Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
BMC Biotechnol ; 17(1): 42, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28499450

RESUMO

BACKGROUND: Polysialic acid (polySia) is a carbohydrate modification of the neural cell adhesion molecule (NCAM), which is implicated in neural differentiation and plays an important role in tumor development and metastasis. Polysialylation of NCAM is mediated by two Golgi-resident polysialyltransferases (polyST) ST8SiaII and ST8SiaIV. Intracellular antibodies (intrabodies; IB) expressed inside the ER and retaining proteins passing the ER such as cell surface receptors or secretory proteins provide an efficient means of protein knockdown. To inhibit the function of ST8SiaII and ST8SiaIV specific ER IBs were generated starting from two corresponding hybridoma clones. Both IBs αST8SiaII-IB and αST8SiaIV-IB were constructed in the scFv format and their functions characterized in vitro and in vivo. RESULTS: IBs directed against the polySTs prevented the translocation of the enzymes from the ER to the Golgi-apparatus. Co-immunoprecipitation of ST8SiaII and ST8SiaIV with the corresponding IBs confirmed the intracellular interaction with their cognate antigens. In CHO cells overexpressing ST8SiaII and ST8SiaIV, respectively, the transfection with αST8SiaII-IB or αST8SiaIV-IB inhibited significantly the cell surface expression of polysialylated NCAM. Furthermore stable expression of ST8SiaII-IB, ST8SiaIV-IB and luciferase in the rhabdomyosarcoma cell line TE671 reduced cell surface expression of polySia and delayed tumor growth if cells were xenografted into C57BL/6 J RAG-2 mice. CONCLUSION: Data obtained strongly indicate that αST8SiaII-IB and αST8SiaIV-IB are promising experimental tools to analyze the individual role of the two enzymes during brain development and during migration and proliferation of tumor cells.


Assuntos
Anticorpos/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Ácidos Siálicos/metabolismo , Sialiltransferases/metabolismo , Animais , Anticorpos/genética , Anticorpos/imunologia , Sequência de Bases , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Moléculas de Adesão de Célula Nervosa/imunologia , Plasmídeos/genética , Plasmídeos/metabolismo , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Sialiltransferases/genética , Sialiltransferases/imunologia , Transplante Heterólogo
2.
Adv Exp Med Biol ; 917: 77-93, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27236553

RESUMO

ER intrabodies are recombinant antibody fragments produced and retained in the endoplasmatic reticulum (ER) of a cell or an organism with the purpose to induce phenotypes generated by interfering with the intracellular processing or by changing the location of the recognized antigen. The most common application is the generation of functional knockdowns of membrane proteins, which cannot reach their natural location on the cell surface when they are retained in the ER by the intrabody. Phenotypes generated by interfering with the secretion of extracellular or plasma proteins can be analyzed in a similar way. So far, most ER intrabody studies relied on scFv fragments subcloned from hybridoma lines. Recently, several large international research consortia have started to provide antibodies, with the final goal to cover substantial parts of the human proteome. For practical reasons of throughput and effort, in these consortia the most appropriate method to generate the necessary large numbers of monoclonal antibodies is in vitro selection, typically employing phage or yeast display. These methods provide the antibody genes right from the start, thereby facilitating the application of ER antibody approaches. On the other end, the first transgenic mice expressing an ER intrabody has recently been described. This moves the ER intrabody approach finally to level with classic in vivo knockout strategies - but also offers novel capabilities to the researchers. Promising new perspectives may originate from the fact that the knockdown is restricted to the protein level, that a graded knockdown strength can be achieved, or that the targeting of individual posttranslational modifications will be possible with previously impossible specificity. Finally, the link of today's high throughput recombinant antibody generation to a knock down phenotype is now possible with a single cloning step. It can therefore be expected that we will see a much quicker growth of the number of successful applications of ER intrabody technology in the near future than it has been seen in its first two decades.


Assuntos
Anticorpos/imunologia , Núcleo Celular/metabolismo , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Técnicas de Silenciamento de Genes/métodos , Proteoma/imunologia , Animais , Anticorpos/genética , Anticorpos/metabolismo , Núcleo Celular/imunologia , Humanos , Camundongos
3.
Antibodies (Basel) ; 13(1)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38390872

RESUMO

Pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) remains a deadly cancer worldwide with a need for new therapeutic approaches. A dysregulation in the equilibrium between pro- and anti-inflammatory responses with a predominant immunosuppressive inflammatory reaction in advanced stage tumors seem to contribute to tumor growth and metastasis. The current therapies do not include strategies against pro-tumorigenic inflammation in cancer patients. We have shown that the upregulated cell surface expression of Toll-like Receptor (TLR) 2 and of TLR9 inside PDAC cells maintain chronic inflammatory responses, support chemotherapeutic resistance, and mediate tumor progression in human pancreatic cancer. We further demonstrated intracellular TLR2 and TLR9 targeting using specific intrabodies, which resulted in downregulated inflammatory signaling. In this study, we tested, for the first time, an intrabody-mediated TLR blockade in human TLR2- and TLR9-expressing pancreatic cancer cells for its effects on inflammatory signaling-mediated tumor growth. Newly designed anti-TLR2- and anti-TLR9-specific intrabodies inhibited PDAC growth. Co-expression analysis of the intrabodies and corresponding human TLRs showed efficient retention and accumulation of both intrabodies within the endoplasmic reticulum (ER), while co-immunoprecipitation studies indicated both intrabodies interacting with their cognate TLR antigen within the pancreatic cancer cells. Cancer cells with attenuated proliferation expressing accumulated TLR2 and TRL9 intrabodies demonstrated reduced STAT3 phosphorylation signaling, while apoptotic markers Caspases 3 and 8 were upregulated. To conclude, our results demonstrate the TLR2 and TLR9-specific intrabody-mediated signaling pathway inhibition of autoregulatory inflammation inside cancer cells and their proliferation, resulting in the suppression of pancreatic tumor cell growth. These findings underscore the potential of specific intrabody-mediated TLR inhibition in the ER relevant for tumor growth inhibition and open up a new therapeutic intervention strategy for the treatment of pancreatic cancer.

4.
Methods ; 56(3): 338-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22037249

RESUMO

Intrabodies are recombinantly expressed intracellular antibody fragments that can be used to specifically bind and inhibit the function of cellular proteins of interest. Intrabodies can be targeted to various cell compartments by attaching an appropriate localization peptide sequence to them. An efficient strategy with a high success rate is to anchor intrabodies in the endoplasmatic reticulum where they can inhibit transitory target proteins by binding and preventing them to reach their site of action. Intrabodies can be assembled from antibody gene fragments from various sources into dedicated expression vectors. Conventionally, antibody cDNA sequences are derived from selected hybridoma cell clones that express antibodies with the desired specificity. Alternatively, appropriate clones can be isolated by affinity selection from an antibody in vitro display library. Here an evaluation of endoplasmatic reticulum targeted intrabodies with respect to other knockdown approaches is given and the characteristics of various intrabody expression vectors are discussed. A step by step protocol is provided that was repeatedly used to construct intrabodies derived from diverse antibody isotypes producing hybridoma cell clones. The inactivation of the cell surface receptor neural cell adhesion molecule (NCAM) by a highly efficacious novel endoplasmatic reticulum-anchored intrabody is demonstrated.


Assuntos
Anticorpos/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Técnicas de Transferência de Genes , Vetores Genéticos , Recombinação Homóloga , Humanos , Dados de Sequência Molecular , Mutação , Moléculas de Adesão de Célula Nervosa/antagonistas & inibidores , Biblioteca de Peptídeos , Proteínas/fisiologia , RNA Antissenso
5.
Cell Mol Biol Lett ; 18(3): 433-46, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23893288

RESUMO

Toll-like receptor 9 (TLR9) is a component of the innate immune system, which recognizes the DNA of both pathogens and hosts. Thus, it can drive autoimmune diseases. Intracellular antibodies expressed inside the ER block transitory protein functions by inhibiting the translocation of the protein from the ER to its subcellular destination. Here, we describe the construction and characterization of an anti-TLR9 ER intrabody (αT9ib). The respective single-chain Fv comprises the variable domains of the heavy and light chain of a monoclonal antibody (mAb; 5G5) towards human and murine TLR9. Co-expression of αT9ib and mouse TLR9 in HEK293 cells resulted in co-localization of both molecules with the ER marker calnexin. Co-immunoprecipitation of mouse TLR9 with αT9ib indicated that αT9ib interacts with its cognate antigen. The expression of αT9ib inhibited NF-κB-driven reporter gene activation upon CpG DNA challenge but not the activation of TLR3 or TLR4. Consequently, TLR9-driven TNFα production was inhibited in RAW264.7 macrophages upon transfection with the αT9ib expression plasmid. The αT9ib-encoding open reading frame was integrated into an adenoviral cosmid vector to produce the recombinant adenovirus (AdV)-αT9ib. Transduction with AdVαT9ib specifically inhibited TLR9-driven cellular TNFα release. These data strongly indicate that αT9ib is a very promising experimental tool to block TLR9 signaling.


Assuntos
Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo/imunologia , Anticorpos de Cadeia Única/imunologia , Receptor Toll-Like 9/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos/imunologia , Sequência de Bases , Calnexina/imunologia , Calnexina/metabolismo , Linhagem Celular , Clonagem Molecular , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Citometria de Fluxo , Células HEK293 , Humanos , Immunoblotting , Luciferases/genética , Luciferases/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , NF-kappa B/metabolismo , Ligação Proteica/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Antibodies (Basel) ; 11(3)2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35892709

RESUMO

Tumor cells are characterized by overexpressed tumor-associated antigens or mutated neoantigens, which are expressed on the cell surface or intracellularly. One strategy of cancer immunotherapy is to target cell-surface-expressed tumor-associated antigens (TAAs) with therapeutic antibodies. For targeting TAAs or neoantigens, adoptive T-cell therapies with activated autologous T cells from cancer patients transduced with novel recombinant TCRs or chimeric antigen receptors have been successfully applied. Many TAAs and most neoantigens are expressed in the cytoplasm or nucleus of tumor cells. As alternative to adoptive T-cell therapy, the mRNA of intracellular tumor antigens can be depleted by RNAi, the corresponding genes or proteins deleted by CRISPR-Cas or inactivated by kinase inhibitors or by intrabodies, respectively. Intrabodies are suitable to knockdown TAAs and neoantigens without off-target effects. RNA sequencing and proteome analysis of single tumor cells combined with computational methods is bringing forward the identification of new neoantigens for the selection of anti-cancer intrabodies, which can be easily performed using phage display antibody repertoires. For specifically delivering intrabodies into tumor cells, the usage of new capsid-modified adeno-associated viruses and lipid nanoparticles coupled with specific ligands to cell surface receptors can be used and might bring cancer intrabodies into the clinic.

7.
BMC Biotechnol ; 10: 31, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20388199

RESUMO

BACKGROUND: Toll-like receptor (TLR) 2 is a component of the innate immune system and senses specific pathogen associated molecular patterns (PAMPs) of both microbial and viral origin. Cell activation via TLR2 and other pattern recognition receptors (PRRs) contributes to sepsis pathology and chronic inflammation both relying on overamplification of an immune response. Intracellular antibodies expressed and retained inside the endoplasmatic reticulum (ER-intrabodies) are applied to block translocation of secreted and cell surface molecules from the ER to the cell surface resulting in functional inhibition of the target protein. Here we describe generation and application of a functional anti-TLR2 ER intrabody (alphaT2ib) which was generated from an antagonistic monoclonal antibody (mAb) towards human and murine TLR2 (T2.5) to inhibit the function of TLR2. alphaT2ib is a scFv fragment comprising the variable domain of the heavy chain and the variable domain of the light chain of mAb T2.5 linked together by a synthetic (Gly4Ser)3 amino acid sequence. RESULTS: Coexpression of alphaT2ib and mouse TLR2 in HEK293 cells led to efficient retention and accumulation of TLR2 inside the ER compartment. Co-immunoprecipitation of human TLR2 with alphaT2ib indicated interaction of alphaT2ib with its cognate antigen within cells. alphaT2ib inhibited NF-kappaB driven reporter gene activation via TLR2 but not through TLR3, TLR4, or TLR9 if coexpressed in HEK293 cells. Co-transfection of human TLR2 with increasing amounts of the expression plasmid encoding alphaT2ib into HEK293 cells demonstrated high efficiency of the TLR2-alphaT2ib interaction. The alphaT2ib open reading frame was integrated into an adenoviral cosmid vector for production of recombinant adenovirus (AdV)-alphaT2ib. Transduction with AdValphaT2ib specifically inhibited TLR2 surface expression of murine RAW264.7 and primary macrophages derived from bone marrow (BMM). Furthermore, TLR2 activation dependent TNFalpha mRNA accumulation, as well as TNFalpha translation and release by macrophages were largely abrogated upon transduction of alphaT2ib. alphaT2ib was expressed in BMM and splenocytes over 6 days upon systemic infection with AdValphaT2ib. Systemic transduction applying AdValphaT2ib rendered immune cells largely non-responsive to tripalmitoyl-peptide challenge. Our results show persistent paralysis of TLR2 activity and thus inhibition of immune activation. CONCLUSION: The generated anti-TLR2 scFv intrabody inhibits specifically and very efficiently TLR2 ligand-driven cell activation in vitro and ex vivo. This indicates a therapeutic potential of alphaT2ib in microbial or viral infections.


Assuntos
Anticorpos Monoclonais/biossíntese , Macrófagos/metabolismo , Anticorpos de Cadeia Única/biossíntese , Receptor 2 Toll-Like/metabolismo , Adenoviridae , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Sequência de Bases , Linhagem Celular , Retículo Endoplasmático/metabolismo , Vetores Genéticos , Humanos , Interleucina-6/análise , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transdução de Sinais , Anticorpos de Cadeia Única/imunologia , Receptor 2 Toll-Like/imunologia , Transfecção , Fator de Necrose Tumoral alfa/análise
8.
PLoS One ; 14(4): e0215062, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30990863

RESUMO

Interferon α (IFNα) counteracts viral infections by activating various IFNα-stimulated genes (ISGs). These genes encode proteins that block viral transport into the host cell and inhibit viral replication, gene transcription and translation. Due to the existence of 14 different, highly homologous isoforms of mouse IFNα, an IFNα knockout mouse has not yet been established by genetic knockout strategies. An scFv intrabody for holding back IFNα isoforms in the endoplasmic reticulum (ER) and thus counteracting IFNα secretion is reported. The intrabody was constructed from the variable domains of the anti-mouse IFNα rat monoclonal antibody 4EA1 recognizing the 5 isoforms IFNα1, IFNα2, IFNα4, IFNα5, IFNα6. A soluble form of the intrabody had a KD of 39 nM to IFNα4. It could be demonstrated that the anti-IFNα intrabody inhibits clearly recombinant IFNα4 secretion by HEK293T cells. In addition, the secretion of IFNα4 was effectively inhibited in stably transfected intrabody expressing RAW 264.7 macrophages and dendritic D1 cells. Colocalization of the intrabody with IFNα4 and the ER marker calnexin in HEK293T cells indicated complex formation of intrabody and IFNα4 inside the ER. Intracellular binding of intrabody and antigen was confirmed by co-immunoprecipitation. Complexes of endogenous IFNα and intrabody could be visualized in the ER of Poly (I:C) stimulated RAW 264.7 macrophages and D1 dendritic cells. Infection of macrophages and dendritic cells with the vesicular stomatitis virus VSV-AV2 is attenuated by IFNα and IFNß. The intrabody increased virus proliferation in RAW 264.7 macrophages and D1 dendritic cells under IFNß-neutralizing conditions. To analyze if all IFNα isoforms are recognized by the intrabody was not in the focus of this study. Provided that binding of the intrabody to all isoforms was confirmed, the establishment of transgenic intrabody mice would be promising for studying the function of IFNα during viral infection and autoimmune diseases.


Assuntos
Células Dendríticas/imunologia , Retículo Endoplasmático/imunologia , Interferon-alfa/antagonistas & inibidores , Macrófagos/imunologia , Anticorpos de Cadeia Única/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Interferon-alfa/efeitos dos fármacos , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Células RAW 264.7
9.
Protein Sci ; 26(5): 925-945, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28271570

RESUMO

Single domain antibodies (sdAbs) from camels or sharks comprise only the variable heavy chain domain. Human sdAbs comprise the variable domain of the heavy chain (VH) or light chain (VL) and can be selected from human antibodies. SdAbs are stable, nonaggregating molecules in vitro and in vivo compared to complete antibodies and scFv fragments. They are excellent novel inhibitors of cytosolic/nuclear proteins because they are correctly folded inside the cytosol in contrast to scFv fragments. SdAbs are unique because of their excellent specificity and possibility to target posttranslational modifications such as phosphorylation sites, conformers or interaction regions of proteins that cannot be targeted with genetic knockout techniques and are impossible to knockdown with RNAi. The number of inhibiting cytosolic/nuclear sdAbs is increasing and usage of synthetic single pot single domain antibody libraries will boost the generation of these fascinating molecules without the need of immunization. The most frequently selected antigenic epitopes belong to viral and oncogenic proteins, followed by toxins, proteins of the nervous system as well as plant- and drosophila proteins. It is now possible to select functional sdAbs against virtually every cytosolic/nuclear protein and desired epitope. The development of new endosomal escape protein domains and cell-penetrating peptides for efficient transfection broaden the application of inhibiting sdAbs. Last but not least, the generation of relatively new cell-specific nanoparticles such as polymersomes and polyplexes carrying cytosolic/nuclear sdAb-DNA or -protein will pave the way to apply cytosolic/nuclear sdAbs for inhibition of viral infection and cancer in the clinic.


Assuntos
Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Anticorpos de Cadeia Única/química , Proteínas Virais/antagonistas & inibidores , Viroses/metabolismo , Animais , Citosol/metabolismo , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Engenharia de Proteínas/métodos , Anticorpos de Cadeia Única/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Viroses/genética
10.
J Immunol Methods ; 300(1-2): 146-59, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15946674

RESUMO

The human vascular endothelial growth factor receptor-2 (VEGFR-2/KDR) and its ligand vascular endothelial growth factor (VEGF) play an essential role in tumor angiogenesis and in haematological malignancies. To inhibit VEGF induced signalling, intrabodies derived from two scFv fragments recognizing the VEGF receptor were generated. When these intrabodies were expressed in endothelial cells, they blocked the transport of KDR to the cell surface. We developed a cell culture model using porcine aortic endothelial cells overexpressing KDR for testing the efficiency of anti-KDR intrabodies. The two intrabodies were targeted to the ER and colocalized with the KDR receptor in an intracellular compartment. No degradation of the receptor was observed. An immature incomplete glycosylated protein of 195 kDa was detected, suggesting that the intrabodies affect the maturation of the receptor. Despite the presence of significant amounts of receptor protein, the inactivation by one of the two intrabodies was highly effective, resulting in complete functional inhibition of KDR and inhibition of in vitro angiogenesis. The new intrabody appears to be a powerful tool with which to inhibit KDR function.


Assuntos
Fragmentos de Imunoglobulinas/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Expressão Gênica , Glicosilação , Humanos , Fragmentos de Imunoglobulinas/genética , Técnicas In Vitro , Peso Molecular , Neovascularização Fisiológica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Suínos , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
MAbs ; 7(6): 1010-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26252565

RESUMO

Intracellular antibodies (intrabodies) are recombinant antibody fragments that bind to target proteins expressed inside of the same living cell producing the antibodies. The molecules are commonly used to study the function of the target proteins (i.e., their antigens). The intrabody technology is an attractive alternative to the generation of gene-targeted knockout animals, and complements knockdown techniques such as RNAi, miRNA and small molecule inhibitors, by-passing various limitations and disadvantages of these methods. The advantages of intrabodies include very high specificity for the target, the possibility to knock down several protein isoforms by one intrabody and targeting of specific splice variants or even post-translational modifications. Different types of intrabodies must be designed to target proteins at different locations, typically either in the cytoplasm, in the nucleus or in the endoplasmic reticulum (ER). Most straightforward is the use of intrabodies retained in the ER (ER intrabodies) to knock down the function of proteins passing the ER, which disturbs the function of members of the membrane or plasma proteomes. More effort is needed to functionally knock down cytoplasmic or nuclear proteins because in this case antibodies need to provide an inhibitory effect and must be able to fold in the reducing milieu of the cytoplasm. In this review, we present a broad overview of intrabody technology, as well as applications both of ER and cytoplasmic intrabodies, which have yielded valuable insights in the biology of many targets relevant for drug development, including α-synuclein, TAU, BCR-ABL, ErbB-2, EGFR, HIV gp120, CCR5, IL-2, IL-6, ß-amyloid protein and p75NTR. Strategies for the generation of intrabodies and various designs of their applications are also reviewed.


Assuntos
Anticorpos/imunologia , Técnicas de Silenciamento de Genes/métodos , Espaço Intracelular/imunologia , Proteoma/imunologia , Animais , Anticorpos/genética , Anticorpos/metabolismo , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Citoplasma/imunologia , Citoplasma/metabolismo , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Humanos , Espaço Intracelular/metabolismo , Modelos Imunológicos
12.
J Cell Mol Med ; 11(1): 54-70, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17367501

RESUMO

Intracellular antibodies (intrabodies) constitute a potent tool to neutralize the function of target proteins inside specific cell compartments (cytosol, nucleus, mitochondria and ER). The intrabody technology is an attractive alternative to the generation of gene-targeted knockout animals and complements or replaces knockdown techniques such as antisense-RNA, RNAi and RNA aptamers. This article focuses on intrabodies targeted to the ER. Intracellular anti-bodies expressed and retained inside the ER (ER intrabodies) are shown to be highly efficient in blocking the translocation of secreted and cell surface molecules from the ER to the cell surface. The advantage of ER intrabodies over cytoplasmic intrabodies is that they are correctly folded and easier to select. A particular advantage of the intrabody technology over existing ones is the possibility of inhibiting selectively post-translational modifications of proteins. The main applications of ER intrabodies so far have been (i) inactivation of oncogenic receptors and (ii) functional inhibition of virus envelope proteins and virus-receptor molecules on the surface of host cells. In cancer research, the number of in vivo mouse models for evaluation of the therapeutic potential of intrabodies is increasing. In the future, endosomal localized receptors involved in bacterial and viral infections, intracellular oncogenic receptors and enzymes involved in glycosylation of tumour antigens might be new targets for ER intrabodies.


Assuntos
Anticorpos/imunologia , Anticorpos/metabolismo , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Proteínas/antagonistas & inibidores , Animais , Anticorpos/genética , Membrana Celular/metabolismo , Humanos , Modelos Imunológicos , Processamento de Proteína Pós-Traducional , Proteínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA