969 resultados para MURINE MELANOMA
Resumo:
T helper (Th) 9 cells are an important subpopulation of the CD4+ T helper cells. Due to their ability to secrete Interleukin-(IL-)9, Th9 cells essentially contribute to the expulsion of parasitic helminths from the intestinal tract but they play also an immunopathological role in the course of asthma. Recently, a beneficial function of Th9 cells in anti-tumor immune responses was published. In a murine melanoma tumor model Th9 cells were shown to enhance the anti-melanoma immune response via the recruitment of CD8+ T cells, dendritic cells and mast cells. In contrast to Th9 effector cells regulatory T cells (Tregs) are able to control an immune response with the aid of different suppressive mechanisms. Based on their ability to suppress an immune response Tregs are believed to be beneficial in asthma by diminishing excessive allergic reactions. However, concerning cancer they can have a detrimental function because Tregs inhibit an effective anti-tumor immune reaction. Thus, the analysis of Th9 suppression by Tregs is of central importance concerning the development of therapeutic strategies for the treatment of cancer and allergic diseases and was therefore the main objective of this PhD thesis.rnIn general it could be demonstrated that the development of Th9 cells can be inhibited by Tregs in vitro. The production of the lineage-specific cytokine IL-9 by developing Th9 cells was completely suppressed at a Treg/Th9 ratio of 1:1 on the transcriptional (qRT-PCR) as well as on the translational level (ELISA). In contrast, the expression of IRF4 that was found to strongly promote Th9 development was not reduced in the presence of Tregs, suggesting that IRF4 requires additional transcription factors to induce the differentiation of Th9 cells. In order to identify such factors, which regulate Th9 development and therefore represent potential targets for Treg-mediated suppressive mechanisms, a transcriptome analysis using “next-generation sequencing” was performed. The expression of some genes which were found to be up- or downregulated in Th9 cells in the presence of Tregs was validated with qRT-PCR. Time limitations prevented a detailed functional analysis of these candidate genes. Nevertheless, the analysis of the suppressive mechanisms revealed that Tregs probably suppress Th9 cells via the increase of the intracellular cAMP concentration. In contrast, IL-9 production by differentiated Th9 cells was only marginally affected by Tregs in vitro and in vivo analysis (asthma, melanoma model). Hence, Tregs represent very effective inhibitors of Th9 development whereas they have only a minimal suppressive influence on differentiated Th9 cells.rn
Resumo:
Da nicht-synonyme tumorspezifische Punktmutationen nur in malignen Geweben vorkommen und das veränderte Proteinprodukt vom Immunsystem als „fremd“ erkannt werden kann, stellen diese einen bisher ungenutzten Pool von Zielstrukturen für die Immuntherapie dar. Menschliche Tumore können individuell bis zu tausenden nicht-synonymer Punktmutationen in ihrem Genom tragen, welche nicht der zentralen Immuntoleranz unterliegen. Ziel der vorliegenden Arbeit war die Hypothese zu untersuchen, dass das Immunsystem in der Lage sein sollte, mutierte Epitope auf Tumorzellen zu erkennen und zu klären, ob auf dieser Basis eine wirksame mRNA (RNA) basierte anti-tumorale Vakzinierung etabliert werden kann. Hierzu wurde von Ugur Sahin und Kollegen, das gesamte Genom des murinen B16-F10 Melanoms sequenziert und bioinformatisch analysiert. Im Rahmen der NGS Sequenzierung wurden mehr als 500 nicht-synonyme Punktmutationen identifiziert, von welchen 50 Mutationen selektiert und durch Sanger Sequenzierung validiert wurden. rnNach der Etablierung des immunologischen Testsysteme war eine Hauptfragestellung dieser Arbeit, die selektierten nicht-synonyme Punktmutationen in einem in vivo Ansatz systematisch auf Antigenität zu testen. Für diese Studien wurden mutierte Sequenzen in einer Länge von 27 Aminosäuren genutzt, in denen die mutierte Aminosäure zentral positioniert war. Durch die Länge der Peptide können prinzipiell alle möglichen MHC Klasse-I und -II Epitope abgedeckt werden, welche die Mutation enthalten. Eine Grundidee des Projektes Ansatzes ist es, einen auf in vitro transkribierter RNA basierten oligotopen Impfstoff zu entwickeln. Daher wurden die Impfungen naiver Mäuse sowohl mit langen Peptiden, als auch in einem unabhängigen Ansatz mit peptidkodierender RNA durchgeführt. Die Immunphänotypisierung der Impfstoff induzierten T-Zellen zeigte, dass insgesamt 16 der 50 (32%) mutierten Sequenzen eine T-Zellreaktivität induzierten. rnDie Verwendung der vorhergesagten Epitope in therapeutischen Vakzinierungsstudien bestätigten die Hypothese das mutierte Neo-Epitope potente Zielstrukturen einer anti-tumoralen Impftherapie darstellen können. So wurde in therapeutischen Tumorstudien gezeigt, dass auf Basis von RNA 9 von 12 bestätigten Epitopen einen anti-tumoralen Effekt zeigte.rnÜberaschenderweise wurde bei einem MHC Klasse-II restringierten mutiertem Epitop (Mut-30) sowohl in einem subkutanen, als auch in einem unabhängigen therapeutischen Lungenmetastasen Modell ein starker anti-tumoraler Effekt auf B16-F10 beobachtet, der dieses Epitop als neues immundominantes Epitop für das B16-F10 Melanom etabliert. Um den immunologischen Mechanismus hinter diesem Effekt näher zu untersuchen wurde in verschieden Experimenten die Rolle von CD4+, CD8+ sowie NK-Zellen zu verschieden Zeitpunkten der Tumorentwicklung untersucht. Die Analyse des Tumorgewebes ergab, eine signifikante erhöhte Frequenz von NK-Zellen in den mit Mut-30 RNA vakzinierten Tieren. Das NK Zellen in der frühen Phase der Therapie eine entscheidende Rolle spielen wurde anhand von Depletionsstudien bestätigt. Daran anschließend wurde gezeigt, dass im fortgeschrittenen Tumorstadium die NK Zellen keinen weiteren relevanten Beitrag zum anti-tumoralen Effekt der RNA Vakzinierung leisten, sondern die Vakzine induzierte adaptive Immunantwort. Durch die Isolierung von Lymphozyten aus dem Tumorgewebe und deren Einsatz als Effektorzellen im IFN-γ ELISPOT wurde nachgewiesen, dass Mut-30 spezifische T-Zellen das Tumorgewebe infiltrieren und dort u.a. IFN-γ sekretieren. Dass diese spezifische IFN-γ Ausschüttung für den beobachteten antitumoralen Effekt eine zentrale Rolle einnimmt wurde unter der Verwendung von IFN-γ -/- K.O. Mäusen bestätigt.rnDas Konzept der individuellen RNA basierten mutationsspezifischen Vakzine sieht vor, nicht nur mit einem mutations-spezifischen Epitop, sondern mit mehreren RNA-kodierten Mutationen Patienten zu impfen um der Entstehung von „escape“-Mutanten entgegenzuwirken. Da es nur Erfahrung mit der Herstellung und Verabreichung von Monotop-RNA gab, also RNA die für ein Epitop kodiert, war eine wichtige Fragestellungen, inwieweit Oligotope, welche die mutierten Sequenzen sequentiell durch Linker verbunden als Fusionsprotein kodieren, Immunantworten induzieren können. Hierzu wurden Pentatope mit variierender Position des einzelnen Epitopes hinsichtlich ihrer in vivo induzierten T-Zellreaktivitäten charakterisiert. Die Experimente zeigten, dass es möglich ist, unabhängig von der Position im Pentatop eine Immunantwort gegen ein Epitop zu induzieren. Des weiteren wurde beobachtet, dass die induzierten T-Zellfrequenzen nach Pentatop Vakzinierung im Vergleich zur Nutzung von Monotopen signifikant gesteigert werden kann.rnZusammenfassend wurde im Rahmen der vorliegenden Arbeit präklinisch erstmalig nachgewiesen, dass nicht-synonyme Mutationen eine numerisch relevante Quelle von Zielstrukturen für die anti-tumorale Immuntherapie darstellen. Überraschenderweise zeigte sich eine dominante Induktion MHC-II restringierter Immunantworten, welche partiell in der Lage waren massive Tumorabstoßungsreaktionen zu induzieren. Im Sinne einer Translation der gewonnenen Erkenntnisse wurde ein RNA basiertes Oligotop-Format etabliert, welches Eingang in die klinische Testung des Konzeptes fand.rn
Resumo:
Antigenic changes present in nonantigenic tumor cells exposed to UV radiation (UV) in vitro were investigated by addressing the following questions: (1) Are antigenic variants (AV) produced that are rejected in normal but not immunosuppressed mice? (2) Does generation of AV depend upon intrinsic properties of the cells exposed or result from the action of UV? (3) Is antigenic modification induced by UV due to increased histocompatibility antigen expression? (4) Do AV crossreact immunologically with parental tumor or with other AV? and (5) Is the UV-associated common antigen expressed on UV-induced tumors present on UV-irradiated tumor cells? AV were generated at different frequencies following in vitro UV irradiation of a spontaneous murine fibrosarcoma (51% of cell lines tested), a murine melanoma (56%), and two melanoma clones (100% and 11%). This indicated that the percentage of AV produced is an intrinsic property of the cell line exposed. The increased antigenicity did not correlate with an increased expression of class I histocompatibility antigens. Immunological experiments demonstrated that the AV and parental cells shared a determinant that was susceptible to immune recognition, but incapable of inducing immunity. In contrast, the AV were noncrossreactive, suggesting that variant-specific antigens were also expressed. Finally, the AV were recognized by UV-induced suppressor cells, indicating that the UV-associated common antigen expressed by UV-induced tumors was also present. This investigation provides new information on the susceptibility of tumors to antigenic modification by UV and on the relationship between tumor antigens and neoplastic transformation. Furthermore, it suggests an immunological approach for cancer therapy. ^
Resumo:
Heparanase, an endo-$\beta$-D-glucuronidase, has been associated with melanoma metastasis. Polyclonal antibodies directed against the murine N-terminal heparanase peptide detected a M$\sb{\rm r}\sim 97,000$ protein upon SDS-polyacrylamide gel electrophoresis of mouse melanoma and human melanoma cell lysates. In an indirect immunocytochemical study, metastatic human A375-SM and mouse B16-BL6 melanoma cells were stained with the anti-heparanase antibodies. Heparanase antigen was localized in the cytoplasm of permeabilized melanoma cells as well as at the cell surface of unpermeabilized cells. Immunohistochemical staining of frozen sections from syngeneic mouse organs containing micrometastases of B16-BL6 melanoma demonstrated heparanase localized in metastatic melanoma cells, but not in adjacent normal tissues. Similar studies using frozen sections of malignant melanomas resected from patients indicated that heparanase is localized in invading melanoma cells, but not in adjacent connective tissues.^ Monoclonal antibodies directed against murine heparanase were developed and characterized. Monoclonal antibody 10E5, an IgM, precipitated and inhibitated the enzymatic activity of heparanase. A 2.6 kb cDNA was isolated from a human melanoma $\lambda$gt11 cDNA library using the monoclonal antibody 10E5. Heparan sulfate cleavage activity was detected in the lysogen lysates from E. Coli Y1089 infected with the $\lambda$gt11 cDNA and this activity was inhibited in the presence of 10-fold excess of heparin, a potent inhibitor of heparanase. The nucleotide sequence of the cDNA was determined and insignificant homology was found with the gene sequences currently known. The cDNA hybridized to a 3.2-3.4 kb mRNA in human A375 melanoma, WI-38 fibroblast, and THP-1 leukemia cells using Northern blots.^ Heparanase expression was examined using Western and Northern blots. In comparison to human A375-P melanoma cells, the quantity of 97,000 protein recognized by the polyclonal anti-heparanase antibodies doubled in the metastatic variant A375-SM cells and the quantity of 3.2-3.4 kb mRNA doubled in A375MetMix, a metastatic variant similar to A375-SM cells. In B16 murine melanoma cell, the intensity of the 97,000 protein increased more than 2 times comparing with B16-F1 cells. The extent in the increase of the protein and the mRNA levels is comparable to the change of heparanase activity observed in those cells.^ In summary, the studies suggest that (a) the N-terminus of the heparanase molecule in mouse and human is antigenically related; (b) heparanase antigens are localized at the cell surface and in the cytoplasm of metastatic human and mouse melanoma cells; (c) heparanase antigens are localized in invasive and metastatic murine and human melanomas in vivo, but not in adjacent normal tissues; (d) heparanase molecule appeared to be differentially expressed at the transcriptional as well as at the translational level; and (e) the size of human heparanase mRNA is 3.2-3.4 kilobase. ^
Resumo:
Metastasis is the complex process of tumor cell spread which is responsible for the majority of cancer-related deaths. Metastasis necessitates complex phenotypic changes, many of which are mediated by changes in the activities of cell surface molecules. One of these is cell surface $\beta$1,4-galactosyltransferase (GalTase), which is elevated on more highly metastatic cells. In this study, both molecular and biochemical methods were used to perturb and manipulate cell surface GalTase levels on K1735 murine melanoma cell lines in order to examine its function in metastasis.^ As expected, highly metastatic K1735 variants have higher cell surface GalTase than poorly metastatic variants. Stably transfected K1735 cell lines that overexpress surface GalTase were created. These cell lines were assayed for metastatic ability using an invasion chamber with Matrigel-coated filter inserts. Cells with increased surface GalTase were uniformly more invasive than neo transfected controls. With multiple cell lines, there was a direct correlation (r = 0.918) between surface GalTase activity and invasiveness. Homologous recombination was used to create K1735 cells with decreased levels of surface GalTase. These cells were uniformly less invasive than controls. Cell surface GalTase was inhibited using two different biochemical strategies. In both cases, inhibition of surface GalTase led to a decrease in in vivo metastatic ability of K1735 cells. This is the first direct experimental evidence addressing GalTase function in metastasis. These data provide several lines of independent evidence which show that cell surface GalTase facilitates invasion and metastasis. ^
Resumo:
To investigate the hypothesis that increased malignant potential correlates with increased levels of genetic instability, the following parameters of instability were measured: (1) spontaneous mutation rates for ouabain resistance in murine cell lines of different malignant potentials, (2) the background prevalence of 6-thioguanine (6-TG) resistance in clone 4 (highly metastatic) and clone 19 (poorly metastatic) of the K1735 murine melanoma, (3) the prevalence of ouabain resistant variants in three murine cell lines and their variants after exposure to the mutagen MNNG, (4) the rate of generation of major karyotypic abnormalities in B16 F1 (poorly metastatic) and B16 F10 (highly metastatic) murine melanoma, and (5) analysis of the G-banded karyotypes of cloned B16 F1 and B16 F10 melanoma.^ No correlation of increased spontaneous mutation rates with increased malignant potential was found in repeated experiments with three murine cell lines and their variants of different malignant potential. The background prevalence of g-TG resistance was not significantly different for the poorly and highly metastatic clones of K1735 melanoma. The studies with MNNG-induced mutation showed no increased sensitivity of the highly metastatic variants of the three murine cell lines to mutagenesis. Neither did the rate of generation of major karyotypic abnormalities correlate with malignant potential. However, certain karyotypic differences were demonstrated after G-banding of the B16 F1 and F10 melanomas.^ One hypothesis which is consistent with these results is that the rate of generation of genetic abnormalities need not be strongly related to the degree of malignant potential. An increased prevalence of genetic changes may merely reflect the accumulation of abnormalities while their rate of production remains constant. The presence of specific nonrandom changes likely is the main determinant of malignant potential rather than the rate of production of random changes. ^
Resumo:
Recent data suggest that the generation of new lymphatic vessels (i.e. lymphangiogenesis) may be a rate-limiting step in the dissemination of tumor cells to regional lymph nodes. However, efforts to study the cellular and molecular interactions that take place between tumor cells and lymphatic endothelial cells have been limited due to a lack of lymphatic endothelial cell lines available for study. ^ I have used a microsurgical approach to establish conditionally immortalized lymphatic endothelial cell lines from the afferent mesenteric lymphatic vessels of mice. Characterization of lymphatic endothelial cells, and tumor-associated lymphatic vessels revealed high expression levels of VCAM-1, which is known to facilitate adhesion of some tumor cells to vascular endothelial cells. Further investigation revealed that murine melanoma cells selected for high expression of α4, a counter-receptor for VCAM-1, demonstrated enhanced adhesion to lymphatic endothelial cells in vitro, and increased tumorigenicity and lymphatic metastasis in vivo, despite similar lymphatic vessel numbers. ^ Next, I examined the effects of growth factors that regulate lymphangiogenesis, and report that several growth factors are capable of activating survival and proliferation pathways of lymphatic endothelial cells. The dual protein tyrosine kinase inhibitor AEE788 (EGFR and VEGFR-2) inhibited the activation of Akt and MAPK in lymphatic endothelial cells responding to multiple growth factors. Moreover, oral treatment of mice with AEE788 decreased lymphatic vessel density and production of lymphatic metastasis by human colon cancer cells growing in the cecum of nude mice. ^ In the last set of experiments, I investigated the surgical management of lymphatic metastasis using a novel model of sentinel lymphadenectomy in live mice bearing subcutaneous B16-BL6 melanoma. The data demonstrate that this procedure when combined with wide excision of the primary melanoma, significantly enhanced survival of syngeneic C57BL/6 mice. ^ Collectively, these results indicate that the production of lymphatic metastasis depends on lymphangiogenesis, tumor cell adhesion to lymphatic endothelial cells, and proliferation of tumor cells in lymph nodes. Thus, lymphatic metastasis is a multi-step, complex, and active process that depends upon multiple interactions between tumor cells and tumor associated lymphatic endothelial cells. ^
Resumo:
The adenovirus type 5 E1A gene products have numerous functions in cells, which serve as useful tools in studying the mechanisms of either oncogenesis or tumor suppression. To understand the mechanisms of E1A-mediated tumor suppression, we introduced an Ad5 E1A gene into murine melanoma cells, and characterized E1A-mediated biological functions both in vitro and in vivo. The results of the study indicated that: (i) Ad5 E1A mediated tumor suppression in rodent tumor cells; (ii) E1A-mediated tumor suppression is associated with E1A-mediated apoptosis in vivo.^ To determine which functional region(s) of E1A is(are) required for E1A-mediated apoptosis and whether E1A-mediated apoptosis is required for E1A-mediated tumor suppression, we established stable transfectants of E1A mutants, which have deletion mutation at either the N-terminal (p300-binding) or the CR2 (pRb-binding) domain or both, and then characterized biological functions both in vitro and in vivo. The results of the study indicate that the CR2 domain of E1A is required for E1A-mediated apoptosis, while the N-terminal domain of E1A is dispensable. Interestingly, either of the two domains is able to mediate tumor suppression, since mutant E1A with a single deletion at either domain still suppressed tumor growth. Importantly, deletion mutations at both the N-terminal and the CR2 domains of E1A abrogated E1A-mediated tumor suppression, suggesting both regions are required for E1A-mediated tumor suppression. The results demonstrate that E1A-mediated apoptosis is not the only mechanism for E1A-mediated tumor suppression. Thus, the N-terminal and CR2 domains of E1A mediated two independent mechanisms of tumor suppression.^ To understand the mechanism of E1A-mediated apoptosis, we examined the temporal relationship of molecular events during the apoptotic cascades after UV radiation and serum depletion in both the E1A-expressing cells and parental cells. Kinetic analysis of JNK activity indicates that the JNK pathway is greatly increased in response to UV light in E1A transfectants, suggesting that extracellular stress stimuli have been converted into intracellular stress signals with greater magnitude in E1A transfectants than those in parental cells. Thus, E1A-mediated sensitization precedes these events. As ceramide has been proposed as second messenger and upstream activator of JNK pathway for stress-induced apoptosis, we also examined the roles of ceramide in apoptosis and the relationship with JNK pathway. The results indicate that E1A transfectants do not have increased sensitivity to ceramide. Therefore, E1A-mediated sensitization to UV radiation cannot be attributed to an increased sensitivity to ceramide. Furthermore, UV-induced JNK activation correlates with UV-induced apoptosis, while lethal dose of ceramide does not activate JNK. Thus, activation of JNK pathway is independent of the ceramide pathway. In addition, E1A transfectants also have increased activation of NF-kB in response to UV. These results suggest that E1A-mediated sensitization is an early event which associates with conversion of extracellular stress stimuli into amplified intracellular signals. The mechanism of E1A-mediated sensitization and its relationship with other pathways are discussed. ^
Resumo:
α-Melanocyte stimulating hormone (α-MSH) analogs, cyclized through site-specific rhenium (Re) and technetium (Tc) metal coordination, were structurally characterized and analyzed for their abilities to bind α-MSH receptors present on melanoma cells and in tumor-bearing mice. Results from receptor-binding assays conducted with B16 F1 murine melanoma cells indicated that receptor-binding affinity was reduced to approximately 1% of its original levels after Re incorporation into the cyclic Cys4,10, d-Phe7–α-MSH4-13 analog. Structural analysis of the Re–peptide complex showed that the disulfide bond of the original peptide was replaced by thiolate–metal–thiolate cyclization. A comparison of the metal-bound and metal-free structures indicated that metal complexation dramatically altered the structure of the receptor-binding core sequence. Redesign of the metal binding site resulted in a second-generation Re–peptide complex (ReCCMSH) that displayed a receptor-binding affinity of 2.9 nM, 25-fold higher than the initial Re–α-MSH analog. Characterization of the second-generation Re–peptide complex indicated that the peptide was still cyclized through Re coordination, but the structure of the receptor-binding sequence was no longer constrained. The corresponding 99mTc- and 188ReCCMSH complexes were synthesized and shown to be stable in phosphate-buffered saline and to challenges from diethylenetriaminepentaacetic acid (DTPA) and free cysteine. In vivo, the 99mTcCCMSH complex exhibited significant tumor uptake and retention and was effective in imaging melanoma in a murine-tumor model system. Cyclization of α-MSH analogs via 99mTc and 188Re yields chemically stable and biologically active molecules with potential melanoma-imaging and therapeutic properties.
Resumo:
Messenger RNA transcripts of the highly pigmented murine melanoma B16-F1 cells were compared with those from their weakly pigmented derivative B16-F10 cells by differential display. A novel gene called msg1 (melanocyte-specific gene) was found to be expressed at high levels in B16-F1 cells but at low levels in B16-F10 cells. Expression of msg1 was undetectable in the amelanotic K1735 murine melanoma cells. The pigmented murine melanocyte cell line melan-a expressed msg1, as did pigmented primary cultures of murine and human melanocytes; however, seven amelanotic or very weakly pigmented human melanoma cell lines were negative. Transformation of murine melanocytes by transfection with v-Ha-ras or Ela was accompanied by depigmentation and led to complete loss of msg1 expression. The normal tissue distribution of msg1 mRNA transcripts in adult mice was confined to melanocytes and testis. Murine msg1 and human MSG1 genes encode a predicted protein of 27 kDa with 75% overall amino acid identity and 96% identity within the C-terminal acidic domain of 54 amino acids. This C-terminal domain was conserved with 76% amino acid identity in another protein product of a novel human gene, MRG1 (msg1-related gene), isolated from normal human melanocyte cDNA by 5'-rapid amplification of cDNA ends based on the homology to msg1. The msg1 protein was localized to the melanocyte nucleus by immunofluorescence cytochemistry. We conclude that msg1 encodes a nuclear protein, is melanocyte-specific, and appears to be lost in depigmented melanoma cells.
Resumo:
Vaccination with cytokine-producing tumor cells generates potent immune responses against tumors outside the central nervous system (CNS). The CNS, however, is a barrier to allograft and xenograft rejection, and established tumors within the CNS have failed to respond to other forms of systemic immunotherapy. To determine what barriers the "immunologically privileged" CNS would pose to cytokine-assisted tumor vaccines and what cytokines would be most efficacious against tumors within the CNS, we irradiated B16 murine melanoma cells producing murine interleukin 2 (IL-2), IL-3, IL-4, IL-6, gamma-interferon, or granulocyte-macrophage colony stimulating factor (GM-CSF) and used these cells as subcutaneous vaccines against tumors within the brain. Under conditions where untransfected B16 cells had no effect, cells producing IL-3, IL-6, or GM-CSF increased the survival of mice challenged with viable B16 cells in the brain. Vaccination with B16 cells producing IL-4 or gamma-interferon had no effect, and vaccination with B16 cells producing IL-2 decreased survival time. GM-CSF-producing vaccines were also able to increase survival in mice with pre-established tumors. The response elicited by GM-CSF-producing vaccines was found to be specific to tumor type and to be abrogated by depletion of CD8+ cells. Unlike the immunity generated against subcutaneous tumors by GM-CSF, however, the effector responses generated against tumors in the CNS were not dependent on CD4+ cells. These data suggest that cytokine-producing tumor cells are very potent stimulators of immunity against tumors within the CNS, but effector responses in the CNS may be different from those obtained against subcutaneous tumors.
Resumo:
Although the ability of UV irradiation to induce pigmentation in vivo and in vitro is well documented, the intracellular signals that trigger this response are poorly understood. We have recently shown that increasing DNA repair after irradiation enhances UV-induced melanization. Moreover, addition of small DNA fragments, particularly thymine dinucleotides (pTpT), selected to mimic sequences excised during the repair of UV-induced DNA photoproducts, to unirradiated pigment cells in vitro or to guinea pig skin in vivo induces a pigment response indistinguishable from UV-induced tanning. Here we present further evidence that DNA damage and/or the repair of this damage increases melanization. (i) Treatment with the restriction enzyme Pvu II or the DNA-damaging chemical agents methyl methanesulfonate (MMS) or 4-nitroquinoline 1-oxide (4-NQO) produces a 4- to 10-fold increase in melanin content in Cloudman S91 murine melanoma cells and an up to 70% increase in normal human melanocytes, (ii) UV irradiation, MMS, and pTpT all upregulate the mRNA level for tyrosinase, the rate-limiting enzyme in melanin biosynthesis. (iii) Treatment with pTpT or MMS increases the response of S91 cells to melanocyte-stimulating hormone (MSH) and increases the binding of MSH to its cell surface receptor, as has been reported for UV irradiation. Together, these data suggest that UV-induced DNA damage and/or the repair of this damage is an important signal in the pigmentation response to UV irradiation. Because Pvu II acts exclusively on DNA and because MMS and 4-NQO, at the concentrations used, primarily interact with DNA, such a stimulus alone appears sufficient to induce melanogenesis. Of possible practical importance, the dinucleotide pTpT mimics most, if not all, of the effects of UV irradiation on pigmentation, tyrosinase mRNA regulation, and response to MSH without the requirement for antecedent DNA damage.
Resumo:
Although both CD4+ and CD8+ T cells are clearly required to generate long-lasting anti-tumor immunity induced by s.c. vaccination with interleukin 2 (IL-2)-transfected, irradiated M-3 clone murine melanoma cells, some controversy continues about the site and mode of T-cell activation in this system. Macrophages, granulocytes, and natural killer cells infiltrate the vaccination site early after injection into either syngeneic euthymic DBA/2 mice or athymic nude mice and eliminate the inoculum within 48 hr. We could not find T cells at the vaccination site, which argues against the concept that T-cell priming by the IL-2-secreting cancer cells occurs directly at that location. However, reverse transcription-PCR revealed transcripts indicative of T-cell activation and expansion in the draining lymph nodes of mice immunized with the IL-2-secreting vaccine but not in mice vaccinated with untransfected, irradiated M-3 cells. We therefore propose that the antigen-presenting cells, which invade the vaccination site, process tumor-derived antigens and, subsequently, initiate priming of tumor-specific T lymphocytes in lymphoid organs. These findings suggest a three-stage process for the generation of effector T cells after vaccination with IL-2-secreting tumor cells: (i) tumor-antigen uptake and processing at the site of injection by antigen-presenting cells, (ii) migration of antigen-presenting cells into the regional draining lymph nodes, where T-cell priming occurs, and (iii) circulation of activated T cells that either perform or initiate effector mechanisms leading to tumor cell destruction.
Resumo:
Nas últimas décadas, muito interesse tem sido focado no potencial biotecnológico das microalgas, principalmente devido à identificação de diversas substâncias bioativas sintetizadas por estas, especialmente ácidos graxos poliinsaturados (PUFAs). PUFAs ômega-3 (PUFAs ω-3) têm sido estudados pela promoção de efeitos benéficos em muitas doenças crônicas, incluindo o câncer, ao contrário de PUFAs ômega-6 (PUFAs ω-6) que, de forma geral, são conhecidos por agravar o desenvolvimento destes quadros. Este estudo objetivou testar a atividade antiproliferativa de extratos lipídicos de duas microalgas marinhas, Isochrysis galbana e Thalassiosira pseudonana, ricas principalmente em PUFAs ω-3, em uma linhagem celular de melanoma, B16F10. Adicionalmente, o efeito dos precursores dos PUFAs ω-3, α-linolênico (ALA), e dos PUFAs ω-6, linoleico (LA) na proliferação celular, foi avaliado nesta linhagem e em uma linhagem melanocítica normal, Melan-A, pelo método MTT. Foi demonstrado que os extratos lipídicos estudados são capazes de induzir inibição de proliferação tempo e concentração dependente na linhagem B16F10 e esta atividade foi também exercida pelo ALA de forma independente das concentrações testadas. Além disso, o extrato lipídico da microalga Thalassiosira pseudonana também induziu citotoxicidade na linhagem B16F10. Desse modo, é possível sugerir que o efeito antiproliferativo dos extratos possa estar relacionado com a alta concentração de ω-3 PUFAs em relação a ω- 6 PUFAs, especialmente para a Thalassiosira pseudonana Adicionalmente, a linhagem Melan-A não foi sensível ao tratamento com o ALA, sugerindo, dessa forma, um efeito antitumoral para este composto. Este estudo ainda indica a possibilidade do uso de microalgas como fontes promissoras de substâncias antitumorais.
Resumo:
Tese de Doutoramento em Ciências Veterinárias na Especialidade de Ciências Biológicas e Biomédicas