Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arthritis Rheumatol ; 73(6): 1021-1032, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33314777

RESUMO

OBJECTIVE: To identify novel heterozygous LPIN2 mutations in a patient with Majeed syndrome and characterize the pathomechanisms that lead to the development of sterile osteomyelitis. METHODS: Targeted genetic analysis and functional studies assessing monocyte responses, macrophage differentiation, and osteoclastogenesis were conducted to compare the pathogenesis of Majeed syndrome to interleukin-1 (IL-1)-mediated diseases including neonatal-onset multisystem inflammatory disease (NOMID) and deficiency of the IL-1 receptor antagonist (DIRA). RESULTS: A 4-year-old girl of mixed ethnic background presented with sterile osteomyelitis and elevated acute-phase reactants. She had a 17.8-kb deletion on the maternal LPIN2 allele and a splice site mutation, p.R517H, that variably spliced out exons 10 and 11 on the paternal LPIN2 allele. The patient achieved long-lasting remission receiving IL-1 blockade with canakinumab. Compared to controls, monocytes and monocyte-derived M1-like macrophages from the patient with Majeed syndrome and those with NOMID or DIRA had elevated caspase 1 activity and IL-1ß secretion. In contrast, lipopolysaccharide-stimulated, monocyte-derived, M2-like macrophages from the patient with Majeed syndrome released higher levels of osteoclastogenic mediators (IL-8, IL-6, tumor necrosis factor, CCL2, macrophage inflammatory protein 1α/ß, CXCL8, and CXCL1) compared to NOMID patients and healthy controls. Accelerated osteoclastogenesis in the patient with Majeed syndrome was associated with higher NFATc1 levels, enhanced JNK/MAPK, and reduced Src kinase activation, and partially responded to JNK inhibition and IL-1 (but not IL-6) blockade. CONCLUSION: We report 2 novel compound heterozygous disease-causing mutations in LPIN2 in an American patient with Majeed syndrome. LPIN2 deficiency drives differentiation of proinflammatory M2-like macrophages and enhances intrinsic osteoclastogenesis. This provides a model for the pathogenesis of sterile osteomyelitis which differentiates Majeed syndrome from other IL-1-mediated autoinflammatory diseases.


Assuntos
Anemia Diseritropoética Congênita/genética , Síndromes de Imunodeficiência/genética , Inflamação/genética , Macrófagos/imunologia , Proteínas Nucleares/genética , Osteogênese/genética , Osteomielite/genética , Anemia Diseritropoética Congênita/tratamento farmacológico , Anemia Diseritropoética Congênita/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Estudos de Casos e Controles , Pré-Escolar , Síndromes Periódicas Associadas à Criopirina/genética , Síndromes Periódicas Associadas à Criopirina/imunologia , Feminino , Doenças Hereditárias Autoinflamatórias/genética , Doenças Hereditárias Autoinflamatórias/imunologia , Heterozigoto , Humanos , Síndromes de Imunodeficiência/tratamento farmacológico , Síndromes de Imunodeficiência/imunologia , Inflamação/imunologia , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/imunologia , MAP Quinase Quinase 4/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Transcrição NFATC/metabolismo , Proteínas Nucleares/imunologia , Osteomielite/tratamento farmacológico , Osteomielite/imunologia , Quinases da Família src/metabolismo
2.
Adv Exp Med Biol ; 1185: 353-358, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884637

RESUMO

Neurotrophic factors can promote the survival of degenerating retinal cells through the activation of STAT3 pathway. Thus, augmenting STAT3 activation in the retina has been proposed as potential therapy for retinal dystrophies. On the other hand, aberrant activation of STAT3 pathway is oncogenic and implicated in diverse human diseases. Furthermore, the STAT3/SOCS3 axis has been shown to induce the degradation of rhodopsin during retinal inflammation. In this study, we generated and used mice with constitutive activation of STAT3 pathway in the retina to evaluate the safety and consequences of enhancing STAT3 activities in the retina as a potential treatment for retinal degenerative diseases. We show that long-term activation of the STAT3 pathway can induce retinal degenerative changes and also exacerbate uveitis and other intraocular inflammatory diseases. Mechanisms underlying the development of vision impairment in the STAT3c-Tg mice derived in part from STAT3-mediated inhibition of rhodopsin and overexpression of SOCS3 in the retina. These results suggest that much caution should be exercised in the use of STAT3 augmentation therapy for retinal dystrophies.


Assuntos
Envelhecimento , Retina/patologia , Degeneração Retiniana/patologia , Fator de Transcrição STAT3/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteínas Supressoras da Sinalização de Citocina , Uveíte/patologia
3.
Nat Commun ; 9(1): 3506, 2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30158636

RESUMO

The transcription factor Nrf2 is a critical regulator of inflammatory responses. If and how Nrf2 also affects cytosolic nucleic acid sensing is currently unknown. Here we identify Nrf2 as an important negative regulator of STING and suggest a link between metabolic reprogramming and antiviral cytosolic DNA sensing in human cells. Here, Nrf2 activation decreases STING expression and responsiveness to STING agonists while increasing susceptibility to infection with DNA viruses. Mechanistically, Nrf2 regulates STING expression by decreasing STING mRNA stability. Repression of STING by Nrf2 occurs in metabolically reprogrammed cells following TLR4/7 engagement, and is inducible by a cell-permeable derivative of the TCA-cycle-derived metabolite itaconate (4-octyl-itaconate, 4-OI). Additionally, engagement of this pathway by 4-OI or the Nrf2 inducer sulforaphane is sufficient to repress STING expression and type I IFN production in cells from patients with STING-dependent interferonopathies. We propose Nrf2 inducers as a future treatment option in STING-dependent inflammatory diseases.


Assuntos
Proteínas de Membrana/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Vírus de DNA/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Proteínas de Membrana/genética , Camundongos , Fator 2 Relacionado a NF-E2/genética , Células RAW 264.7 , RNA Mensageiro/metabolismo , Succinatos/farmacologia
4.
J Clin Invest ; 125(11): 4196-211, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26524591

RESUMO

Autosomal recessive mutations in proteasome subunit ß 8 (PSMB8), which encodes the inducible proteasome subunit ß5i, cause the immune-dysregulatory disease chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE), which is classified as a proteasome-associated autoinflammatory syndrome (PRAAS). Here, we identified 8 mutations in 4 proteasome genes, PSMA3 (encodes α7), PSMB4 (encodes ß7), PSMB9 (encodes ß1i), and proteasome maturation protein (POMP), that have not been previously associated with disease and 1 mutation in PSMB8 that has not been previously reported. One patient was compound heterozygous for PSMB4 mutations, 6 patients from 4 families were heterozygous for a missense mutation in 1 inducible proteasome subunit and a mutation in a constitutive proteasome subunit, and 1 patient was heterozygous for a POMP mutation, thus establishing a digenic and autosomal dominant inheritance pattern of PRAAS. Function evaluation revealed that these mutations variably affect transcription, protein expression, protein folding, proteasome assembly, and, ultimately, proteasome activity. Moreover, defects in proteasome formation and function were recapitulated by siRNA-mediated knockdown of the respective subunits in primary fibroblasts from healthy individuals. Patient-isolated hematopoietic and nonhematopoietic cells exhibited a strong IFN gene-expression signature, irrespective of genotype. Additionally, chemical proteasome inhibition or progressive depletion of proteasome subunit gene transcription with siRNA induced transcription of type I IFN genes in healthy control cells. Our results provide further insight into CANDLE genetics and link global proteasome dysfunction to increased type I IFN production.


Assuntos
Doenças Hereditárias Autoinflamatórias/genética , Interferon Tipo I/biossíntese , Lipodistrofia/genética , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Sequência de Aminoácidos , Células Cultivadas , Fibroblastos , Regulação da Expressão Gênica , Genótipo , Doenças Hereditárias Autoinflamatórias/imunologia , Doenças Hereditárias Autoinflamatórias/metabolismo , Humanos , Interferon Tipo I/genética , Lipodistrofia/imunologia , Lipodistrofia/metabolismo , Modelos Moleculares , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Conformação Proteica , Subunidades Proteicas , Interferência de RNA , RNA Interferente Pequeno/genética , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Síndrome , Transcrição Gênica
5.
Nat Genet ; 46(10): 1140-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25217959

RESUMO

Inflammasomes are innate immune sensors that respond to pathogen- and damage-associated signals with caspase-1 activation, interleukin (IL)-1ß and IL-18 secretion, and macrophage pyroptosis. The discovery that dominant gain-of-function mutations in NLRP3 cause the cryopyrin-associated periodic syndromes (CAPS) and trigger spontaneous inflammasome activation and IL-1ß oversecretion led to successful treatment with IL-1-blocking agents. Herein we report a de novo missense mutation (c.1009A > T, encoding p.Thr337Ser) affecting the nucleotide-binding domain of the inflammasome component NLRC4 that causes early-onset recurrent fever flares and macrophage activation syndrome (MAS). Functional analyses demonstrated spontaneous inflammasome formation and production of the inflammasome-dependent cytokines IL-1ß and IL-18, with the latter exceeding the levels seen in CAPS. The NLRC4 mutation caused constitutive caspase-1 cleavage in cells transduced with mutant NLRC4 and increased production of IL-18 in both patient-derived and mutant NLRC4-transduced macrophages. Thus, we describe a new monoallelic inflammasome defect that expands the monogenic autoinflammatory disease spectrum to include MAS and suggests new targets for therapy.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ligação ao Cálcio/genética , Inflamassomos/genética , Inflamação/genética , Síndrome de Ativação Macrofágica/genética , Mutação de Sentido Incorreto , Sequência de Aminoácidos , Criança , Exoma/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação/sangue , Interleucina-18/sangue , Interleucina-18/metabolismo , Síndrome de Ativação Macrofágica/sangue , Macrófagos/metabolismo , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
6.
Cell Immunol ; 289(1-2): 140-4, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24791699

RESUMO

Clinical trials suggest that BAFF inhibitors such as atacicept (TACI-IgG) and belimumab (anti-BAFF antibody) could not reduce memory B-cell numbers, although they reduced the numbers of CD20(+) naïve B cells and activated B cells. In the present study, we explored the way to reduce memory B-cell numbers. First, we used TACI-IgG to treat murine lupus. We found that TACI-IgG was effective in reducing mature B cell numbers. Accordingly it controlled the level of the anti-dsDNA antibody in lupus-like mice. In addition, TACI-IgG up-regulated memory B cells in murine lupus. Furthermore, we found that TACI-IgG up-regulated IL-15 expression in lupus-like mice. Thus, the combination of TACI-IgG and anti-IL-15 antibodies was explored to understand their effects on the treatment of murine lupus. Compared to treatments with TACI-IgG or anti-IL-15 alone, the combination of TACI-IgG and anti-IL-15 antibodies efficiently ameliorated murine lupus phenotypes. The study provides hints for the clinical application of BAFF- and IL-15-specific therapeutic agents.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Linfócitos B/imunologia , Interleucina-15/imunologia , Lúpus Eritematoso Sistêmico/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos Antinucleares/imunologia , Fator Ativador de Células B/imunologia , DNA/imunologia , Quimioterapia Combinada , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/imunologia , Imunossupressores/uso terapêutico , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Transdução de Sinais/imunologia
7.
J Immunol ; 192(9): 4192-201, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24670802

RESUMO

Clinical trials have shown that BAFF inhibitors do not reduce memory B cell levels but can reduce the number of mature B cells. It remains uncertain whether BAFF affects memory-maintaining cytokines such as IL-15. We found that BAFF suppressed IL-15 expression in B cells from lupus-like or experimental allergic encephalomyelitis mice. When BAFF was blocked with atacicept-IgG, IL-15 expression was upregulated in lupus-like or experimental allergic encephalomyelitis mice. Finally, we showed that BAFF suppressed IL-15 expression in transitional 2 B cells by reducing Foxo1 expression and inducing Foxo1 phosphorylation. This study suggests that BAFF suppresses IL-15 expression in autoimmune diseases, and this opens up the possible opportunity for the clinical application of BAFF- and IL-15-specific therapeutic agents.


Assuntos
Doenças Autoimunes/imunologia , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Interleucina-15/biossíntese , Animais , Doenças Autoimunes/metabolismo , Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Western Blotting , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Interleucina-15/imunologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Technol Cancer Res Treat ; 13(6): 551-60, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24000979

RESUMO

Electroporation (EP) is a method used to physically deliver therapeutic molecules such as plasmid DNA directly to tissues. It has been used safely and successfully in clinical studies and preclinical cancer models to deliver genes to a variety of tissues. In cancer research cytokine therapy is emerging as a promising tool that can be used to boost the host response to tumor antigens. The delivery of cytokines as recombinant proteins can result in toxicity and other adverse effects; however the delivery of cytokine genes using EP has been shown to be safe and effective. Interleukin 15 (IL-15) is a cytokine that promotes the innate as well as the adaptive immune response to cancer cells and bacterial pathogens. In this study we used EP to deliver a human IL-15 plasmid (phIL-15) directly to tumors to examine its anti-cancer effects. B16.F10 melanoma tumors were induced in C57BL/6J mice and phIL-15 was delivered three times over the course of a week. Expression of the transgene, tumor volume, long-term survival and resistance to challenge were monitored in these animals. Delivery of IL-15 plasmid by EP resulted in increased IL-15 expression within the tumor compared to the injection only control. This expression peaked at 12 to 18 hours after the first delivery and was sustained at lower levels after the second and third deliveries. The delivery of the phIL-15 resulted in tumor regression, long-term survival and greater protection against tumor recurrence when cancer cells were reintroduced compared to control plasmid. From these results we can conclude that the delivery of IL-15 plasmid to tumors using EP is a promising avenue to investigate for its anti-tumor effects, however more work needs to be done to increase the stability of the gene once it is delivered and to elucidate the anti-tumor mechanism.


Assuntos
Eletroporação , Técnicas de Transferência de Genes , Interleucina-15/genética , Melanoma Experimental/genética , Melanoma Experimental/patologia , Animais , Linhagem Celular Tumoral , Eletroporação/métodos , Feminino , Expressão Gênica , Humanos , Melanoma Experimental/imunologia , Melanoma Experimental/mortalidade , Melanoma Experimental/terapia , Camundongos , Plasmídeos/genética , Carga Tumoral
9.
Mol Immunol ; 57(2): 129-37, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24084099

RESUMO

Dysregulation of T-cell survival and apoptosis is the common cause of autoimmune diseases such as multiple sclerosis (MS). However, the factors inducing imbalance of T-cell survival and apoptosis in MS remains unclear. Here, we show that the resistance to apoptosis was associated with high levels of B-cell activating factor (BAFF). Blockade of BAFF with TACI (transmembrane activator and calcium modulator and cyclophilin ligand interactor)-IgG significantly reduced T-cell survival in myelin oligodendroglia glycoprotein (MOG)-induced chronic experimental allergic encephalitis (EAE). Furthermore, BAFF induced anti-apoptotic molecule Bcl2 expression in T cells by up-regulating osteopontin (OPN) secretion from B cells. BAFF mainly induced OPN expression in splenic CD21(-)CD23(+) B cells via a NF-kB dependent signaling pathway. In addition, we found that BAFF and OPN levels were increased in MS patients similar to the results obtained from our mice research. The study suggests that BAFF regulates T-cell survival by inducing OPN secretion in B cells in autoimmune diseases.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Osteopontina/metabolismo , Animais , Apoptose , Fator Ativador de Células B/biossíntese , Linfócitos B/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Humanos , Imunoglobulina G , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , NF-kappa B/metabolismo , Osteopontina/biossíntese , Fragmentos de Peptídeos/imunologia , Receptores de Complemento 3d/biossíntese , Receptores de IgE/biossíntese , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo
10.
J Biol Chem ; 287(36): 30436-43, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22761423

RESUMO

An important feature of the adaptive immune response is its remarkable capacity to regulate the duration of inflammatory responses, and effector T cells have been shown to limit excessive immune responses by producing anti-inflammatory cytokines such as IL-10 and IL-27. However, how anti-inflammatory cytokines mediate their suppressive activities is not well understood. In this study, we show that STAT3 contributes to mechanisms that control the duration of T cell proliferation by regulating the subcellular location of FoxO1 and FoxO3a, two Class O Forkhead transcription factors that mediate lymphocyte quiescence and inhibit T cell activation. We show that active FoxO1 and FoxO3a reside exclusively in the nucleus of naïve T cells whereas inactive pFoxO1 and pFoxO3a were most abundant in activated T cells and sequestered in their cytoplasm in association with unphosphorylated STAT3 (U-STAT3) and 14-3-3. We further show that FoxO1/FoxO3a rapidly relocalized into the nucleus in response to pSTAT3 activation by IL-6 or IL-10, and the accumulation of FoxO1/FoxO3a in their nuclei coincided with increased expression of p27(Kip1) and p21(WAF1). STAT3 inhibitors completely abrogated cytokine-induced translocation of FoxO1/FoxO3a into the nucleus. In naïve or resting STAT3-deficient T cells, expression of pFoxO1/pFoxO3a was predominantly in the cytoplasm and correlated with defects in p27(Kip1) and p21(WAF1) expression, suggesting requirement of STAT3 for importation or retention of FoxO in the nucleus and attenuation of lymphocyte proliferation. Taken together, these results suggest that U-STAT3 collaborates with 14-3-3 to sequester pFoxO1/pFoxO3a in cytoplasm and thus prolong T cell activation, whereas pSTAT3 activation by anti-inflammatory cytokines would curtail the duration of TCR activation and re-establish lymphocyte quiescence by inducing nuclear localization of FoxO1/FoxO3a and FoxO-mediated expression of growth-inhibitory proteins.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/imunologia , Proteínas 14-3-3/metabolismo , Transporte Ativo do Núcleo Celular/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Núcleo Celular/genética , Núcleo Celular/imunologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/imunologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/imunologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Citoplasma/genética , Citoplasma/imunologia , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Células Jurkat , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia
11.
Biomaterials ; 33(10): 3036-46, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22244695

RESUMO

A large-scale in vitro 3D tumor model was generated to evaluate gene delivery procedures in vivo. This 3D tumor model consists of a "tissue-like" spheroid that provides a micro-environment supportive of melanoma proliferation, allowing cells to behave similarly to cells in vivo. This functional spheroid measures approximately 1 cm in diameter and can be used to effectively evaluate plasmid transfection when testing various electroporation (EP) electrode applicators. In this study, we identified EP conditions that efficiently transfect green fluorescent protein (GFP) and interleukin 15 (IL-15) plasmids into tumor cells residing in the 3D construct. We found that plasmids delivered using a 6-plate electrode applying 6 pulses with nominal electric field strength of 500 V/cm and pulse-length of 20 ms produced significant increase of GFP (7.3-fold) and IL-15 (3.0-fold) expression compared to controls. This in vitro 3D model demonstrates the predictability of cellular response toward delivery techniques, limits the numbers of animals employed for transfection studies, and may facilitate future developments of clinical trials for cancer therapies in vivo.


Assuntos
Eletroporação/métodos , Melanoma/metabolismo , Melanoma/patologia , Plasmídeos/metabolismo , Transfecção/métodos , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Eletrodos , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interleucina-15/genética , Camundongos , Camundongos Endogâmicos C57BL , Esferoides Celulares/patologia
12.
In Vitro Cell Dev Biol Anim ; 45(9): 523-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19533253

RESUMO

An in vitro 3D model was developed utilizing a synthetic microgravity environment to facilitate studying the cell interactions. 2D monolayer cell culture models have been successfully used to understand various cellular reactions that occur in vivo. There are some limitations to the 2D model that are apparent when compared to cells grown in a 3D matrix. For example, some proteins that are not expressed in a 2D model are found up-regulated in the 3D matrix. In this paper, we discuss techniques used to develop the first known large, free-floating 3D tissue model used to establish tumor spheroids. The bioreactor system known as the High Aspect Ratio Vessel (HARVs) was used to provide a microgravity environment. The HARVs promoted aggregation of keratinocytes (HaCaT) that formed a construct that served as scaffolding for the growth of mouse melanoma. Although there is an emphasis on building a 3D model with the proper extracellular matrix and stroma, we were able to develop a model that excluded the use of matrigel. Immunohistochemistry and apoptosis assays provided evidence that this 3D model supports B16.F10 cell growth, proliferation, and synthesis of extracellular matrix. Immunofluorescence showed that melanoma cells interact with one another displaying observable cellular morphological changes. The goal of engineering a 3D tissue model is to collect new information about cancer development and develop new potential treatment regimens that can be translated to in vivo models while reducing the use of laboratory animals.


Assuntos
Técnicas de Cultura de Células/métodos , Melanoma/patologia , Modelos Biológicos , Esferoides Celulares/patologia , Ausência de Peso , Animais , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Matriz Extracelular/metabolismo , Imunofluorescência , Humanos , Camundongos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA