Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
JCI Insight ; 6(2)2021 01 25.
Article En | MEDLINE | ID: mdl-33320838

Limited experimental evidence bridges nutrition and cancer immunosurveillance. Here, we show that ketogenic diet (KD) - or its principal ketone body, 3-hydroxybutyrate (3HB), most specifically in intermittent scheduling - induced T cell-dependent tumor growth retardation of aggressive tumor models. In conditions in which anti-PD-1 alone or in combination with anti-CTLA-4 failed to reduce tumor growth in mice receiving a standard diet, KD, or oral supplementation of 3HB reestablished therapeutic responses. Supplementation of KD with sucrose (which breaks ketogenesis, abolishing 3HB production) or with a pharmacological antagonist of the 3HB receptor GPR109A abolished the antitumor effects. Mechanistically, 3HB prevented the immune checkpoint blockade-linked upregulation of PD-L1 on myeloid cells, while favoring the expansion of CXCR3+ T cells. KD induced compositional changes of the gut microbiota, with distinct species such as Eisenbergiella massiliensis commonly emerging in mice and humans subjected to carbohydrate-low diet interventions and highly correlating with serum concentrations of 3HB. Altogether, these results demonstrate that KD induces a 3HB-mediated antineoplastic effect that relies on T cell-mediated cancer immunosurveillance.


Diet, Ketogenic , Ketone Bodies/administration & dosage , Neoplasms, Experimental/diet therapy , Neoplasms, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , 3-Hydroxybutyric Acid/administration & dosage , 3-Hydroxybutyric Acid/metabolism , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Female , Gastrointestinal Microbiome/immunology , Humans , Immune Checkpoint Inhibitors/administration & dosage , Ketone Bodies/metabolism , Kidney Neoplasms/diet therapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Melanoma, Experimental/diet therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Receptors, G-Protein-Coupled/antagonists & inhibitors
2.
Sci Rep ; 10(1): 20645, 2020 11 26.
Article En | MEDLINE | ID: mdl-33244152

Electroporation in combination with chemotherapy is an established treatment used on solid malignancies that results in enhanced chemotherapeutic uptake. Recent advances have begun to transition to the use of non-toxic compounds, such as calcium, in lieu of chemotherapy, which can also induce tumour cell death. While the effect of treatment on tumour cell death has been well characterized and has been shown to induce an immunogenic form of cell death, the effect of treatment on intratumoural immune cells has not been investigated. Here we present data showing the effect of calcium electroporation on immune cells, using melanoma-conditioned bone marrow-derived macrophages. Similar to tumour cells, macrophage cell membranes are susceptible to poration following treatment and subsequently reseal. Macrophages are less susceptible to calcium electroporation induced cell death in comparison to B16F10 melanoma cells. However treatment with electroporation with or without bleomycin or calcium was shown to affect macrophage phenotype and function. Coculture of calcium electroporated macrophages revealed that both the capacity of macrophages to stimulate and direct T cell responses are affected following exposure to treatment. We conclude that calcium electroporation has the potential to boost the immunogenic capacity of exposed tumour associated macrophages, and further research is warranted to determine if calcium electroporation can be optimised to generate systemic anti-cancer immune responses.


Calcium/pharmacology , Cell Survival/drug effects , Macrophages/drug effects , Melanoma, Experimental/diet therapy , Animals , Bleomycin/pharmacology , Bone Marrow/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Electroporation/methods , Female , Immunity/drug effects , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes/drug effects
3.
Phytother Res ; 33(12): 3228-3241, 2019 Dec.
Article En | MEDLINE | ID: mdl-31486124

The peel of Citrus unshiu Marcow. fruits (CU) has long been used as a traditional medicine that has therapeutic effects against pathogenic diseases, including asthma, vomiting, dyspepsia, blood circulation disorders, and various types of cancer. In this study, we investigated the effect of CU peel on metastatic melanoma, a highly aggressive skin cancer, in B16F10 melanoma cells, and in B16F10 cells inoculated-C57BL/6 mice. Our results show that ethanol extracts of CU (EECU) inhibited cell growth and increased the apoptotic cells in B16F10 cells. EECU also stimulated the induction of mitochondria-mediated intrinsic pathway, with reduced mitochondrial membrane potential and increased generation of intracellular reactive oxygen species. Furthermore, EECU suppressed the migration, invasion, and colony formation of B16F10 cells. In addition, the oral administration of EECU reduced serum lactate dehydrogenase activity without weight loss, hepatotoxicity, nor nephrotoxicity in B16F10 cell-inoculated mice. Moreover, EECU markedly suppressed lung hypertrophy, the number and expression of metastatic tumor nodules, and the expression of inflammatory tumor necrosis factor-alpha in lung tissue. In conclusion, our findings suggest that the inhibitory effect of EECU on the metastasis of melanoma indicates that it may be regarded as a potential therapeutic herbal drug for melanoma.


Citrus/chemistry , Fruit/chemistry , Melanoma, Experimental/diet therapy , Neoplasm Metastasis/drug therapy , Animals , Apoptosis , Mice , Mice, Inbred C57BL
4.
Cell Metab ; 27(4): 828-842.e7, 2018 04 03.
Article En | MEDLINE | ID: mdl-29551590

Dietary restriction (DR) was shown to impact on tumor growth with very variable effects depending on the cancer type. However, how DR limits cancer progression remains largely unknown. Here, we demonstrate that feeding mice a low-protein (Low PROT) isocaloric diet but not a low-carbohydrate (Low CHO) diet reduced tumor growth in three independent mouse cancer models. Surprisingly, this effect relies on anticancer immunosurveillance, as depleting CD8+ T cells, antigen-presenting cells (APCs), or using immunodeficient mice prevented the beneficial effect of the diet. Mechanistically, we established that a Low PROT diet induces the unfolded protein response (UPR) in tumor cells through the activation of IRE1α and RIG1 signaling, thereby resulting in cytokine production and mounting an efficient anticancer immune response. Collectively, our data suggest that a Low PROT diet induces an IRE1α-dependent UPR in cancer cells, enhancing a CD8-mediated T cell response against tumors.


CD8-Positive T-Lymphocytes/immunology , Diet, Protein-Restricted , Endoribonucleases/metabolism , Immunologic Surveillance , Neoplasms, Experimental/diet therapy , Neoplasms, Experimental/immunology , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Colorectal Neoplasms/diet therapy , Colorectal Neoplasms/immunology , Endoribonucleases/genetics , Female , Lymphocyte Depletion , Lymphoma/diet therapy , Lymphoma/immunology , Melanoma, Experimental/diet therapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , RNA Helicases/metabolism , Signal Transduction
5.
Anticancer Res ; 37(7): 3461-3471, 2017 07.
Article En | MEDLINE | ID: mdl-28668835

BACKGROUND/AIM: Tumor-associated macrophages (TAMs), together with splenic CD11b+ cells, help maintain the tumor microenvironment. The immunomodulatory compound imiquimod (IQM) stimulates innate immune cells, including macrophages, to induce antitumor effects. In order to elucidate the effects of IQM on the tumor microenvironment, we investigated the immunomodulatory effect of IQM during melanoma growth by using the B16F10 melanoma model. MATERIALS AND METHODS: To elucidate the immunomodulatory effects of IQM on the tumor microenvironment, we isolated CD11b+ TAMs and splenic CD11b+ cells and evaluated the immunomodulatory effects of IQM, using the B16F10 melanoma model. RESULTS: IQM suppressed B16F10 melanoma growth in parallel with reduction of Foxp3+ regulatory T cells (Tregs) at the tumor site, caused by the down-regulation of CCL22 production by tumor-derived and splenic CD11b+ cells. Subsequently, we investigated the antitumor or tumor-loading effects of splenic CD11b+ cells on B16F10 melanoma growth in vivo. B16F10 melanoma growth was accelerated by splenic CD11b+ cells from untreated mice, but was inhibited by splenic CD11b+ cells from IQM-treated mice. Consistent with these results, Foxp3+ Tregs were significantly decreased in tumors of mice implanted with both melanoma and splenic CD11b+ cells from topical IQM-treated mice. Furthermore, intratumoral administration of anti-CCL22 antibody inhibited B16F10 melanoma growth by decreasing Treg recruitment at the tumor site. CONCLUSION: Our results suggest a possible mechanism for the antitumor immune response induced by IQM through tumor-associated macrophages.


Aminoquinolines/immunology , Aminoquinolines/pharmacology , Chemokine CCL22/metabolism , Immunologic Factors/immunology , Macrophages/drug effects , Melanoma, Experimental/diet therapy , Melanoma, Experimental/metabolism , Animals , CD11b Antigen/immunology , CD11b Antigen/metabolism , Cell Line, Tumor , Chemokine CCL22/immunology , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Imiquimod , Immunologic Factors/pharmacology , Macrophages/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
Biomed Res Int ; 2015: 746093, 2015.
Article En | MEDLINE | ID: mdl-25839040

The objective of this study was the synthesis of folic acid- (FA-) targeted polymeric micelles of Synperonic PE/F 127-cholesteryl hemisuccinate (PF127-Chol) for specific delivery of docetaxel (DTX). Targeted or nontargeted micelles loaded with DTX were prepared via dialysis method. The effects of processing variables on the physicochemical properties of targeted micelles were evaluated using a full factorial design. After the optimization of the polymer/drug ratio, the organic solvent type used for the preparation of the micelles, and the temperature of dialyzing medium, the in vitro cytotoxicity and cellular uptake of the optimized micelles were studied on B16F10 melanoma cells by flow cytometry and fluorescent microscopy. The anticancer efficacy of DTX-loaded FA-PF127-Chol was evaluated in mice bearing melanoma tumor. Optimized targeted micelles had the particle size of 171.3 nm, zeta potential of -7.8 mV, PDI of 0.325, and a high encapsulation efficiency that released the drug within 144 h. The MTT assay indicated that targeted micelles carrying DTX were significantly more cytotoxic, had higher cellular uptake, and reduced the tumor volume significantly more than the nontargeted micelles and the free drug. FA-PF127-Chol could be, therefore, a promising biomaterial for tumors overexpressing folate receptors.


Antineoplastic Agents/chemistry , Cholesterol Esters/chemistry , Drug Carriers/chemistry , Folic Acid/chemistry , Melanoma, Experimental/diet therapy , Polymers/chemistry , Taxoids/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Docetaxel , Drug Delivery Systems/methods , Hep G2 Cells , Humans , Mice , Micelles , Particle Size , Taxoids/pharmacology
7.
Biomed Res Int ; 2015: 479798, 2015.
Article En | MEDLINE | ID: mdl-25861631

Specific phenol compounds including rhododendrol (RD), a skin-brightening ingredient in cosmetics, are reported to induce leukoderma, inducing a social problem, and the elucidation of mechanism of leukoderma is strongly demanded. This study investigated the relationship among the cytotoxicities of six phenol compounds on B16F10 melanoma cells and HaCaT keratinocytes and generated reactive oxygen species (ROS). As a result, the cytotoxicity of RD on B16F10 cells was higher than that on HaCaT cells, and RD significantly increased intracellular ROS and hydrogen peroxide (H2O2) levels in B16F10 cells. Furthermore, although raspberry ketone (RK), RD derivative, also increased intracellular ROS in B16F10 cells, increase in ROS was suppressed by disodium dihydrogen ethylenediaminetetraacetate dehydrate (EDTA). The amounts of increased ROS with RK in HaCaT cells without melanocyte were further increased by tyrosinase. Therefore, tyrosinase, a metalloprotein having copper, was speculated to be one of causative agents allowing phenol compounds to work as a prooxidant. Hydroxyl radical was generated by adding a mixture of tyrosinase and H2O2 to RD, and the amount of the radical was further increased by UVB, indicating that RD cytotoxicity was caused by intracellularly increased ROS, which possibly related to phenol induced prooxidants.


Hydrogen Peroxide/pharmacology , Melanocytes/drug effects , Phenols/pharmacology , Cell Line , Cell Line, Tumor , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Melanocytes/metabolism , Melanoma, Experimental/diet therapy , Monophenol Monooxygenase/metabolism , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/metabolism , Ultraviolet Rays
8.
Molecules ; 19(9): 12940-8, 2014 Aug 25.
Article En | MEDLINE | ID: mdl-25157464

Chlorogenic acid (CGA), the ester formed between caffeic acid and l-quinic acid, is a widespread phenolic compound. It is part of the human diet, found in foods such as coffee, apples, pears, etc. CGA is also was widely used in cosmetics, but the effects of CGA on melanogenesis are unknown. In this study, we analyzed the effects of CGA on cell proliferation, melanin content and tyrosinase of B16 murine melanoma cells. Additionally, the enzymatic reactions of CGA in B16 melanoma cells lytic solution were detected by UV spectrophotometry. Results showed CGA at 30 and 60 µM significantly suppresses cell proliferation. 8-MOP at 100 µM significantly promotes cell proliferation, but CGA can counter this. Incubated for 24 h, CGA (500 µM) improves melanogenesis while suppressing tyrosinase activity in B16 melanoma cells or 8-methoxypsoralen (8-MOP) co-incubated B16 melanoma cells. After 12 h, B16 melanoma cell treatment with CGA leads to an increase in melanin accumulation, however, after 48 h there is a decrease in melanin production which correlates broadly with a decrease in tyrosinase activity. CGA incubated with lytic solution 24 h turned brown at 37 °C. The formation of new products (with a maximum absorption at 295 nm) is associated with reduction of CGA (maximum absorption at 326 nm). Therefore, CGA has its two sidesroles in melanogenesis of B16 melanoma cells. CGA is a likely a substrate of melanin, but the metabolic product(s) of CGA may suppress melanogenesis in B16 melanoma cells by inhibiting tyrosinase activity.


Chlorogenic Acid/administration & dosage , Melanins/biosynthesis , Melanoma, Experimental/diet therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Humans , Melanoma, Experimental/pathology , Mice , Monophenol Monooxygenase/metabolism , Quinic Acid/metabolism
9.
Int Immunopharmacol ; 21(2): 494-500, 2014 Aug.
Article En | MEDLINE | ID: mdl-24953854

Inflammation is closely associated with tumor growth, which is mediated by the activation of bone marrow-derived CD11b(+) cells. Here, we investigated whether anti-inflammatory dexamethasone (Dex), a synthetic glucocorticoid (GC), could regulate tumor growth and CD11b(+) myeloid bone marrow cells (BMCs) in lymphocyte (R1), monocyte (R2) and granulocyte (R3) regions of FSC-SSC dot plot. The growth of B16F10 mouse melanoma tumor was inhibited in Dex-injected group. Lung metastasis was decreased and the lifespan was elongated in Dex-injected mice with tumor resection. Intravenous injection of B16F10 cells increased the percentage of CD11b(+) myeloid BMCs in R1 and R2 regions from 3h to 72h. In contrast, little changes in the percentage of CD11b(+) myeloid BMCs were detected in R3 region. Among CD11b(+) myeloid BMCs, the percentage of CD11b(+)Gr-1(+) cells was increased in R1, R2 and R3 regions. Absolute number of CD11b(+) and CD11b(+)Gr-1(+) cells was enhanced in R1 region from 3h to 72 h. B16F10 tumor growth was significantly increased by intravenous injection of CD11b(+) BMCs. Tumor-bearing mice showed an increase in the percentage of CD11b(+) myeloid BMCs in R2 region and CD11b(+)Gr-1(+) cells in R2 and R3 regions, which are reduced by intravenous injection with Dex. Absolute number of CD11b(+)Gr-1(+) cells was enhanced in R2 and R3 regions. Tumor growth was significantly inhibited by intravenous injection of BMCs collected from Dex-treated tumor-bearing mice. Taken together, data demonstrate that tumor regression by Dex was resulted from the alteration of CD11b(+) myeloid BMCs and their inhibitory function to tumor growth. It suggests that CD11b(+) myeloid BMCs could regulate antitumor efficacy of GCs such as Dex.


Bone Marrow Cells/drug effects , Bone Marrow/drug effects , CD11b Antigen/metabolism , Dexamethasone/pharmacology , Myeloid Cells/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Female , Granulocytes/drug effects , Granulocytes/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Melanoma, Experimental/diet therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Myeloid Cells/metabolism , Receptors, Chemokine/metabolism
10.
Invest New Drugs ; 32(4): 587-97, 2014 Aug.
Article En | MEDLINE | ID: mdl-24691673

We previously selected two melanin-targeting radioligands [(125)I]ICF01035 and [(125)I]ICF01040 for melanoma-targeted (125)I radionuclide therapy according to their pharmacological profile in mice bearing B16F0 tumors. Here we demonstrate in vitro that these compounds present different radiotoxicities in relation to melanin and acidic vesicle contents in B16F0, B16F0 PTU and A375 cell lines. ICF01035 is effectively observed in nuclei of achromic (A375) melanoma or in melanosomes of melanized melanoma (B16F0), while ICF01040 stays in cytoplasmic vesicles in both cells. [(125)I]ICF01035 induced a similar survival fraction (A50) in all cell lines and led to a significant decrease in S-phase cells in amelanotic cell lines. [(125)I]ICF01040 induced a higher A50 in B16 cell lines compared to [(125)I]ICF01035 ones. [(125)I]ICF01040 induced a G2/M blockade in both A375 and B16F0 PTU, associated with its presence in cytoplasmic acidic vesicles. These results suggest that the radiotoxicity of [(125)I]ICF01035 and [(125)I]ICF01040 are not exclusively reliant on DNA alterations compatible with γ rays but likely result from local dose deposition (Auger electrons) leading to toxic compound leaks from acidic vesicles. In vivo, [(125)I]ICF01035 significantly reduced the number of B16F0 lung colonies, enabling a significant increase in survival of the treated mice. Targeting melanosomes or acidic vesicles is thus an option for future melanoma therapy.


Acridines/administration & dosage , Iodine Radioisotopes/administration & dosage , Melanoma, Experimental/diet therapy , Melanoma, Experimental/drug therapy , Radiopharmaceuticals/administration & dosage , Acridines/metabolism , Animals , Cell Line, Tumor , Electrons , Humans , Iodine Radioisotopes/metabolism , Male , Mice , Mice, Inbred C57BL , Radiopharmaceuticals/metabolism
11.
J Surg Res ; 188(2): 451-8, 2014 May 15.
Article En | MEDLINE | ID: mdl-24576779

BACKGROUND: To determine the effect of docosahexaenoic acid (DHA) on the growth of human melanoma in vitro and in vivo and to better understand the potential role of the G protein-coupled receptors (GPRs) in mediating this effect. MATERIALS AND METHODS: For in vitro studies, human melanoma and control fibroblast cells were treated with DHA and TAK-875 (selective GPR40 agonist) and a cell viability assay was performed to determine cell counts. A murine subcutaneous xenograft model of human melanoma was used to test the effect of dietary treatment with an omega-3 fatty acid (FA) rich diet compared with an omega-6 FA rich diet on the growth of human melanoma in vivo. A similar animal model was used to test the effect of oral TAK-875 on the growth of established melanoma tumors in vivo. RESULTS: DHA has an inhibitory effect on the growth of human melanoma both in vitro and in vivo. Tumors from animals on the omega-3 FA rich diet were 69% smaller in weight (P = 0.005) and 76% smaller in volume compared with tumors from animals on the omega-6 FA rich diet. TAK-875 has an inhibitory effect on the growth of human melanoma both in vitro and in vivo. Tumors from animals treated with TAK-875 were 46% smaller in weight (P = 0.07), 62% smaller in volume (P = 0.03), and grew 77% slower (P = 0.04) compared with the placebo group. CONCLUSIONS: DHA and TAK-875 have a profound and selective inhibitory effect on the growth of human melanoma both in vitro and in vivo.


Benzofurans/therapeutic use , Docosahexaenoic Acids/therapeutic use , Melanoma, Experimental/diet therapy , Melanoma, Experimental/drug therapy , Receptors, G-Protein-Coupled/metabolism , Sulfones/therapeutic use , Animals , Benzofurans/pharmacology , Cell Line, Tumor , Docosahexaenoic Acids/pharmacology , Humans , Melanoma, Experimental/metabolism , Mice , Receptors, G-Protein-Coupled/agonists , Sulfones/pharmacology , Xenograft Model Antitumor Assays
12.
Nutr Cancer ; 61(4): 518-29, 2009.
Article En | MEDLINE | ID: mdl-19838924

Methionine (Met) deprivation stress (MDS) is proposed in association with chemotherapy in the treatment of some cancers. A synergistic effect of this combination is generally acknowledged. However, little is known on the mechanism of the response to this therapeutic strategy. A model of B16 melanoma tumor in vivo was treated by MDS alone and in combination with chloroethylnitrosourea (CENU). It was applied recent developments in proton-NMR spectroscopy-based metabolomics for providing information on the metabolic response of tumors to MDS and combination with chemotherapy. MDS inhibited tumor growth during the deprivation period and growth resumption thereafter. The combination of MDS with CENU induced an effective time-dependent synergy on growth inhibition. Metabolite profiling during MDS showed a decreased Met content (P < 0.01) despite the preservation of the protein content, disorders in sulfur-containing amino acids, glutamine/proline, and phospholipid metabolism [increase of glycerophosphorylcholine (P < 0.01), decrease in phosphocholine (P < 0.05)]. The metabolic profile of MDS combined with CENU and ANOVA analysis revealed the implication of Met and phospholipid metabolism in the observed synergy, which may be interpreted as a Met-sparing metabolic reprogramming of tumors. It follows that combination therapy of MDS with CENU seems to intensify adaptive processes, which may set limitations to this therapeutic strategy.


Adaptation, Physiological/drug effects , Antineoplastic Agents/therapeutic use , Melanoma, Experimental/therapy , Methionine/deficiency , Methionine/metabolism , Nitrosourea Compounds/therapeutic use , Phospholipids/metabolism , Analysis of Variance , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Combined Modality Therapy , Confidence Intervals , Growth Inhibitors/therapeutic use , Liver/drug effects , Male , Melanoma, Experimental/chemistry , Melanoma, Experimental/diet therapy , Melanoma, Experimental/metabolism , Metabolome/drug effects , Methionine/analysis , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Organ Size/drug effects , Phosphatidylethanolamine N-Methyltransferase/metabolism , Phospholipids/analysis , Random Allocation , Stress, Physiological/drug effects , Time Factors , Tumor Burden/drug effects
13.
J Cutan Pathol ; 36(10): 1053-62, 2009 Oct.
Article En | MEDLINE | ID: mdl-19187112

BACKGROUND: Diet seems to represent, directly or indirectly, 35% of all cancer reports. In this study, the influence of dietary protein on the growth of melanoma B16F10 was evaluated through analyses of cell cycle phases and proliferative capacity. METHODS: Flow cytometry and argyrophilic nucleolar organizer regions (AgNORs) technique were applied in mice bearing B16F10 melanoma cells fed on different dietary proteins. All data were submitted to statistical analyses. RESULTS: The G0/G1 phase increased for the animal groups fed bovine collagen hydrolysate (BCH) or BCH-P1 + whey protein isolate (WPI), compared with mice receiving only WPI, for all dietary groups treated and nontreated with paclitaxel. Mice that received BCH + WPI treated with paclitaxel showed the highest percentage of apoptosis compared with WPI group. AgNORs, total nucleolar organizer regions (NORs)/cells and dot number/cell for all dietary protein groups nontreated with paclitaxel were higher than for the WPI. The only two dietary protein groups treated with paclitaxel that presented higher total NORs and dot number/cell than the WPI group were BCH + WPI and BCH-P1 + WPI. CONCLUSIONS: A significantly lower proliferative capacity and larger number of cells in the G0/G1 phase were observed for the dietary protein groups combining the two collagen hydrolysates, BCH or BCH-P1 with WPI, treated with paclitaxel.


Cell Cycle/physiology , Dietary Proteins/pharmacology , Melanoma, Experimental/diet therapy , Melanoma, Experimental/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cattle , Cell Cycle/drug effects , Cell Proliferation/drug effects , Collagen/pharmacology , Diet , Melanoma, Experimental/drug therapy , Mice , Milk Proteins/pharmacology , Nucleolus Organizer Region/drug effects , Nucleolus Organizer Region/pathology , Paclitaxel/pharmacology , Whey Proteins
14.
Immunol Cell Biol ; 86(3): 277-88, 2008.
Article En | MEDLINE | ID: mdl-18268518

Bovine lactoferrin (bLf), an iron-containing natural defence protein found in bodily secretions, has been reported to inhibit carcinogenesis and the growth of tumours. Here, we investigated whether natural bLf and iron-saturated forms of bLf differ in their ability to augment cancer chemotherapy. bLf was supplemented into the diet of C57BL/6 mice that were subsequently challenged subcutaneously with tumour cells, and treated by chemotherapy. Chemotherapy eradicated large (0.6 cm diameter) EL-4 lymphomas in mice that had been fed iron-saturated bLf (here designated Lf(+)) for 6 weeks prior to chemotherapy, but surprisingly not in mice that were fed lesser iron-saturated forms of bLf, including apo-bLf (4% iron saturated), natural bLf (approximately 15% iron saturated) and 50% iron-saturated bLf. Lf(+)-fed mice bearing either EL-4, Lewis lung carcinoma or B16 melanoma tumours completely rejected their tumours within 3 weeks following a single injection of either paclitaxel, doxorubicin, epirubicin or fluorouracil, whereas mice fed the control diet were resistant to chemotherapy. Lf(+) had to be fed to mice for more than 2 weeks prior to chemotherapy to be wholly effective in eradicating tumours from all mice, suggesting that it acts as a competence factor. It significantly reduced tumour vascularity and blood flow, and increased antitumour cytotoxicity, tumour apoptosis and the infiltration of tumours by leukocytes. Lf(+) bound to the intestinal epithelium and was preferentially taken up within Peyer's patches. It increased the production of Th1 and Th2 cytokines within the intestine and tumour, including TNF, IFN-gamma, as well as nitric oxide that have been reported to sensitize tumours to chemotherapy. Importantly, it restored both red and white peripheral blood cell numbers depleted by chemotherapy, potentially fortifying the mice against cancer. In summary, bLf is a potent natural adjuvant and fortifying agent for augmenting cancer chemotherapy, but needs to be saturated with iron to be effective.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Lewis Lung/drug therapy , Lactoferrin/administration & dosage , Lymphoma/drug therapy , Melanoma, Experimental/drug therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Pharmaceutic/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/immunology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/diet therapy , Carcinoma, Lewis Lung/immunology , Cattle , Cytotoxicity, Immunologic/drug effects , Dietary Supplements , Drug Resistance, Neoplasm/drug effects , Immunohistochemistry , Iron/chemistry , Lactoferrin/chemistry , Lactoferrin/immunology , Lymphoma/diet therapy , Lymphoma/immunology , Melanoma, Experimental/blood supply , Melanoma, Experimental/diet therapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy
15.
Biochem Pharmacol ; 69(12): 1693-704, 2005 Jun 15.
Article En | MEDLINE | ID: mdl-15935145

Bovine serum amine oxidase (BSAO) oxidatively deaminates polyamines containing primary amine groups, spermidine and spermine, to form the cytotoxic products hydrogen peroxide and aldehyde(s). Polyamines are present at elevated levels in many tumor tissues. The aims of the study were to evaluate the anti-tumoral activities of native and immobilized BSAO in mouse melanoma and also to determine the mechanism of tumor cell death. C57BL mice received a subcutaneous injection of B16 melanoma cells to induce formation of tumors, prior to antitumor treatments with native and immobilized BSAO. The enzyme was immobilized in a poly(ethylene glycol) (PEG) biocompatible matrix. Antitumor treatments consisted of a single injection of enzyme into the tumor. When immobilized BSAO (2.5mU) was injected into the tumor, there was a marked decrease of 70% of the tumor growth. This was compared with a decrease of only 32% of tumor size when the same amount of native BSAO was administered. The type of cell death was analysed in tumors that were treated with native or immobilized BSAO. When tumors were treated with immobilized BSAO, there was induction of a high level of apoptosis (around 70%), compared to less than 10% with the native enzyme. Apoptotic cell death was assessed by nuclear chromatin condensation using Hoechst staining and labelling of externalized phosphatidylserine using Annexin V. However, native BSAO, probably due to a burst of cytotoxic products, induced a high level of necrosis of about 40%, compared to less than 10% with immobilized BSAO. In conclusion, the advantage is that immobilized BSAO can act by allowing the slow release of cytotoxic products, which induces tumor cell death by apoptosis rather than necrosis.


Amine Oxidase (Copper-Containing)/blood , Amine Oxidase (Copper-Containing)/therapeutic use , Antineoplastic Agents/therapeutic use , Enzymes, Immobilized/therapeutic use , Growth Inhibitors/therapeutic use , Melanoma, Experimental/drug therapy , Animals , Cattle , Cell Death/drug effects , Cell Line, Tumor , Dietary Supplements/adverse effects , Drug Screening Assays, Antitumor/methods , Female , Melanoma, Experimental/diet therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Skin/drug effects , Skin/injuries , Skin/pathology , Spermine/adverse effects , Spermine/therapeutic use
16.
Cancer Lett ; 124(2): 181-6, 1998 Feb 27.
Article En | MEDLINE | ID: mdl-9500208

The present study investigated the effect of dietary supplementation of flaxseed, the richest source of lignans, on experimental metastasis of B16BL6 murine melanoma cells in C57BL/6 mice. Mice were fed a basal diet or the basal diet supplemented with 2.5, 5 or 10% flaxseed for 2 weeks before and after the intravenous injection of 0.75 x 10(5) melanoma cells. At necropsy, the number of tumors that developed in the lungs was counted, the cross-sectional area of tumors was measured and the volumes of tumors were calculated. The median number of tumors in mice fed the 2.5, 5 and 10% flaxseed-supplemented diets was 32, 54 and 63% lower than that of the controls, respectively. The addition of flaxseed to the diet also caused a dose-dependent decrease in the tumor cross-sectional area and the tumor volume. These results provide the first experimental evidence that flaxseed reduces metastasis and inhibits the growth of the metastatic secondary tumors in animals. It is concluded that flaxseed may be a useful nutritional adjuvant to prevent metastasis in cancer patients.


Dietary Supplements , Flax , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/diet therapy , Melanoma, Experimental/secondary , Animals , Disease Models, Animal , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplastic Cells, Circulating/pathology , Tumor Cells, Cultured
17.
Nutr Cancer ; 29(1): 1-6, 1997.
Article En | MEDLINE | ID: mdl-9383777

The purpose of the present study was to determine the effect of dietary supplementation of soybean protein isolate (SPI) on experimental metastasis of B16BL6 murine melanoma cells in C57BL/6 mice. Four groups of mice were fed a basal AIN-93G diet or the basal diet supplemented with 10%, 15%, or 20% SPI for two weeks before and after the intravenous injection of 0.75 x 10(5) cells. At necropsy the number of tumors that developed in the lungs and their cross-sectional area were determined, and tumor volume was calculated. In the control group, 12 of the 15 mice had > or = 11 lung tumors. In contrast, only 3 or 4 of the 15 mice fed the SPI diets had > or = 11 tumors. The incidence of metastasis was 93%, 60%, 53%, and 53%, and the median number of lung tumors was 53, 2, 2, and 1 in mice fed the basal, 10%, 15%, and 20% SPI diets, respectively. Tumor cross-sectional area and tumor volume of SPI groups were significantly decreased compared with the controls. These results demonstrate that dietary supplementation of SPI reduced pulmonary metastasis of B16BL6 cells in mice and inhibited the growth of tumors that developed in the lungs. It is concluded that soybeans may be a useful adjuvant for preventing metastatic diseases in cancer patients.


Dietary Supplements , Glycine max , Melanoma, Experimental/diet therapy , Animals , Injections, Intravenous , Male , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Tumor Cells, Cultured
18.
Adv Exp Med Biol ; 354: 171-83, 1994.
Article En | MEDLINE | ID: mdl-7520663

We have shown that Tyr and Phe restriction suppresses the malignant phenotype of the highly invasive and metastatic BL6 variant of B16 murine melanoma. Lung-colonizing abilities of Tyr- and Phe-modulated in vivo and in vitro variants of BL6 are inhibited following intravenous inoculation into mice fed normal diet. Although this antimetastatic effect of Tyr and Phe restriction is most likely not due to differences in attachment to endothelium, our data indicate that major impacts of Tyr and Phe restriction are at the level of the tumor, itself. Modulation of host immune responses, which in turn suppresses metastasis, does not appear to contribute significantly to the altered phenotype. Although numbers and function of T cells, mast cells, and NK cells are affected by Tyr and Phe restriction, they are not involved in the Tyr- and Phe-mediated suppression of tumor growth, metastasis, or angiogenesis. Our data do not rule out the importance of other host factors involved in the Tyr and Phe modulation of tumor phenotype. The outcome of this modulation results most likely from complex Tyr/Phe-tumor-host interactions.


Animal Nutritional Physiological Phenomena , Melanoma, Experimental/diet therapy , Phenylalanine/administration & dosage , Tyrosine/administration & dosage , Animals , Diet , Female , Killer Cells, Natural/physiology , Lung/pathology , Mast Cells/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic , Phenotype , Tumor Cells, Cultured
19.
Anticancer Res ; 13(2): 523-8, 1993.
Article En | MEDLINE | ID: mdl-8517667

We previously showed that restriction of tyrosine (Tyr) and phenylalanine (Phe) in vivo dramatically suppresses the metastatic phenotype of B16-BL6 (BL6) murine melanoma. Present results indicate a direct effect of Tyr and Phe restriction on the tumor in the absence of host selection pressures. Lung colonizing ability of BL6 is dramatically suppressed after one passage in vitro in media containing low levels of Tyr and Phe. This antimetastatic effect is immediate, stable for at least 5 in vitro passages in Tyr and Phe restricted media, and evident event after levels of Tyr and Phe are restored to normal. Heterogeneity for lung colonizing ability is suppressed, as evidence by fewer tumor colonies formed by clones following i.v. inoculation into mice fed normal diet. This suppression of BL6 metastatic phenotype is not due to differential clearance and retention in the lung or to decreased growth, but is specific for these two amino acids. As the mechanism(s) for the antitumor effects of Tyr and Phe restriction are detailed, the relevance of Tyr and Phe restriction as an early adjuvant to effective cancer treatment can be explored.


Lung Neoplasms/secondary , Melanoma, Experimental/diet therapy , Melanoma, Experimental/secondary , Phenylalanine/deficiency , Tyrosine/deficiency , Animals , Cell Division/drug effects , Cell Division/physiology , Diet , Lung Neoplasms/diet therapy , Mice , Phenotype , Phenylalanine/therapeutic use , Tyrosine/therapeutic use
...