Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Haematologica ; 104(9): 1841-1852, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30792198

RESUMO

CD20 monoclonal antibody therapies have significantly improved the outlook for patients with B-cell malignancies. However, many patients acquire resistance, demonstrating the need for new and improved drugs. We previously demonstrated that the natural process of antibody hexamer formation on targeted cells allows for optimal induction of complement-dependent cytotoxicity. Complement-dependent cytotoxicity can be potentiated by introducing a single point mutation such as E430G in the IgG Fc domain that enhances intermolecular Fc-Fc interactions between cell-bound IgG molecules, thereby facilitating IgG hexamer formation. Antibodies specific for CD37, a target that is abundantly expressed on healthy and malignant B cells, are generally poor inducers of complement-dependent cytotoxicity. Here we demonstrate that introduction of the hexamerization-enhancing mutation E430G in CD37-specific antibodies facilitates highly potent complement-dependent cytotoxicity in chronic lymphocytic leukemia cells ex vivo Strikingly, we observed that combinations of hexamerization-enhanced CD20 and CD37 antibodies cooperated in C1q binding and induced superior and synergistic complement-dependent cytotoxicity in patient-derived cancer cells compared to the single agents. Furthermore, CD20 and CD37 antibodies colocalized on the cell membrane, an effect that was potentiated by the hexamerization-enhancing mutation. Moreover, upon cell surface binding, CD20 and CD37 antibodies were shown to form mixed hexameric antibody complexes consisting of both antibodies each bound to their own cognate target, so-called hetero-hexamers. These findings provide novel insights into the mechanisms of synergy in antibody-mediated complement-dependent cytotoxicity and provide a rationale to explore Fc-engineering and antibody hetero-hexamerization as a tool to enhance the cooperativity and therapeutic efficacy of antibody combinations.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD20/imunologia , Antígenos de Neoplasias/imunologia , Proteínas do Sistema Complemento/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Leucemia Linfocítica Crônica de Células B/genética , Tetraspaninas/imunologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Linhagem Celular Tumoral , Complemento C1q/imunologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Imunoglobulina G/imunologia , Leucemia Linfocítica Crônica de Células B/sangue , Mutação , Ligação Proteica , Rituximab/farmacologia
2.
PLoS Biol ; 14(1): e1002344, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26736041

RESUMO

IgG antibodies can organize into ordered hexamers on cell surfaces after binding their antigen. These hexamers bind the first component of complement C1 inducing complement-dependent target cell killing. Here, we translated this natural concept into a novel technology platform (HexaBody technology) for therapeutic antibody potentiation. We identified mutations that enhanced hexamer formation and complement activation by IgG1 antibodies against a range of targets on cells from hematological and solid tumor indications. IgG1 backbones with preferred mutations E345K or E430G conveyed a strong ability to induce conditional complement-dependent cytotoxicity (CDC) of cell lines and chronic lymphocytic leukemia (CLL) patient tumor cells, while retaining regular pharmacokinetics and biopharmaceutical developability. Both mutations potently enhanced CDC- and antibody-dependent cellular cytotoxicity (ADCC) of a type II CD20 antibody that was ineffective in complement activation, while retaining its ability to induce apoptosis. The identified IgG1 Fc backbones provide a novel platform for the generation of therapeutics with enhanced effector functions that only become activated upon binding to target cell-expressed antigen.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Imunoglobulina G/metabolismo , Imunoterapia/métodos , Animais , Linhagem Celular Tumoral , Ativação do Complemento , Feminino , Humanos , Imunoglobulina G/genética , Camundongos SCID , Mutação , Transplante de Neoplasias , Polimerização
3.
Proc Natl Acad Sci U S A ; 110(13): 5145-50, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23479652

RESUMO

The promise of bispecific antibodies (bsAbs) to yield more effective therapeutics is well recognized; however, the generation of bsAbs in a practical and cost-effective manner has been a formidable challenge. Here we present a technology for the efficient generation of bsAbs with normal IgG structures that is amenable to both antibody drug discovery and development. The process involves separate expression of two parental antibodies, each containing single matched point mutations in the CH3 domains. The parental antibodies are mixed and subjected to controlled reducing conditions in vitro that separate the antibodies into HL half-molecules and allow reassembly and reoxidation to form highly pure bsAbs. The technology is compatible with standard large-scale antibody manufacturing and ensures bsAbs with Fc-mediated effector functions and in vivo stability typical of IgG1 antibodies. Proof-of-concept studies with HER2×CD3 (T-cell recruitment) and HER2×HER2 (dual epitope targeting) bsAbs demonstrate superior in vivo activity compared with parental antibody pairs.


Assuntos
Anticorpos Biespecíficos/biossíntese , Fragmentos Fab das Imunoglobulinas/biossíntese , Fragmentos Fc das Imunoglobulinas/biossíntese , Imunoglobulina G/biossíntese , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/genética , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/química , Imunoglobulina G/genética , Células Jurkat , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
4.
Mol Cancer Ther ; 19(10): 2126-2138, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32847982

RESUMO

Higher-order death receptor 5 (DR5) clustering can induce tumor cell death; however, therapeutic compounds targeting DR5 have achieved limited clinical efficacy. We describe HexaBody-DR5/DR5, an equimolar mixture of two DR5-specific IgG1 antibodies with an Fc-domain mutation that augments antibody hexamerization after cell surface target binding. The two antibodies do not compete for binding to DR5 as demonstrated using binding competition studies, and binding to distinct epitopes in the DR5 extracellular domain was confirmed by crystallography. The unique combination of dual epitope targeting and increased IgG hexamerization resulted in potent DR5 agonist activity by inducing efficient DR5 outside-in signaling and caspase-mediated cell death. Preclinical studies in vitro and in vivo demonstrated that maximal DR5 agonist activity could be achieved independent of Fc gamma receptor-mediated antibody crosslinking. Most optimal agonism was observed in the presence of complement complex C1, although without inducing complement-dependent cytotoxicity. It is hypothesized that C1 may stabilize IgG hexamers that are formed after binding of HexaBody-DR5/DR5 to DR5 on the plasma membrane, thereby strengthening DR5 clustering and subsequent outside-in signaling. We observed potent antitumor activity in vitro and in vivo in large panels of patient-derived xenograft models representing various solid cancers. The results of our preclinical studies provided the basis for an ongoing clinical trial exploring the activity of HexaBody-DR5/DR5 (GEN1029) in patients with malignant solid tumors.


Assuntos
Epitopos/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos
5.
Blood Cancer J ; 10(3): 30, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32341336

RESUMO

Tetraspanin CD37 has recently received renewed interest as a therapeutic target for B-cell malignancies. Although complement-dependent cytotoxicity (CDC) is a powerful Fc-mediated effector function for killing hematological cancer cells, CD37-specific antibodies are generally poor inducers of CDC. To enhance CDC, the E430G mutation was introduced into humanized CD37 monoclonal IgG1 antibodies to drive more efficient IgG hexamer formation through intermolecular Fc-Fc interactions after cell surface antigen binding. DuoHexaBody-CD37, a bispecific CD37 antibody with the E430G hexamerization-enhancing mutation targeting two non-overlapping epitopes on CD37 (biparatopic), demonstrated potent and superior CDC activity compared to other CD37 antibody variants evaluated, in particular ex vivo in patient-derived chronic lymphocytic leukemia cells. The superior CDC potency was attributed to enhanced IgG hexamerization mediated by the E430G mutation in combination with dual epitope targeting. The mechanism of action of DuoHexaBody-CD37 was shown to be multifaceted, as it was additionally capable of inducing efficient antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vitro. Finally, potent anti-tumor activity in vivo was observed in cell line- and patient-derived xenograft models from different B-cell malignancy subtypes. These encouraging preclinical results suggest that DuoHexaBody-CD37 (GEN3009) may serve as a potential therapeutic antibody for the treatment of human B-cell malignancies.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antígenos de Neoplasias/imunologia , Leucemia Linfocítica Crônica de Células B/terapia , Linfoma de Células B/terapia , Receptores Fc/imunologia , Tetraspaninas/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/patologia , Linhagem Celular Tumoral , Desenvolvimento de Medicamentos , Células HEK293 , Xenoenxertos , Humanos , Imunoglobulina G/imunologia , Leucemia Linfocítica Crônica de Células B/imunologia , Linfoma de Células B/imunologia , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Receptores Fc/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia
6.
Leuk Res ; 32(1): 113-20, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17521720

RESUMO

Mutation of the tumor suppressor PTEN results in loss of its PI3-kinase counteracting function. PI3-kinase stimulates tumor formation by PKB/Akt-mediated cell proliferation and prevention of apoptosis. PI3-kinase may also activate Rho-GTPases and their regulatory GEFs to promote invasion. Here we have analyzed the function of the Rac-specific activator, Tiam1, in PI3-kinase-induced T-lymphomagenesis. Mice with a T cell-specific Pten deletion developed T-lymphomas with enhanced PKB/Akt phosphorylation. However, these T-lymphomas infiltrated more frequently into various organs in Tiam1-deficient mice compared to wild type mice. Surprisingly, Tiam1-deficient lymphomas showed increased Rac activity, suggesting that the lack of Tiam1 is compensated by alternative Rac-activating mechanisms that lead to increased progression of PI3-kinase-induced T-lymphomas.


Assuntos
Deleção de Genes , Fatores de Troca do Nucleotídeo Guanina/farmacologia , Linfoma de Células T/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Progressão da Doença , Linfoma de Células T/patologia , Camundongos , Camundongos Mutantes , Fosfatidilinositol 3-Quinases/farmacologia , Transdução de Sinais , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
7.
Cancer Res ; 66(20): 10007-15, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17047063

RESUMO

Down-regulation of the epithelial cell-cell adhesion molecule E-cadherin is frequently associated with tumor formation and progression. Besides its role in physical cell-cell adhesion, E-cadherin is also thought to be involved in intracellular signaling in normal epithelial cells. In these cells, the Armadillo catenin p120ctn binds to the cytoplasmic domain of E-cadherin and stabilizes the adhesion complexes. On loss of E-cadherin, cytoplasmic p120ctn might accumulate and contribute to tumor malignancy. We used suppression subtractive hybridization to search for genes regulated by E-cadherin expression. We isolated human Nanos1 as a transcript of which levels decrease on E-cadherin reexpression in a human breast cancer cell line. The hNanos1 protein bears a COOH-terminal (CCHC)(2) zinc finger domain and belongs to an evolutionarily conserved protein family sharing functions in germ cell development in both vertebrates and invertebrates. We found an inverse correlation between E-cadherin and hNanos1 expression in various cell lines and under diverse conditions. Conditional expression of hNanos1 in human colorectal DLD1 cancer cells functionally abolished cell-cell adhesion. It induced cytoplasmic translocation of p120ctn, as well as strong migratory and invasive properties. We also found that the NH(2)-terminal domain of hNanos1, which is conserved only among mammals, interacts with p120ctn. hNanos1 counteracted the stimulatory effect of p120ctn on cell protrusion formation. Together, these findings describe a new function for hNanos1 as a downstream effector of E-cadherin loss contributing to tumor progression. Targeting hNanos1 might be a promising strategy in the treatment of E-cadherin-negative tumors in particular.


Assuntos
Caderinas/genética , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Tatus , Caderinas/biossíntese , Caderinas/metabolismo , Cateninas , Adesão Celular/fisiologia , Moléculas de Adesão Celular/biossíntese , Linhagem Celular Tumoral , Clonagem Molecular , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfoproteínas/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Dedos de Zinco , delta Catenina
8.
Methods Enzymol ; 407: 269-81, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16757331

RESUMO

The Tiam1 gene encodes a guanine nucleotide exchange factor (GEF) that specifically activates the Rho-like GTPase Rac. In vitro studies indicate that Tiam1 localizes to adherens junctions and plays a role in the formation and maintenance of cadherin-based cell adhesions, thereby regulating migration of epithelial cells. In vivo studies implicate Tiam1 in various aspects of tumorigenesis. In this chapter, we discuss the use of the DMBA/TPA chemical carcinogenesis protocol in Tiam1-deficient mice to study the role of Tiam1 in Ras-induced skin tumors. This two-stage carcinogenesis protocol allows us to study initiation, promotion, and progression of tumors in a Tiam1-positive and Tiam1-negative background. Moreover, we describe methods to study the role of Tiam1 in susceptibility to apoptosis, cell growth, and Ras transformation by in vivo and in vitro experiments. The latter makes use of tumor cells and primary embryonic fibroblasts and keratinocytes isolated from mice.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas ras/fisiologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Apoptose/fisiologia , Separação Celular , Regulação para Baixo , Queratinócitos/citologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/patologia , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
9.
Mol Immunol ; 67(2 Pt A): 117-30, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25697848

RESUMO

Complement is recognized as a key player in a wide range of normal as well as disease-related immune, developmental and homeostatic processes. Knowledge of complement components, structures, interactions, and cross-talk with other biological systems continues to grow and this leads to novel treatments for cancer, infectious, autoimmune- or age-related diseases as well as for preventing transplantation rejection. Antibodies are superbly suited to be developed into therapeutics with appropriate complement stimulatory or inhibitory activity. Here we review the design, development and future of antibody-based drugs that enhance or dampen the complement system.


Assuntos
Anticorpos/imunologia , Ativação do Complemento/imunologia , Inativadores do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Anticorpos/uso terapêutico , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/uso terapêutico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/terapia , Humanos , Inflamação/imunologia , Inflamação/terapia , Modelos Imunológicos , Neoplasias/imunologia , Neoplasias/terapia
10.
BMC Genomics ; 5(1): 11, 2004 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15018647

RESUMO

BACKGROUND: Activation of proto-oncogenes by DNA amplification is an important mechanism in the development and maintenance of cancer cells. Until recently, identification of the targeted genes relied on labour intensive and time consuming positional cloning methods. In this study, we outline a straightforward and efficient strategy for fast and comprehensive cloning of amplified and overexpressed genes. RESULTS: As a proof of principle, we analyzed neuroblastoma cell line IMR-32, with at least two amplification sites along the short arm of chromosome 2. In a first step, overexpressed cDNA clones were isolated using a PCR based subtractive cloning method. Subsequent deposition of these clones on a custom microarray and hybridization with IMR-32 DNA, resulted in the identification of clones that were overexpressed due to gene amplification. Using this approach, amplification of all previously reported amplified genes in this cell line was detected. Furthermore, four additional clones were found to be amplified, including the TEM8 gene on 2p13.3, two anonymous transcripts, and a fusion transcript, resulting from 2p13.3 and 2p24.3 fused sequences. CONCLUSIONS: The combinatorial strategy of subtractive cDNA cloning and array CGH analysis allows comprehensive amplicon dissection, which opens perspectives for improved identification of hitherto unknown targeted oncogenes in cancer cells.


Assuntos
DNA Complementar/genética , Perfilação da Expressão Gênica/métodos , Hibridização de Ácido Nucleico/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Linhagem Celular Tumoral , Cromossomos Humanos Par 2/genética , Clonagem Molecular/métodos , RNA Helicases DEAD-box , DNA/genética , DNA Complementar/química , Feminino , Amplificação de Genes , Proteínas de Homeodomínio/genética , Humanos , Hibridização in Situ Fluorescente , Masculino , Proteína Meis1 , Proteína Proto-Oncogênica N-Myc , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , RNA Helicases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
11.
MAbs ; 6(2): 392-402, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24492309

RESUMO

The human epidermal growth factor receptor (HER)2 provides an excellent target for selective delivery of cytotoxic drugs to tumor cells by antibody-drug conjugates (ADC) as has been clinically validated by ado-trastuzumab emtansine (Kadcyla(TM)). While selecting a suitable antibody for an ADC approach often takes specificity and efficient antibody-target complex internalization into account, the characteristics of the optimal antibody candidate remain poorly understood. We studied a large panel of human HER2 antibodies to identify the characteristics that make them most suitable for an ADC approach. As a model toxin, amenable to in vitro high-throughput screening, we employed Pseudomonas exotoxin A (ETA') fused to an anti-kappa light chain domain antibody. Cytotoxicity induced by HER2 antibodies, which were thus non-covalently linked to ETA', was assessed for high and low HER2 expressing tumor cell lines and correlated with internalization and downmodulation of HER2 antibody-target complexes. Our results demonstrate that HER2 antibodies that do not inhibit heterodimerization of HER2 with related ErbB receptors internalize more efficiently and show greater ETA'-mediated cytotoxicity than antibodies that do inhibit such heterodimerization. Moreover, stimulation with ErbB ligand significantly enhanced ADC-mediated tumor kill by antibodies that do not inhibit HER2 heterodimerization. This suggests that the formation of HER2/ErbB-heterodimers enhances ADC internalization and subsequent killing of tumor cells. Our study indicates that selecting HER2 ADCs that allow piggybacking of HER2 onto other ErbB receptors provides an attractive strategy for increasing ADC delivery and tumor cell killing capacity to both high and low HER2 expressing tumor cells.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/terapia , Desenho de Fármacos , Imunoterapia/métodos , Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Dimerização , Endocitose , Fator de Crescimento Epidérmico/metabolismo , Feminino , Humanos , Imunotoxinas/uso terapêutico , Agregação de Receptores/efeitos dos fármacos , Receptor ErbB-2/imunologia , Transdução de Sinais/efeitos dos fármacos , Trastuzumab
12.
Science ; 343(6176): 1260-3, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24626930

RESUMO

Complement activation by antibodies bound to pathogens, tumors, and self antigens is a critical feature of natural immune defense, a number of disease processes, and immunotherapies. How antibodies activate the complement cascade, however, is poorly understood. We found that specific noncovalent interactions between Fc segments of immunoglobulin G (IgG) antibodies resulted in the formation of ordered antibody hexamers after antigen binding on cells. These hexamers recruited and activated C1, the first component of complement, thereby triggering the complement cascade. The interactions between neighboring Fc segments could be manipulated to block, reconstitute, and enhance complement activation and killing of target cells, using all four human IgG subclasses. We offer a general model for understanding antibody-mediated complement activation and the design of antibody therapeutics with enhanced efficacy.


Assuntos
Membrana Celular/imunologia , Ativação do Complemento , Complemento C1/imunologia , Imunoglobulina G/química , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Lipossomos , Conformação Proteica , Multimerização Proteica
13.
J Cell Sci ; 121(Pt 8): 1183-92, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18349077

RESUMO

Tiam1 is a ubiquitously expressed activator of the small GTPase Rac. Previously, we found that Tiam1 knockout (KO) mice are resistant to DMBA-induced skin tumorigenicity, which correlated with increased apoptosis in keratinocytes of the skin epidermis. Here, we have studied the mechanisms by which Tiam1 protects against apoptosis. We found that Tiam1-KO keratinocytes show increased apoptosis in response to apoptotic stimuli, including growth factor deprivation and heat-shock treatment. Expression of catalytically active Tiam1, but not inactive Tiam1, rescues the apoptosis susceptibility of Tiam1-KO keratinocytes, indicating that this defect is caused by impaired Tiam1-mediated Rac activation. Apoptosis induced by growth factor starvation correlates with impaired ERK phosphorylation in Tiam1-KO keratinocytes. Moreover, Tiam1-KO keratinocytes contain lower levels of intracellular reactive oxygen species (ROS) when compared with wild-type cells. The ROS content of keratinocytes is dependent on both Tiam1 and the activity of NADPH oxidase (Nox), and is required for ERK-mediated survival signaling. Indeed, Tiam1 deficiency or the inhibition of intracellular ROS production blocks ERK phosphorylation and sensitizes wild-type keratinocytes to apoptotic stimuli. Our results indicate that the Rac activator Tiam1 controls the intracellular redox balance by Nox-mediated ROS production, which regulates ERK phosphorylation and the susceptibility of keratinocytes to apoptotic signaling.


Assuntos
Apoptose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Queratinócitos/metabolismo , Animais , Animais Recém-Nascidos , Sobrevivência Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Queratinócitos/citologia , Camundongos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA