Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Oncol ; 9: 25, 2019.
Article in English | MEDLINE | ID: mdl-30740361

ABSTRACT

Microtubules are important drug targets in tumor cells, owing to their role in supporting and determining the cell shape, organelle movement and cell division. The complementarity-determining regions (CDRs) of immunoglobulins have been reported to be a source of anti-tumor peptide sequences, independently of the original antibody specificity for a given antigen. We found that, the anti-Lewis B mAb light-chain CDR1 synthetic peptide Rb44, interacted with microtubules and induced depolymerization, with subsequent degradation of actin filaments, leading to depolarization of mitochondrial membrane-potential, increase of ROS, cell cycle arrest at G2/M, cleavage of caspase-9, caspase-3 and PARP, upregulation of Bax and downregulation of Bcl-2, altogether resulting in intrinsic apoptosis of melanoma cells. The in vitro inhibition of angiogenesis was also an Rb44 effect. Peritumoral injection of Rb44L1 delayed growth of subcutaneously grafted melanoma cells in a syngeneic mouse model. L1-CDRs from immunoglobulins and their interactions with tubulin-dimers were explored to interpret effects on microtubule stability. The opening motion of tubulin monomers allowed for efficient L1-CDR docking, impairment of dimer formation and microtubule dissociation. We conclude that Rb44 VL-CDR1 is a novel peptide that acts on melanoma microtubule network causing cell apoptosis in vitro and melanoma growth inhibition in vivo.

2.
FEBS Open Bio ; 6(9): 885-901, 2016 09.
Article in English | MEDLINE | ID: mdl-27642552

ABSTRACT

Antibody-derived peptides modulate functions of the immune system and are a source of anti-infective and antitumor substances. Recent studies have shown that they comprise amino acid sequences of immunoglobulin complementarity-determining regions, but also fragments of constant regions. VH CDR3 of murine mAb AC-1001 displays antimetastatic activities using B16F10-Nex2 murine melanoma cells in a syngeneic model. The peptide was cytotoxic in vitro in murine and human melanoma cells inducing reactive oxygen species (ROS) and apoptosis by the intrinsic pathway. Signs of autophagy were also suggested by the increased expression of LC3/LC3II and Beclin 1 and by ultrastructural evidence. AC-1001 H3 bound to both G- and F-actin and inhibited tumor cell migration. These results are important evidence of the antitumor activity of Ig CDR-derived peptides.

3.
Peptides ; 85: 1-15, 2016 11.
Article in English | MEDLINE | ID: mdl-27575453

ABSTRACT

The present work aims at investigating the mechanism of action of the Rb9 peptide, which contains the VHCDR 3 sequence of anti-sodium-dependent phosphate transport protein 2B (NaPi2B) monoclonal antibody RebMab200 and displayed antitumor properties. Short peptides corresponding to the hypervariable complementarity-determining regions (CDRs) of immunoglobulins have been associated with antimicrobial, antiviral, immunomodulatory and antitumor activities regardless of the specificity of the antibody. We have shown that the CDR derived peptide Rb9 induced substrate hyperadherence, inhibition of cell migration and matrix invasion in melanoma and other tumor cell lines. Rb9 also inhibited metastasis of murine melanoma in a syngeneic mouse model. We found that Rb9 binds to and interferes with Hsp90 chaperone activity causing attenuation of FAK-Src signaling and downregulation of active Rac1 in B16F10-Nex2 melanoma cells. The peptide also bound to an adhesion G-protein coupled receptor, triggering a concentration-dependent synthesis of cAMP and activation of PKA and VASP signaling as well as IP-3 dependent Ca2+ release. Hsp90 is highly expressed on the cell surface of melanoma cells, and synthetic agents that target Hsp90 are promising cancer therapeutic drugs. Based on their remarkable antitumor effects, the CDR-H3-derived peptides from RebMab200, and particularly the highly soluble and stable Rb9, are novel candidates to be further studied as potential antitumor drugs, selectively acting on cancer cell motility and invasion.


Subject(s)
Complementarity Determining Regions/genetics , HSP90 Heat-Shock Proteins/genetics , Melanoma, Experimental/drug therapy , Peptides/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Complementarity Determining Regions/immunology , HSP90 Heat-Shock Proteins/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Neoplasm Invasiveness/genetics , Neuropeptides/genetics , Peptides/administration & dosage , Peptides/immunology , Receptors, G-Protein-Coupled/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/immunology , rac1 GTP-Binding Protein/genetics
4.
Sci Rep ; 5: 14310, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26391685

ABSTRACT

Short peptide sequences from complementarity-determining regions (CDRs) of different immunoglobulins may exert anti-infective, immunomodulatory and antitumor activities regardless of the specificity of the original monoclonal antibody (mAb). In this sense, they resemble early molecules of innate immunity. C36L1 was identified as a bioactive light-chain CDR1 peptide by screening 19 conserved CDR sequences targeting murine B16F10-Nex2 melanoma. The 17-amino acid peptide is readily taken up by melanoma cells and acts on microtubules causing depolymerization, stress of the endoplasmic reticulum and intrinsic apoptosis. At low concentrations, C36L1 inhibited migration, invasion and proliferation of B16F10-Nex2 cells with cell cycle arrest at G2/M phase, by regulating the PI3K/Akt signaling axis involving Rho-GTPase and PTEN mediation. Peritumor injection of the peptide delayed growth of subcutaneously grafted melanoma cells. Intraperitoneal administration of C36L1 induced a significant immune-response dependent anti-tumor protection in a syngeneic metastatic melanoma model. Dendritic cells stimulated ex-vivo by the peptide and transferred to animals challenged with tumor cells were equally effective. The C36 VL CDR1 peptide is a promising microtubule-interacting drug that induces tumor cell death by apoptosis and inhibits metastases of highly aggressive melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Complementarity Determining Regions/chemistry , Melanoma/metabolism , Melanoma/pathology , Microtubules/metabolism , Peptides/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Melanoma/drug therapy , Melanoma/immunology , Melanoma, Experimental , Mice , Microtubules/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Metastasis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tubulin/chemistry , Tubulin/metabolism , rho GTP-Binding Proteins/metabolism
5.
Molecules ; 20(7): 12757-68, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26184150

ABSTRACT

Nectandra megapotamica (Spreng.) Mez. (Lauraceae) is a well-known Brazilian medicinal plant that has been used in folk medicine to treat several diseases. In continuation of our ongoing efforts to discover new bioactive natural products from the Brazilian flora, this study describes the identification of cytotoxic compounds from the MeOH extract of N. megapotamica (Lauraceae) leaves using bioactivity-guided fractionation. This approach resulted in the isolation and characterization of eight tetrahydrofuran neolignans: calopeptin (1), machilin-G (2), machilin-I (3), aristolignin (4), nectandrin A (5), veraguensin (6), ganschisandrin (7), and galgravin (8). Different assays were conducted to evaluate their cytotoxic activities and to determine the possible mechanism(s) related to the activity displayed against human leukemia cells. The most active compounds 4, 5 and 8 gave IC50 values of 14.2 ± 0.7, 16.9 ± 0.8 and 16.5 ± 0.8 µg/mL, respectively, against human leukemia (HL-60) tumor cells. Moreover, these compounds induced specific apoptotic hallmarks, such as plasma membrane bleb formation, nuclear DNA condensation, specific chromatin fragmentation, phosphatidyl-serine exposure on the external leaflet of the plasma membrane, cleavage of PARP as well as mitochondrial damage, which as a whole could be related to the intrinsic apoptotic pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cytotoxins/pharmacology , Lauraceae/chemistry , Lignans/pharmacology , Plant Leaves/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Brazil , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Survival/drug effects , Cytotoxins/chemistry , Cytotoxins/isolation & purification , DNA Fragmentation/drug effects , HL-60 Cells , HeLa Cells , Humans , Inhibitory Concentration 50 , Lignans/chemistry , Lignans/isolation & purification , MCF-7 Cells , Melanoma, Experimental , Mice , Organ Specificity , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Plant Extracts/chemistry , Plants, Medicinal , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Proteolysis , Structure-Activity Relationship
6.
Peptides ; 68: 113-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25305549

ABSTRACT

Mastoparan is an α-helical and amphipathic tetradecapeptide obtained from the venom of the wasp Vespula lewisii. This peptide exhibits a wide variety of biological effects, including antimicrobial activity, increased histamine release from mast cells, induction of a potent mitochondrial permeability transition and tumor cell cytotoxicity. Here, the effects of mastoparan in malignant melanoma were studied using the murine model of B16F10-Nex2 cells. In vitro, mastoparan caused melanoma cell death by the mitochondrial apoptosis pathway, as evidenced by the Annexin V-FITC/PI assay, loss of mitochondrial membrane potential (ΔΨm), generation of reactive oxygen species, DNA degradation and cell death signaling. Most importantly, mastoparan reduced the growth of subcutaneous melanoma in syngeneic mice and increased their survival. The present results show that mastoparan induced caspase-dependent apoptosis in melanoma cells through the intrinsic mitochondrial pathway protecting the mice against tumor development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Melanoma, Experimental/drug therapy , Peptides/pharmacology , Wasp Venoms/pharmacology , Animals , Cell Line, Tumor , Cell Shape/drug effects , Drug Screening Assays, Antitumor , Intercellular Signaling Peptides and Proteins , Male , Melanoma, Experimental/pathology , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Neoplasm Transplantation , Oxidative Stress , Tumor Burden/drug effects
7.
J Immunol Methods ; 414: 11-9, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25205401

ABSTRACT

On searching for melanoma transcription factors in a project focusing on internal antitumor peptide sequences from transcription factors, we found that a highly immunogenic component emerged upon using a subtraction tolerization method of immunization. While several conventional immunization procedures using whole melanoma cells induced a plethora of low affinity antibodies of various specificities, the subtraction tolerization method efficiently elicited mono-specific antibodies that recognized Wilms' tumor protein 1 (WT1), which is known as an important marker in melanoma prognosis and treatment. For the tolerization step, pre-immunization of Balb/c mice with a membrane-rich preparation of glioblastoma U87 cells was used. The subsequent immunizations with SK-MEL-28 melanoma cells elicited antibodies strongly reacting with 50 and 55 kDa proteins, identified as WT1. Remarkably, this was the only component strongly reactive with these antibodies in a melanoma cell lysate. WT1 was then chosen as a target for selecting internally bioactive peptides. A hydrophilic Trojan peptide containing most of the zinc finger-2 domain of WT1 was synthesized and shown to inhibit SK-MEL-28 melanoma growth in vitro. The peptide WT1-pTj was also protective in vivo in a metastatic melanoma model and peptide-stimulated syngeneic dendritic cells reproduced the anti-melanoma effect of the unprotected peptide. Identification of antitumor peptides derived from major transcription factors represents a new tool to be explored in cancer research aiming at new therapeutic drugs.


Subject(s)
Antibodies, Neoplasm/immunology , Cancer Vaccines/immunology , Melanoma, Experimental/immunology , WT1 Proteins/immunology , Animals , Biomarkers, Tumor/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/immunology , Female , Humans , Immune Tolerance/immunology , Immunization/methods , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Peptides/immunology , Peptides/therapeutic use , WT1 Proteins/therapeutic use
8.
Pharmacogn Mag ; 10(Suppl 2): S363-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24991116

ABSTRACT

BACKGROUND: Pyrostegia venusta (Ker. Gawl.) Miers (Bignoniacea) is a medicinal plant from the Brazilian Cerrado used to treat leucoderma and common diseases of the respiratory system. OBJECTIVE: To investigate the antitumor activity of P.venusta extracts against melanoma. MATERIALS AND METHODS: The cytotoxic activity and tumor induced cell death of heptane extract (HE) from P. venusta flowers was evaluated against murine melanoma B16F10-Nex2 cells in vitro and in a syngeneic model in vivo. RESULTS: We found that HE induced apoptosis in melanoma cells by disruption of the mitochondrial membrane potential, induction of reactive oxygen species and late apoptosis evidenced by plasma membrane blebbing, cell shrinkage, chromatin condensation and DNA fragmentation, exposure of phosphatidylserine on the cell surface and activation of caspase-2,-3,-8,-9. HE was also protective against singeneyc subcutaneous melanoma HE compounds were also able to induce cell cycle arrest at G2/M phases on tumor cells. On fractionation of HE in silica gel we isolated a cytotoxic fraction that contained a mixture of saturated hydrocarbons identified by (1)H NMR and GC-MS analyses. Predominant species were octacosane (C28H58-36%) and triacontane (C30H62-13%), which individually showed significant cytotoxic activity against murine melanoma B16F10-Nex2 cells in vitro and a very promising antitumor protection against subcutaneous melanoma in vivo. CONCLUSION: The results suggest that the components of the heptane extract, mainly octasane and triacontane, which showed antitumor properties in experimental melanoma upon regional administration, might also be therapeutic in human cancer, such as in the mostly epidermal and slowly invasive melanomas, such as acral lentiginous melanoma, as an adjuvant treatment to surgical excision.

9.
Peptides ; 59: 14-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24972300

ABSTRACT

Short synthetic peptides corresponding to sequences of complementarity-determining regions (CDRs) from different immunoglobulin families have been shown to induce antimicrobial, antiviral and antitumor activities regardless of the specificity of the original monoclonal antibody (mAb). Presently, we studied the in vitro and in vivo antitumor activity of synthetic peptides derived from conserved CDR sequences of different immunoglobulins against human tumor cell lines and murine B16F10-Nex2 melanoma aiming at the discovery of candidate molecules for cancer therapy. Four light- and heavy-chain CDR peptide sequences from different antibodies (C36-L1, HA9-H2, 1-H2 and Mg16-H2) showed cytotoxic activity against murine melanoma and a panel of human tumor cell lineages in vitro. Importantly, they also exerted anti-metastatic activity using a syngeneic melanoma model in mice. Other peptides (D07-H3, MN20v1, MS2-H3) were also protective against metastatic melanoma, without showing significant cytotoxicity against tumor cells in vitro. In this case, we suggest that these peptides may act as immune adjuvants in vivo. As observed, peptides induced nitric oxide production in bone-marrow macrophages showing that innate immune cells can also be modulated by these CDR peptides. The present screening supports the search in immunoglobulins of rather frequent CDR sequences that are endowed with specific antitumor properties and may be candidates to be developed as anti-cancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/pharmacology , Immunoglobulins/chemistry , Melanoma/drug therapy , Peptides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Complementarity Determining Regions/immunology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Immunoglobulins/immunology , Immunomodulation , MCF-7 Cells , Melanoma/immunology , Melanoma/pathology , Mice , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
10.
FEBS Open Bio ; 4: 153-61, 2014.
Article in English | MEDLINE | ID: mdl-24490140

ABSTRACT

The Wilms tumor protein 1 (WT1) transcription factor has been associated in malignant melanoma with cell survival and metastasis, thus emerging as a candidate for targeted therapy. A lysine-arginine rich peptide, WT1-pTj, derived from the ZF domain of WT1 was evaluated as an antitumor agent against A2058 human melanoma cells and B16F10-Nex2 syngeneic murine melanoma. Peptide WT1-pTj quickly penetrated human melanoma cells and induced senescence, recognized by increased SA-ß-galactosidase activity, enhanced transcriptional activity of p53, and induction of the cell cycle inhibitors p21 and p27. Moreover, the peptide bound to p53 and competed with WT1 protein for binding to p53. WT1-pTj treatment led to sustained cell growth suppression, abrogation of clonogenicity and G2/M cell cycle arrest. Notably, in vivo studies showed that WT1-pTj inhibited both the metastases and subcutaneous growth of murine melanoma in syngeneic mice, and prolonged the survival of nude mice challenged with human melanoma cells. The 27-amino acid cell-penetrating WT1-derived peptide, depends on C(3) and H(16) for effective antimelanoma activity, inhibits proliferation of WT1-expressing human tumor cell lines, and may have an effective role in the treatment of WT1-expressing malignancies.

11.
Clinics (Sao Paulo) ; 68(7): 1018-27, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23917669

ABSTRACT

OBJECTIVE: Available chemotherapy presents poor control over the development of metastatic melanoma. FTY720 is a compound already approved by the Food and Drug Administration for the treatment of patients with multiple sclerosis. It has also been observed that FTY720 inhibits tumor growth in vivo (experimental models) and in vitro (animal and human tumor cells). The aim of this study was to evaluate the effects of FTY720 on a metastatic melanoma model and in tumor cell lines. METHODS: We analyzed FTY720 efficacy in vivo in a syngeneic murine metastatic melanoma model, in which we injected tumor cells intravenously into C57BL/6 mice and then treated the mice orally with the compound for 7 days. We also treated mice and human tumor cell lines with FTY720 in vitro, and cell viability and death pathways were analyzed. RESULTS: FTY720 treatment limited metastatic melanoma growth in vivo and promoted a dose-dependent decrease in the viability of murine and human tumor cells in vitro. Melanoma cells treated with FTY720 exhibited characteristics of programmed cell death, reactive oxygen species generation, and increased ß-catenin expression. In addition, FTY720 treatment resulted in an immunomodulatory effect in vivo by decreasing the percentage of Foxp3+ cells, without interfering with CD8+ T cells or lymphocyte-producing interferon-gamma. CONCLUSION: Further studies are needed using FTY720 as a monotherapy or in combined therapy, as different types of cancer cells would require a variety of signaling pathways to be extinguished.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Immunosuppressive Agents/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Caspase 3/drug effects , Cell Line, Tumor , Cell Survival , Drug Screening Assays, Antitumor/methods , Fingolimod Hydrochloride , Flow Cytometry , Humans , Lung Neoplasms/secondary , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Reactive Oxygen Species , Sphingosine/therapeutic use , Time Factors
12.
Clinics ; 68(7): 1018-1027, jul. 2013. graf
Article in English | LILACS | ID: lil-680698

ABSTRACT

OBJECTIVE: Available chemotherapy presents poor control over the development of metastatic melanoma. FTY720 is a compound already approved by the Food and Drug Administration for the treatment of patients with multiple sclerosis. It has also been observed that FTY720 inhibits tumor growth in vivo (experimental models) and in vitro (animal and human tumor cells). The aim of this study was to evaluate the effects of FTY720 on a metastatic melanoma model and in tumor cell lines. METHODS: We analyzed FTY720 efficacy in vivo in a syngeneic murine metastatic melanoma model, in which we injected tumor cells intravenously into C57BL/6 mice and then treated the mice orally with the compound for 7 days. We also treated mice and human tumor cell lines with FTY720 in vitro, and cell viability and death pathways were analyzed. RESULTS: FTY720 treatment limited metastatic melanoma growth in vivo and promoted a dose-dependent decrease in the viability of murine and human tumor cells in vitro. Melanoma cells treated with FTY720 exhibited characteristics of programmed cell death, reactive oxygen species generation, and increased β-catenin expression. In addition, FTY720 treatment resulted in an immunomodulatory effect in vivo by decreasing the percentage of Foxp3+ cells, without interfering with CD8+ T cells or lymphocyte-producing interferon-gamma. CONCLUSION: Further studies are needed using FTY720 as a monotherapy or in combined therapy, as different types of cancer cells would require a variety of signaling pathways to be extinguished. .


Subject(s)
Animals , Humans , Male , Mice , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Immunosuppressive Agents/therapeutic use , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Blotting, Western , Cell Line, Tumor , Cell Survival , /drug effects , Drug Screening Assays, Antitumor/methods , Flow Cytometry , Lung Neoplasms/secondary , Microscopy, Electron, Transmission , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Reactive Oxygen Species , Sphingosine/therapeutic use , Time Factors
13.
PLoS One ; 7(6): e38698, 2012.
Article in English | MEDLINE | ID: mdl-22701695

ABSTRACT

BACKGROUND: Malignant melanoma is a deadly type of metastatic skin cancer with increased incidence over the past 30 years. Despite the advanced knowledge on the biology, immunobiology and molecular genetics of melanoma, the alternatives of treatment are limited with poor prognosis. On clinical trials, natural products and among them redox-active quinones have been tested in the attempt to control the growth of cancer cells. Recently, we isolated jacaranone from Pentacalia desiderabilis, a benzoquinone derivative that showed a broad antitumor activity and protective anti-melanoma effect in a syngeneic model. The purified substance is active at micromolar concentrations, is not hemolytic, and is not toxic in naïve mice. METHODOLOGY/PRINCIPAL FINDINGS: The jacaranone antitumor activity was shown against several human cancer cell lines in vitro. Moreover, the induction of apoptosis in murine melanoma cells and jacaranone antitumor activity in vivo, in a melanoma experimental model, were also shown. Jacaranone renders antiproliferative and proapoptotic responses in tumor cells, by acting on Akt and p38 MAPK signaling pathways through generation of reactive oxygen species (ROS). The free radical scavenger N-acetyl-cysteine (NAC) was able to completely suppress cell death induced by jacaranone as it blocked Akt downregulation, p38 MAPK activation as well as upregulation of proapoptotic Bax. Notably, treatment of melanoma growing subcutaneously in mice with jacaranone significantly extended the mean survival times in a dose-dependent manner. CONCLUSIONS/SIGNIFICANCE: The results provide evidence for the mechanisms of action of jacaranone and emphasize the potential use of this quinone for the treatment of melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Asteraceae/chemistry , Benzoquinones/pharmacology , Melanoma/drug therapy , Phytotherapy/methods , Plant Extracts/pharmacology , Signal Transduction/drug effects , Acetylcysteine , Animals , Annexin A5 , Antineoplastic Agents/analysis , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzoquinones/analysis , Benzoquinones/therapeutic use , Blotting, Western , Cell Line, Tumor , Chromatin/metabolism , Colorimetry , Down-Regulation , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nuclear Magnetic Resonance, Biomolecular , Propidium , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Superoxides , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Biochem Biophys Res Commun ; 411(2): 449-54, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21756878

ABSTRACT

Malignant melanoma is one the most aggressive types of cancer and its incidence has gradually increased in the last years, accounting for about 75% of skin cancer deaths. This poor prognosis results from the tumor resistance to conventional drugs mainly by deregulation of apoptotic pathways. The aim of this work was to investigate the cell death mechanism induced by α-pinene and its therapeutic application. Our results demonstrated that α-pinene was able to induce apoptosis evidenced by early disruption of the mitochondrial potential, production of reactive oxygen species, increase in caspase-3 activity, heterochromatin aggregation, DNA fragmentation and exposure of phosphatidyl serine on the cell surface. Most importantly, this molecule was very effective in the treatment of experimental metastatic melanoma reducing the number of lung tumor nodules. This is the first report on the apoptotic and antimetastatic activity of isolated α-pinene.


Subject(s)
Anacardiaceae/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Monoterpenes/therapeutic use , Skin Neoplasms/drug therapy , Animals , Bicyclic Monoterpenes , Cell Line, Tumor , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...