence microscopy. Expression of genes and proteins was carried out by RT-PCR and western blot analyses respectively. FASN and CPT-1 activity was estimated by spectrophotometer. Cytokines expression was analyzed by ELISA. RESULTS: We report that inhibition of FASN with its specific inhibitor orlistat manifests tumor-specific inhibition of cell survival, accompanied by induction of apoptosis. Orlistat-treated tumor cells showed an altered ROS generation, shift in cytokine balance and modulated expression of cell survival regulatory molecules like HSP70, Bcl2, p53, PUMA, Caspase-3 and CAD. It was observed that IFN-γ mediates orlistat-dependent modulation of FASN expression. CONCLUSION AND GENERAL SIGNIFICANCE: In this study, we report some of the so far unexplored novel aspects underlying the molecular mechanisms associated with orlistat-dependent modulation of tumor cell survival. These observations will help in designing antineoplastic therapeutic protocols using orlistat against malignancies of hematological origin.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Fatty acid synthases/antagonistes et inhibiteurs , Lactones/pharmacologie , Lymphome T/traitement médicamenteux , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Cytokines/biosynthèse , Relation dose-effet des médicaments , Protéines du choc thermique HSP70/physiologie , Interféron gamma/physiologie , Lymphome T/anatomopathologie , Mâle , Souris , Souris de lignée BALB C , Orlistat , Espèces réactives de l'oxygène/métabolisme
18.
Toxicol Appl Pharmacol ; 263(1): 111-21, 2012 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-22714040

RÉSUMÉ

The present investigation was undertaken to study if curcumin, which is recognized for its potential as an antineoplastic and immunopotentiating agent, can also influence the process of myelopoiesis in a tumor-bearing host. Administration of curcumin to tumor-bearing host augmented count of bone marrow cell (BMC) accompanied by an up-regulated BMC survival and a declined induction of apoptosis. Curcumin administration modulated expression of cell survival regulatory molecules: Bcl2, p53, caspase-activated DNase (CAD) and p53-upregulated modulator of apoptosis (PUMA) along with enhanced expression of genes of receptors for M-CSF and GM-CSF in BMC. The BMC harvested from curcumin-administered hosts showed an up-regulated colony forming ability with predominant differentiation into bone marrow-derived macrophages (BMDM), responsive for activation to tumoricidal state. The number of F4/80 positive bone marrow resident macrophages (BMM), showing an augmented expression of M-CSF, was also augmented in the bone marrow of curcumin-administered host. In vitro reconstitution experiments indicated that only BMM of curcumin-administered hosts, but not in vitro curcumin-exposed BMM, augmented BMC survival. It suggests that curcumin-dependent modulation of BMM is of indirect nature. Such prosurvival action of curcumin is associated with altered T(H1)/T(H2) cytokine balance in serum. Augmented level of serum-borne IFN-γ was found to mediate modulation of BMM to produce enhanced amount of monokines (IL-1, IL-6, TNF-α), which are suggested to augment the BMC survival. Taken together the present investigation indicates that curcumin can potentiate myelopoiesis in a tumor-bearing host, which may have implications in its therapeutic utility.


Sujet(s)
Curcumine/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Myélopoïèse/effets des médicaments et des substances chimiques , Tumeurs expérimentales/physiopathologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Cytokines/physiologie , Relation dose-effet des médicaments , Test ELISA , Méthode TUNEL , Macrophages/physiologie , Mâle , Souris , Souris de lignée BALB C , Myélopoïèse/physiologie , Transplantation tumorale
19.
Chem Biol Interact ; 199(1): 29-37, 2012 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-22705712

RÉSUMÉ

Pyruvate dehydrogenase kinase (PDK) inhibits pyruvate dehydrogenase (PDH) activity and thus promotes energetic switch from mitochondrial glucose oxidation to cytoplasmic glycolysis in cancerous cells (a phenomenon known as the 'Warburg effect') for their energy need, which facilitates the cancer progression by resisting induction of apoptosis and promoting tumor metastasis. Thus, in the present investigation, we explored the molecular mechanisms of the tumoricidal action of dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor, on cells of a murine T cell lymphoma, designated as Dalton's lymphoma (DL). In vitro treatment of tumor cells with DCA inhibited their survival accompanied by a modulation of the biophysical composition of tumor-conditioned medium with respect to pH, glucose and lactate. DCA treatment also altered expression of HIF1-α and pH regulators: VATPase and MCT1 and production of cytokines: IL-10, IL-6 and IFN-γ. Moreover, we also observed an alteration in the expression of other apoptosis and cell survival regulatory molecules: PUMA, GLUT1, Bcl2, p53, CAD, caspase-3 and HSP70. The study discusses the role of novel molecular mechanisms underlying DCA-dependent inhibition of tumor cell survival. This study shows for the first time that DCA-dependent alteration of tumor cell survival involves altered pH homeostasis and glucose metabolism. Thus, these findings will provide a new insight for therapeutic applications of DCA as a novel antineoplastic agent against T cell lymphoma.


Sujet(s)
Antinéoplasiques/pharmacologie , Acide dichloro-acétique/pharmacologie , Glucose/métabolisme , Lymphome T/traitement médicamenteux , Lymphome T/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Milieux de culture conditionnés/composition chimique , Cytokines/métabolisme , Tests de criblage d'agents antitumoraux , Antienzymes/pharmacologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Transporteur de glucose de type 1/métabolisme , Concentration en ions d'hydrogène , Acide lactique/métabolisme , Mâle , Souris , Souris de lignée BALB C , Transporteurs d'acides monocarboxyliques , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyruvate dehydrogenase acetyl-transferring kinase , Espèces réactives de l'oxygène/métabolisme , Symporteurs , Thymocytes/cytologie , Thymocytes/effets des médicaments et des substances chimiques
20.
J Fluoresc ; 21(3): 1255-63, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21240627

RÉSUMÉ

The complex of o-phenylenediamine (o-PDA) and benzoin (BN) was synthesized adopting solid state reaction by mixing of their melt together followed by chilling. The phase diagram study shows the formation of a complex in 1:1 molar ratio with congruent melting point and two eutectics lying on either side of complex. The formation of complex was confirmed using the FTIR, NMR, mass spectroscopy, powder XRD and DSC studies. The optical properties of the parent component, their complex and few other compositions nearby the complex were studied using absorption and laser luminescence techniques. The significantly higher green/yellow emission was noted with newly synthesized complex as compared to that of their parents as well as other compositions of o- PDA and BN.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...