Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38805257

RESUMO

Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH-dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Macrófagos , Mycobacterium tuberculosis , Fagossomos , Anticorpos de Domínio Único , Humanos , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Concentração de Íons de Hidrogênio , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/metabolismo , Fagossomos/metabolismo , Anticorpos de Domínio Único/metabolismo
2.
Open Biol ; 10(2): 190235, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32019478

RESUMO

Cancer-specific mutations can lead to peptides of unique sequence presented on MHC class I to CD8 T cells. These neoantigens can be potent tumour-rejection antigens, appear to be the driving force behind responsiveness to anti-CTLA-4 and anti-PD1/L1-based therapies and have been used to develop personalized vaccines. The platform for delivering neoantigen-based vaccines has varied, and further optimization of both platform and adjuvant will be necessary to achieve scalable vaccine products that are therapeutically effective at a reasonable cost. Here, we developed a platform for testing potential CD8 T cell tumour vaccine candidates. We used a high-affinity alpaca-derived VHH against MHC class II to deliver peptides to professional antigen-presenting cells. We show in vitro and in vivo that peptides derived from the model antigen ovalbumin are better able to activate naive ovalbumin-specific CD8 T cells when conjugated to an MHC class II-specific VHH when compared with an irrelevant control VHH. We then used the VHH-peptide platform to evaluate a panel of candidate neoantigens in vivo in a mouse model of pancreatic cancer. None of the candidate neoantigens tested led to protection from tumour challenge; however, we were able to show vaccine-induced CD8 T cell responses to a melanoma self-antigen that was augmented by combination therapy with the synthetic cytokine mimetic Neo2/15.


Assuntos
Antígenos de Histocompatibilidade Classe II/imunologia , Interleucina-2/administração & dosagem , Melanoma/tratamento farmacológico , Peptídeos/administração & dosagem , Anticorpos de Domínio Único/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Camelídeos Americanos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Interleucina-2/imunologia , Melanoma/imunologia , Camundongos , Peptídeos/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Vacinas de Subunidades Antigênicas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Immunol Res ; 8(4): 518-529, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32019780

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy is effective in the treatment of cancers of hematopoietic origin. In the immunosuppressive solid tumor environment, CAR T cells encounter obstacles that compromise their efficacy. We developed a strategy to address these barriers by having CAR T cells secrete single-domain antibody fragments [variable heavy domain of heavy chain antibodies (VHH) or nanobodies] that can modify the intratumoral immune landscape and thus support CAR T-cell function in immunocompetent animals. VHHs are small in size and able to avoid domain swapping when multiple nanobodies are expressed simultaneously-features that can endow CAR T cells with desirable properties. The secretion of an anti-CD47 VHH by CAR T cells improves engagement of the innate immune system, enables epitope spreading, and can enhance the antitumor response. CAR T cells that secrete anti-PD-L1 or anti-CTLA-4 nanobodies show improved persistence and demonstrate the versatility of this approach. Furthermore, local delivery of secreted anti-CD47 VHH-Fc fusions by CAR T cells at the tumor site limits their systemic toxicity. CAR T cells can be further engineered to simultaneously secrete multiple modalities, allowing for even greater tailoring of the antitumor immune response.


Assuntos
Antígeno CD47/antagonistas & inibidores , Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Receptores de Antígenos Quiméricos/imunologia , Proteínas Recombinantes/genética , Anticorpos de Domínio Único/imunologia , Animais , Linhagem Celular Tumoral , Granzimas/antagonistas & inibidores , Melanoma Experimental/genética , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores de Antígenos Quiméricos/genética , Proteínas Recombinantes/metabolismo , Anticorpos de Domínio Único/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Microambiente Tumoral
4.
Proc Natl Acad Sci U S A ; 116(28): 14181-14190, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31068469

RESUMO

Extracellular matrix (ECM) deposition is a hallmark of many diseases, including cancer and fibroses. To exploit the ECM as an imaging and therapeutic target, we developed alpaca-derived libraries of "nanobodies" against disease-associated ECM proteins. We describe here one such nanobody, NJB2, specific for an alternatively spliced domain of fibronectin expressed in disease ECM and neovasculature. We showed by noninvasive in vivo immuno-PET/CT imaging that NJB2 detects primary tumors and metastatic sites with excellent specificity in multiple models of breast cancer, including human and mouse triple-negative breast cancer, and in melanoma. We also imaged mice with pancreatic ductal adenocarcinoma (PDAC) in which NJB2 was able to detect not only PDAC tumors but also early pancreatic lesions called pancreatic intraepithelial neoplasias, which are challenging to detect by any current imaging modalities, with excellent clarity and signal-to-noise ratios that outperformed conventional 2-fluorodeoxyglucose PET/CT imaging. NJB2 also detected pulmonary fibrosis in a bleomycin-induced fibrosis model. We propose NJB2 and similar anti-ECM nanobodies as powerful tools for noninvasive detection of tumors, metastatic lesions, and fibroses. Furthermore, the selective recognition of disease tissues makes NJB2 a promising candidate for nanobody-based therapeutic applications.


Assuntos
Carcinogênese/genética , Carcinoma Ductal Pancreático/diagnóstico por imagem , Matriz Extracelular/efeitos dos fármacos , Neoplasias Pancreáticas/diagnóstico por imagem , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Fibrose/patologia , Humanos , Masculino , Camundongos , Neoplasias Pancreáticas/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Compostos Radiofarmacêuticos/farmacologia , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia , Neoplasias Pancreáticas
5.
Annu Rev Cell Dev Biol ; 34: 163-188, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30110557

RESUMO

Molecular biologists and chemists alike have long sought to modify proteins with substituents that cannot be installed by standard or even advanced genetic approaches. We here describe the use of transpeptidases to achieve these goals. Living systems encode a variety of transpeptidases and peptide ligases that allow for the enzyme-catalyzed formation of peptide bonds, and protein engineers have used directed evolution to enhance these enzymes for biological applications. We focus primarily on the transpeptidase sortase A, which has become popular over the past few years for its ability to perform a remarkably wide variety of protein modifications, both in vitro and in living cells.


Assuntos
Aminoaciltransferases/genética , Proteínas de Bactérias/genética , Cisteína Endopeptidases/genética , Peptídeos/genética , Peptidil Transferases/genética , Sequência de Aminoácidos/genética , Aminoaciltransferases/química , Proteínas de Bactérias/química , Catálise , Cisteína Endopeptidases/química , Humanos , Peptídeos/química , Peptidil Transferases/química , Engenharia de Proteínas , Especificidade por Substrato
6.
Proc Natl Acad Sci U S A ; 115(15): 3912-3917, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581255

RESUMO

Ipilimumab, a monoclonal antibody that recognizes cytotoxic T lymphocyte antigen (CTLA)-4, was the first approved "checkpoint"-blocking anticancer therapy. In mouse tumor models, the response to antibodies against CTLA-4 depends entirely on expression of the Fcγ receptor (FcγR), which may facilitate antibody-dependent cellular phagocytosis, but the contribution of simple CTLA-4 blockade remains unknown. To understand the role of CTLA-4 blockade in the complete absence of Fc-dependent functions, we developed H11, a high-affinity alpaca heavy chain-only antibody fragment (VHH) against CTLA-4. The VHH H11 lacks an Fc portion, binds monovalently to CTLA-4, and inhibits interactions between CTLA-4 and its ligand by occluding the ligand-binding motif on CTLA-4 as shown crystallographically. We used H11 to visualize CTLA-4 expression in vivo using whole-animal immuno-PET, finding that surface-accessible CTLA-4 is largely confined to the tumor microenvironment. Despite this, H11-mediated CTLA-4 blockade has minimal effects on antitumor responses. Installation of the murine IgG2a constant region on H11 dramatically enhances its antitumor response. Coadministration of the monovalent H11 VHH blocks the efficacy of a full-sized therapeutic antibody. We were thus able to demonstrate that CTLA-4-binding antibodies require an Fc domain for antitumor effect.


Assuntos
Antígeno CTLA-4/imunologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/administração & dosagem , Neoplasias/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Antígeno CTLA-4/química , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/imunologia , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Domínios Proteicos
7.
Cancer Immunol Res ; 6(4): 389-401, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29459478

RESUMO

Cytokine-based therapies for cancer have not achieved widespread clinical success because of inherent toxicities. Treatment for pancreatic cancer is limited by the dense stroma that surrounds tumors and by an immunosuppressive tumor microenvironment. To overcome these barriers, we developed constructs of single-domain antibodies (VHHs) against PD-L1 fused with IL-2 and IFNγ. Targeting cytokine delivery in this manner reduced pancreatic tumor burden by 50%, whereas cytokines fused to an irrelevant VHH, or blockade of PD-L1 alone, showed little effect. Targeted delivery of IL-2 increased the number of intratumoral CD8+ T cells, whereas IFNγ reduced the number of CD11b+ cells and skewed intratumoral macrophages toward the display of M1-like characteristics. Imaging of fluorescent VHH-IFNγ constructs, as well as transcriptional profiling, demonstrated targeting of IFNγ to the tumor microenvironment. Many tumors and tumor-infiltrating myeloid cells express PD-L1, rendering them potentially susceptible to this form of targeted immunotherapy. Cancer Immunol Res; 6(4); 389-401. ©2018 AACR.


Assuntos
Antígeno B7-H1/metabolismo , Citocinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Anticorpos de Domínio Único/farmacologia , Microambiente Tumoral , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/genética , Modelos Animais de Doenças , Humanos , Melanoma Experimental , Camundongos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Anticorpos de Domínio Único/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
8.
PLoS One ; 12(12): e0189068, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29200433

RESUMO

Sortase A, a calcium-dependent transpeptidase derived from Staphylococcus aureus, is used in a broad range of applications, such as the conjugation of fluorescent dyes and other moieties to proteins or to the surface of eukaryotic cells. In vivo and cell-based applications of sortase have been somewhat limited by the large range of calcium concentrations, as well as by the often transient nature of protein-protein interactions in living systems. In order to use sortase A for cell labeling applications, we generated a new sortase A variant by combining multiple mutations to yield an enzyme that was both calcium-independent and highly active. This variant has enhanced activity for both N- and C-terminal labeling, as well as for cell surface modification under physiological conditions.


Assuntos
Aminoaciltransferases/genética , Proteínas de Bactérias/genética , Cálcio/metabolismo , Cisteína Endopeptidases/genética , Peptidil Transferases/genética , Coloração e Rotulagem/métodos , Staphylococcus aureus/genética , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/química , Cisteína Endopeptidases/metabolismo , Mutação , Peptidil Transferases/metabolismo , Staphylococcus aureus/enzimologia
9.
Proc Natl Acad Sci U S A ; 114(38): 10184-10189, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28874561

RESUMO

CD47 is an antiphagocytic ligand broadly expressed on normal and malignant tissues that delivers an inhibitory signal through the receptor signal regulatory protein alpha (SIRPα). Inhibitors of the CD47-SIRPα interaction improve antitumor antibody responses by enhancing antibody-dependent cellular phagocytosis (ADCP) in xenograft models. Endogenous expression of CD47 on a variety of cell types, including erythrocytes, creates a formidable antigen sink that may limit the efficacy of CD47-targeting therapies. We generated a nanobody, A4, that blocks the CD47-SIRPα interaction. A4 synergizes with anti-PD-L1, but not anti-CTLA4, therapy in the syngeneic B16F10 melanoma model. Neither increased dosing nor half-life extension by fusion of A4 to IgG2a Fc (A4Fc) overcame the issue of an antigen sink or, in the case of A4Fc, systemic toxicity. Generation of a B16F10 cell line that secretes the A4 nanobody showed that an enhanced response to several immune therapies requires near-complete blockade of CD47 in the tumor microenvironment. Thus, strategies to localize CD47 blockade to tumors may be particularly valuable for immune therapy.


Assuntos
Antígeno CD47/antagonistas & inibidores , Imunoterapia/métodos , Melanoma Experimental/terapia , Anticorpos de Domínio Único/uso terapêutico , Anemia/induzido quimicamente , Animais , Antígeno CD47/imunologia , Avaliação Pré-Clínica de Medicamentos , Camundongos Endogâmicos C57BL , Fagocitose , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/farmacologia , Microambiente Tumoral
10.
Nat Commun ; 8(1): 647, 2017 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-28935898

RESUMO

Programmed death ligand 1 (PD-L1) is expressed on a number of immune and cancer cells, where it can downregulate antitumor immune responses. Its expression has been linked to metabolic changes in these cells. Here we develop a radiolabeled camelid single-domain antibody (anti-PD-L1 VHH) to track PD-L1 expression by immuno-positron emission tomography (PET). PET-CT imaging shows a robust and specific PD-L1 signal in brown adipose tissue (BAT). We confirm expression of PD-L1 on brown adipocytes and demonstrate that signal intensity does not change in response to cold exposure or ß-adrenergic activation. This is the first robust method of visualizing murine brown fat independent of its activation state.Current approaches to visualise brown adipose tissue (BAT) rely primarily on markers that reflect its metabolic activity. Here, the authors show that PD-L1 is expressed on brown adipocytes, does not change upon BAT activation, and that BAT volume in mice can be measured by PET-CT with a radiolabeled anti-PD-L1 antibody.


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Antígeno B7-H1/análise , Biomarcadores/análise , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/diagnóstico por imagem , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Camelídeos Americanos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Reprodutibilidade dos Testes
11.
J Exp Med ; 214(8): 2243-2255, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28666979

RESUMO

Immunotherapy using checkpoint-blocking antibodies against targets such as CTLA-4 and PD-1 can cure melanoma and non-small cell lung cancer in a subset of patients. The presence of CD8 T cells in the tumor correlates with improved survival. We show that immuno-positron emission tomography (immuno-PET) can visualize tumors by detecting infiltrating lymphocytes and, through longitudinal observation of individual animals, distinguish responding tumors from those that do not respond to therapy. We used 89Zr-labeled PEGylated single-domain antibody fragments (VHHs) specific for CD8 to track the presence of intratumoral CD8+ T cells in the immunotherapy-susceptible B16 melanoma model in response to checkpoint blockade. A 89Zr-labeled PEGylated anti-CD8 VHH detected thymus and secondary lymphoid structures as well as intratumoral CD8 T cells. Animals that responded to CTLA-4 therapy showed a homogeneous distribution of the anti-CD8 PET signal throughout the tumor, whereas more heterogeneous infiltration of CD8 T cells correlated with faster tumor growth and worse responses. To support the validity of these observations, we used two different transplantable breast cancer models, yielding results that conformed with predictions based on the antimelanoma response. It may thus be possible to use immuno-PET and monitor antitumor immune responses as a prognostic tool to predict patient responses to checkpoint therapies.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Antígeno CTLA-4/antagonistas & inibidores , Neoplasias Mamárias Experimentais/terapia , Animais , Antígeno CTLA-4/fisiologia , Feminino , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons/métodos , Resultado do Tratamento
12.
Proc Natl Acad Sci U S A ; 113(19): E2646-54, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27091975

RESUMO

Therapeutic antitumor antibodies treat cancer by mobilizing both innate and adaptive immunity. CD47 is an antiphagocytic ligand exploited by tumor cells to blunt antibody effector functions by transmitting an inhibitory signal through its receptor signal regulatory protein alpha (SIRPα). Interference with the CD47-SIRPα interaction synergizes with tumor-specific monoclonal antibodies to eliminate human tumor xenografts by enhancing macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), but synergy between CD47 blockade and ADCP has yet to be demonstrated in immunocompetent hosts. Here, we show that CD47 blockade alone or in combination with a tumor-specific antibody fails to generate antitumor immunity against syngeneic B16F10 tumors in mice. Durable tumor immunity required programmed death-ligand 1 (PD-L1) blockade in combination with an antitumor antibody, with incorporation of CD47 antagonism substantially improving response rates. Our results highlight an underappreciated contribution of the adaptive immune system to anti-CD47 adjuvant therapy and suggest that targeting both innate and adaptive immune checkpoints can potentiate the vaccinal effect of antitumor antibody therapy.


Assuntos
Anticorpos Monoclonais Murinos/administração & dosagem , Antígeno CD47/efeitos dos fármacos , Antígeno CD47/imunologia , Vacinas Anticâncer/administração & dosagem , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/imunologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Vacinas Anticâncer/imunologia , Feminino , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Neoplasias Experimentais/patologia , Resultado do Tratamento
13.
Nat Protoc ; 10(3): 508-16, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25719269

RESUMO

Transpeptidation catalyzed by sortase A allows the preparation of proteins that are site-specifically and homogeneously modified with a wide variety of functional groups, such as fluorophores, PEG moieties, lipids, glycans, bio-orthogonal reactive groups and affinity handles. This protocol describes immobilization of sortase A on a solid support (Sepharose beads). Immobilization of sortase A simplifies downstream purification of a protein of interest after labeling of its N or C terminus. Smaller batch and larger-scale continuous-flow reactions require only a limited amount of enzyme. The immobilized enzyme can be reused for multiple cycles of protein modification reactions. The described protocol also works with a Ca(2+)-independent variant of sortase A with increased catalytic activity. This heptamutant variant of sortase A (7M) was generated by combining previously published mutations, and this immobilized enzyme can be used for the modification of calcium-senstive substrates or in instances in which low temperatures are needed. Preparation of immobilized sortase A takes 1-2 d. Batch reactions take 3-12 h and flow reactions proceed at 0.5 ml h(-1), depending on the geometry of the reactor used.


Assuntos
Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Enzimas Imobilizadas/metabolismo , Peptidil Transferases/metabolismo , Engenharia de Proteínas/métodos , Proteínas/metabolismo , Aminoaciltransferases/genética , Proteínas de Bactérias/genética , Catálise , Cisteína Endopeptidases/genética , Mutação/genética , Sefarose
14.
ACS Chem Biol ; 10(2): 460-5, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25360987

RESUMO

Surface display of engineered proteins has many useful applications. The expression of a synthetic chimeric antigen receptor composed of an extracellular tumor-specific antibody fragment linked to a cytosolic activating motif in engineered T cells is now considered a viable approach for the treatment of leukemias. The risk of de novo tumor development, inherent in the transfer of genetically engineered cells, calls for alternative approaches for the functionalization of the lymphocyte plasma membrane. We demonstrate the conjugation of LPXTG-tagged probes and LPXTG-bearing proteins to endogenous acceptors at the plasma membrane in a single step using sortase A. We successfully conjugated biotin probes not only to mouse hematopoietic cells but also to yeast cells, 293T cells, and Toxoplasma gondii. Installation of single domain antibodies on activated CD8 T cell redirects cell-specific cytotoxicity to cells that bear the relevant antigen. Likewise, conjugation of Toxoplasma gondii with single domain antibodies targets the pathogen to cells that express the antigen recognized by these single domain antibodies. This simple and robust enzymatic approach enables engineering of the plasma membrane for research or therapy under physiological reaction conditions that ensure the viability of the modified cells.


Assuntos
Anticorpos Antiprotozoários/fisiologia , Proteínas de Protozoários/metabolismo , Toxoplasma/citologia , Toxoplasma/fisiologia , Animais , Antígenos , Bioengenharia , Linfócitos T CD8-Positivos , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA