Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Immunity ; 42(5): 850-63, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25979419

ABSTRACT

The molecular mechanisms regulating antigen translocation into the cytosol for cross-presentation are under controversial debate, mainly because direct data is lacking. Here, we have provided direct evidence that the activity of the endoplasmic reticulum (ER) translocon protein Sec61 is essential for endosome-to-cytosol translocation. We generated a Sec61-specific intrabody, a crucial tool that trapped Sec61 in the ER and prevented its recruitment into endosomes without influencing Sec61 activity and antigen presentation in the ER. Expression of this ER intrabody inhibited antigen translocation and cross-presentation, demonstrating that endosomal Sec61 indeed mediates antigen transport across endosomal membranes. Moreover, we showed that the recruitment of Sec61 toward endosomes, and hence antigen translocation and cross-presentation, is dependent on dendritic cell activation by Toll-like receptor (TLR) ligands. These data shed light on a long-lasting question regarding antigen cross-presentation and point out a role of the ER-associated degradation machinery in compartments distinct from the ER.


Subject(s)
Antigens/metabolism , CD8-Positive T-Lymphocytes , Cross-Priming/immunology , Cytosol/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , Models, Biological , Animals , Antigens/immunology , Cell Line , Cytosol/immunology , Membrane Proteins/chemistry , Mice , Protein Transport , SEC Translocation Channels
2.
Biol Chem ; 400(3): 323-332, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30240352

ABSTRACT

Radiolabeling of nanobodies with radiometals by chelation has the advantage of being simple, fast and easy to implement in clinical routine. In this study, we validated 68Ga/111In-labeled anti-VCAM-1 nanobodies as potential radiometal-based tracers for molecular imaging of atherosclerosis. Both showed specific targeting of atherosclerotic lesions in ApoE-/- mice. Nevertheless, uptake in lesions and constitutively VCAM-1 expressing organs was lower than previously reported for the 99mTc-labeled analog. We further investigated the impact of different radiolabeling strategies on the in vivo biodistribution of nanobody-based tracers. Comparison of the pharmacokinetics between 68Ga-, 18F-, 111In- and 99mTc-labeled anti-VCAM-1 nanobodies showed highest specific uptake for 99mTc-nanobody at all time-points, followed by the 68Ga-, 111In- and 18F-labeled tracer. No correlation was found with the estimated number of radioisotopes per nanobody, and mimicking specific activity of other radiolabeling methods did not result in an analogous biodistribution. We also demonstrated specificity of the tracer using mice with a VCAM-1 knocked-down phenotype, while showing for the first time the in vivo visualization of a protein knock-down using intrabodies. Conclusively, the chosen radiochemistry does have an important impact on the biodistribution of nanobodies, in particular on the specific targeting, but differences are not purely due to the tracer's specific activity.


Subject(s)
Atherosclerosis/diagnostic imaging , Molecular Imaging , Single-Domain Antibodies/chemistry , Vascular Cell Adhesion Molecule-1/immunology , Animals , Gallium Radioisotopes , Indium Radioisotopes , Isotope Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism
3.
Adv Exp Med Biol ; 917: 77-93, 2016.
Article in English | MEDLINE | ID: mdl-27236553

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Cell Nucleus/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques/methods , Proteome/immunology , Animals , Antibodies/genetics , Antibodies/metabolism , Cell Nucleus/immunology , Humans , Mice
4.
BioDrugs ; 37(2): 181-203, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36729328

ABSTRACT

Numerous toxins translocate to the cytosol in order to fulfil their function. This demonstrates the existence of routes for proteins from the extracellular space to the cytosol. Understanding these routes is relevant to multiple aspects related to therapeutic applications. These include the development of anti-toxin treatments, the potential use of toxins as shuttles for delivering macromolecular cargo to the cytosol or the use of drugs based on toxins. Compared with other strategies for delivery, such as chemicals as carriers for macromolecular delivery or physical methods like electroporation, toxin routes present paths into the cell that potentially cause less damage and can be specifically targeted. The efficiency of delivery via toxin routes is limited. However, low-delivery efficiencies can be entirely sufficient, if delivered cargoes possess an amplification effect or if very few molecules are sufficient for inducing the desired effects. This is known for example from RNA-based vaccines that have been developed during the coronavirus disease 2019 pandemic as well as for other approved RNA-based drugs, which elicited the desired effect despite their typically low delivery efficiencies. The different mechanisms by which toxins enter cells may have implications for their technological utility. We review the mechanistic principles of the translocation pathway of toxins from the extracellular space to the cytosol, the delivery efficiencies, and therapeutic strategies or applications that exploit toxin routes for intracellular delivery.


Subject(s)
Biological Products , COVID-19 , Humans , Biological Products/pharmacology , Biological Products/therapeutic use , Cytosol/metabolism , Pharmaceutical Preparations/metabolism
5.
BioDrugs ; 35(6): 643-671, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34705260

ABSTRACT

Delivering macromolecules into the cytosol or nucleus is possible in vitro for DNA, RNA and proteins, but translation for clinical use has been limited. Therapeutic delivery of macromolecules into cells requires overcoming substantially higher barriers compared to the use of small molecule drugs or proteins in the extracellular space. Breakthroughs like DNA delivery for approved gene therapies and RNA delivery for silencing of genes (patisiran, ONPATTRO®, Alnylam Pharmaceuticals, Cambridge, MA, USA) or for vaccination such as the RNA-based coronavirus disease 2019 (COVID-19) vaccines demonstrated the feasibility of using macromolecules inside cells for therapy. Chemical carriers are part of the reason why these novel RNA-based therapeutics possess sufficient efficacy for their clinical application. A clear advantage of synthetic chemicals as carriers for macromolecule delivery is their favourable properties with respect to production and storage compared to more bioinspired vehicles like viral vectors or more complex drugs like cellular therapies. If biologicals can be applied to intracellular targets, the druggable space is substantially broadened by circumventing the limited utility of small molecules for blocking protein-protein interactions and the limitation of protein-based drugs to the extracellular space. An in depth understanding of the macromolecular cargo types, carrier types and the cell biology of delivery is crucial for optimal application and further development of biologicals inside cells. Basic mechanistic principles of the molecular and cell biological aspects of cytosolic/nuclear delivery of macromolecules, with particular consideration of protein delivery, are reviewed here. The efficiency of macromolecule delivery and applications in research and therapy are highlighted.


Subject(s)
COVID-19 , Pharmaceutical Preparations , Genetic Therapy , Humans , SARS-CoV-2
6.
BioDrugs ; 34(4): 435-462, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32301049

ABSTRACT

To interfere with cell function, many scientists rely on methods that target DNA or RNA due to the ease with which they can be applied. Proteins are usually the final executors of function but are targeted only indirectly by these methods. Recent advances in targeted degradation of proteins based on proteolysis-targeting chimaeras (PROTACs), ubiquibodies, deGradFP (degrade Green Fluorescent Protein) and other approaches have demonstrated the potential of interfering directly at the protein level for research and therapy. Proteins can be targeted directly and very specifically by antibodies, but using antibodies inside cells has so far been considered to be challenging. However, it is possible to deliver antibodies or other proteins into the cytosol using standard laboratory equipment. Physical methods such as electroporation have been demonstrated to be efficient and validated thoroughly over time. The expression of intracellular antibodies (intrabodies) inside cells is another way to interfere with intracellular targets at the protein level. Methodological strategies to target the inside of cells with antibodies, including delivered antibodies and expressed antibodies, as well as applications in the research areas of neurobiology, viral infections and oncology, are reviewed here. Antibodies have already been used to interfere with a wide range of intracellular targets. Disease-related targets included proteins associated with neurodegenerative diseases such as Parkinson's disease (α-synuclein), Alzheimer's disease (amyloid-ß) or Huntington's disease (mutant huntingtin [mHtt]). The applications of intrabodies in the context of viral infections include targeting proteins associated with HIV (e.g. HIV1-TAT, Rev, Vif, gp41, gp120, gp160) and different oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV) and Epstein-Barr virus, and they have been used to interfere with various targets related to different processes in cancer, including oncogenic pathways, proliferation, cell cycle, apoptosis, metastasis, angiogenesis or neo-antigens (e.g. p53, human epidermal growth factor receptor-2 [HER2], signal transducer and activator of transcription 3 [STAT3], RAS-related RHO-GTPase B (RHOB), cortactin, vascular endothelial growth factor receptor 2 [VEGFR2], Ras, Bcr-Abl). Interfering at the protein level allows questions to be addressed that may remain unanswered using alternative methods. This review addresses why direct targeting of proteins allows unique insights, what is currently feasible in vitro, and how this relates to potential therapeutic applications.


Subject(s)
Antibodies/chemistry , Epstein-Barr Virus Infections , Herpesvirus 4, Human/chemistry , Neurobiology , Antibodies/administration & dosage , Humans , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
7.
ACS Chem Biol ; 14(12): 2641-2651, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31633910

ABSTRACT

Ineffective cellular delivery is a common problem in numerous biological applications. Developing delivery reagents that work robustly in a variety of experimental settings remains a challenge. Herein, we report how peptides derived from the prototypical cell penetrating peptide TAT can be used in combination with a small molecule, UNC7938, to deliver macromolecules into the cytosol of cells by a simple co-incubation protocol. We establish successful delivery of peptides, DNA plasmids, and a single-chain variable fragment antibody. We also demonstrate that delivery works in hard-to-transfect mammalian cells and under conditions typically inhibitory to cell-penetrating peptides. Mechanistically, UNC7938 destabilizes the membrane of endosomes. This, in turn, enhances the endosome-leakage activity of cell-penetrating peptides and facilitates the endosomal escape of macromolecules initially internalized by mammalian cells via endocytosis. This combined selective membrane-destabilization represents a new chemical space for delivery tools and provides a novel solution to the problem of endosomal entrapment that often limits the effectiveness of reagent-based delivery approaches.


Subject(s)
Cell-Penetrating Peptides/metabolism , Cytosol/metabolism , Endosomes/metabolism , Macromolecular Substances/metabolism , Cytosol/drug effects , Endosomes/drug effects , Humans , Pyrazines/pharmacology , Pyridines/pharmacology
8.
Methods Mol Biol ; 1513: 201-208, 2017.
Article in English | MEDLINE | ID: mdl-27807839

ABSTRACT

Delivery of proteins to the cytosol of living cells is a promising research tool. Delivery of antibodies in particular bears exciting applications such as in vivo tracking of proteins at endogenous expression levels or interference with cellular processes. In spite of the large number of methods published for protein delivery, successful applications so far are rare. A possible explanation for this is a vast overestimation of the delivery efficiency due to the use of inappropriate detection methods and/or unsuitable positive controls for cytosolic delivery. Therefore, we provide strategies for unequivocally detecting cytoplasmic protein delivery and quantifying protein transformation efficiency. Finally, we present a protocol for efficient protein delivery to the cytosol validated using these methods.


Subject(s)
Cell-Penetrating Peptides/metabolism , Cytosol/metabolism , Drug Carriers , Electroporation/methods , Single-Chain Antibodies/metabolism , Animals , Cell Line , Cell-Penetrating Peptides/chemistry , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , HeLa Cells , Humans , Integrases/genetics , Integrases/metabolism , Myosins/immunology , Myosins/metabolism , Plasmids/chemistry , Plasmids/metabolism , Tissue Fixation/methods , Tubulin/immunology , Tubulin/metabolism
9.
Comput Struct Biotechnol J ; 14: 304-8, 2016.
Article in English | MEDLINE | ID: mdl-27570612

ABSTRACT

Challenges posed by complex diseases such as cancer, chronic viral infections, neurodegenerative disorders and many others have forced researchers to think beyond classic small molecule drugs, exploring new therapeutic strategies such as therapy with RNAi, CRISPR/Cas9 or antibody therapies as single or as combination therapies with existing drugs. While classic antibody therapies based on parenteral application can only reach extracellular targets, intracellular application of antibodies could provide specific advantages but is so far little recognized in translational research. Intrabodies allow high specificity and targeting of splice variants or post translational modifications. At the same time off target effects can be minimized by thorough biochemical characterization. Knockdown of cellular proteins by intrabodies has been reported for a significant number of disease-relevant targets, including ErbB-2, EGFR, VEGFR-2, Metalloproteinase MMP2 and MMP9, ß-amyloid protein, α-synuclein, HIV gp120, HCV core and many others. This review outlines the recent advances in ER intrabody technology and their potential use in therapy.

10.
MAbs ; 7(6): 1010-35, 2015.
Article in English | MEDLINE | ID: mdl-26252565

ABSTRACT

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.


Subject(s)
Antibodies/immunology , Gene Knockdown Techniques/methods , Intracellular Space/immunology , Proteome/immunology , Animals , Antibodies/genetics , Antibodies/metabolism , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cytoplasm/immunology , Cytoplasm/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Humans , Intracellular Space/metabolism , Models, Immunological
11.
MAbs ; 6(4): 943-56, 2014.
Article in English | MEDLINE | ID: mdl-24848507

ABSTRACT

The use of antibodies to target their antigens in living cells is a powerful analytical tool for cell biology research. Not only can molecules be localized and visualized in living cells, but interference with cellular processes by antibodies may allow functional analysis down to the level of individual post-translational modifications and splice variants, which is not possible with genetic or RNA-based methods. To utilize the vast resource of available antibodies, an efficient system to deliver them into the cytosol from the outside is needed. Numerous strategies have been proposed, but the most robust and widely applicable procedure still remains to be identified, since a quantitative ranking of the efficiencies has not yet been done. To achieve this, we developed a novel efficiency evaluation method for antibody delivery based on a fusion protein consisting of a human IgG 1 Fc and the recombination enzyme Cre (Fc-Cre). Applied to suitable GFP reporter cells, it allows the important distinction between proteins trapped in endosomes and those delivered to the cytosol. Further, it ensures viability of positive cells and is unsusceptible to fixation artifacts and misinterpretation of cellular localization in microscopy and flow cytometry. Very low cytoplasmic delivery efficiencies were found for various profection reagents and membrane penetrating peptides, leaving electroporation as the only practically useful delivery method for antibodies. This was further verified by the successful application of this method to bind antibodies to cytosolic components in living cells.


Subject(s)
Cytosol/metabolism , Drug Delivery Systems/methods , Immunoglobulin Constant Regions , Immunoglobulin G , Integrases , Recombinant Fusion Proteins , Endosomes/metabolism , HeLa Cells , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/pharmacology , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Integrases/genetics , Integrases/pharmacokinetics , Integrases/pharmacology , Protein Transport
12.
MAbs ; 6(6): 1394-401, 2014.
Article in English | MEDLINE | ID: mdl-25484057

ABSTRACT

Functional knockdowns mediated by endoplasmatic reticulum-retained antibodies (ER intrabodies) are a promising tool for research because they allow functional interference on the protein level. We demonstrate for the first time that ER intrabodies can induce a knock-down phenotype in mice. Surface VCAM1 was suppressed in bone marrow of heterozygous and homozygous ER intrabody mice (iER-VCAM1 mice). iER-VCAM1 mice did not have a lethal phenotype, in contrast to the constitutive knockout of VCAM1, but adult mice exhibited physiological effects in the form of aberrant distribution of immature B-cells in blood and bone marrow. The capability to regulate knock-down strength may spark a new approach for the functional study of membrane and plasma proteins, which may especially be valuable for generating mouse models that more closely resemble disease states than classic knockouts do.


Subject(s)
Down-Regulation/immunology , Endoplasmic Reticulum/immunology , Single-Chain Antibodies/immunology , Vascular Cell Adhesion Molecule-1/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line , Down-Regulation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Female , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
13.
PLoS One ; 7(1): e30684, 2012.
Article in English | MEDLINE | ID: mdl-22292018

ABSTRACT

BACKGROUND: Although p75 neurotrophin receptor (p75NTR) is the first neurotrophin receptor isolated, its diverse physiological functions and signaling have remained elusive for many years. Loss-of-function phenotypic analyses for p75NTR were mainly focused at the genetic level; however these approaches were impacted by off-target effect, insufficient stability, unspecific stress response or alternative active splicing products. In this study, p75NTR surface expression was suppressed for the first time at the protein level by endoplasmic reticulum (ER) retained intrabodies. RESULTS: Three monoclonal recombinant antibody fragments (scFv) with affinities in the low nanomolar range to murine p75NTR were isolated by antibody phage display. To suppress p75NTR cell surface expression, the encoding genes of these scFvs extended by the ER retention peptide KDEL were transiently transfected into the neuron-like rat pheochromocytoma cell line PC12 and the mouse neuroblastoma x mouse spinal cord hybrid cell line NSC19. The ER retained intrabody construct, SH325-G7-KDEL, mediated a downregulation of p75NTR cell surface expression as shown by flow cytometry. This effect was maintained over a period of at least eight days without activating an unfolded protein response (UPR). Moreover, the ER retention of p75NTR resulted in downregulation of mRNA levels of the anti-apoptotic protein Bcl-xL as well as in strong inhibition of NGF-induced neurite outgrowth in PC12 cells. CONCLUSION: The ER retained intrabody SH325-G7-KDEL not only induces phenotypic knockdown of this p75NTR but also p75NTR-associated cellular responses in PC12 cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neurites/drug effects , Receptor, Nerve Growth Factor/antagonists & inhibitors , bcl-X Protein/genetics , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice , Models, Biological , Neurites/metabolism , Neurites/physiology , PC12 Cells , Protein Transport/drug effects , Protein Transport/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/immunology , Receptor, Nerve Growth Factor/metabolism , Recombinant Fusion Proteins/pharmacology , bcl-X Protein/metabolism
14.
MAbs ; 3(1): 3-16, 2011.
Article in English | MEDLINE | ID: mdl-21099369

ABSTRACT

A growing number of research consortia are now focused on generating antibodies and recombinant antibody fragments that target the human proteome. A particularly valuable application for these binding molecules would be their use inside a living cell, e.g., for imaging or functional intervention. Animal-derived antibodies must be brought into the cell through the membrane, whereas the availability of the antibody genes from phage display systems allows intracellular expression. Here, the various technologies to target intracellular proteins with antibodies are reviewed.


Subject(s)
Antibodies/metabolism , Cytoplasm/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/genetics , Endocytosis , Humans , Molecular Sequence Data , Protein Folding , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL