Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biol Chem ; 299(9): 104927, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37330175

RESUMO

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Assuntos
Anticorpos Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Receptores de Superfície Celular , Anticorpos de Domínio Único , Humanos , Antibacterianos/farmacologia , Heme/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/uso terapêutico , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Anticorpos de Domínio Único/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Antígenos de Bactérias/imunologia , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Camelídeos Americanos , Animais , Ligação Proteica/efeitos dos fármacos , Modelos Moleculares , Simulação de Dinâmica Molecular
2.
Biochem Soc Trans ; 49(5): 2021-2035, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34623375

RESUMO

The RAS superfamily of small GTPases regulates major physiological cellular processes. Mutation or deregulation of these small GTPases, their regulators and/or their effectors are associated with many diseases including cancer. Hence, targeting these classes of proteins is an important therapeutic strategy in cancer. This has been recently achieved with the approval of the first KRASG12C covalent inhibitors for the clinic. However, many other mutants and small GTPases are still considered as 'undruggable' with small molecule inhibitors because of a lack of well-defined pocket(s) at their surface. Therefore, alternative therapeutic strategies have been developed to target these proteins. In this review, we discuss the use of intracellular antibodies and derivatives - reagents that bind their antigen inside the cells - for the discovery of novel inhibitory mechanisms, targetable features and therapeutic strategies to inhibit small GTPases and their downstream pathways. These reagents are also versatile tools used to better understand the biological mechanisms regulated by small GTPases and to accelerate the drug discovery process.


Assuntos
Anticorpos/metabolismo , Proteínas de Repetição de Anquirina Projetadas/metabolismo , Descoberta de Drogas/métodos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neoplasias/enzimologia , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Proteínas de Repetição de Anquirina Projetadas/farmacologia , Humanos , Terapia de Alvo Molecular/métodos , Proteínas Monoméricas de Ligação ao GTP/antagonistas & inibidores , Proteínas Monoméricas de Ligação ao GTP/imunologia , Neoplasias/tratamento farmacológico , Ligação Proteica , Proteólise/efeitos dos fármacos , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/farmacologia
3.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33903242

RESUMO

Infection with obligatory intracellular bacteria is difficult to treat, as intracellular targets and delivery methods of therapeutics are not well known. Ehrlichia translocated factor-1 (Etf-1), a type IV secretion system (T4SS) effector, is a primary virulence factor for an obligatory intracellular bacterium, Ehrlichia chaffeensis In this study, we developed Etf-1-specific nanobodies (Nbs) by immunizing a llama to determine if intracellular Nbs block Etf-1 functions and Ehrlichia infection. Of 24 distinct anti-Etf-1 Nbs, NbD7 blocked mitochondrial localization of Etf-1-GFP in cotransfected cells. NbD7 and control Nb (NbD3) bound to different regions of Etf-1. Size-exclusion chromatography showed that the NbD7 and Etf-1 complex was more stable than the NbD3 and Etf-1 complex. Intracellular expression of NbD7 inhibited three activities of Etf-1 and E. chaffeensis: up-regulation of mitochondrial manganese superoxide dismutase, reduction of intracellular reactive oxygen species, and inhibition of cellular apoptosis. Consequently, intracellular NbD7 inhibited Ehrlichia infection, whereas NbD3 did not. To safely and effectively deliver Nbs into the host cell cytoplasm, NbD7 was conjugated to cyclized cell-permeable peptide 12 (CPP12-NbD7). CPP12-NbD7 effectively entered mammalian cells and abrogated the blockade of cellular apoptosis caused by E. chaffeensis and inhibited infection by E. chaffeensis in cell culture and in a severe combined-immunodeficiency mouse model. Our results demonstrate the development of an Nb that interferes with T4SS effector functions and intracellular pathogen infection, along with an intracellular delivery method for this Nb. This strategy should overcome current barriers to advance mechanistic research and develop therapies complementary or alternative to the current broad-spectrum antibiotic.


Assuntos
Ehrlichia chaffeensis/efeitos dos fármacos , Ehrlichiose/tratamento farmacológico , Anticorpos de Domínio Único/farmacologia , Sistemas de Secreção Tipo IV/genética , Animais , Apoptose/genética , Subpopulações de Linfócitos B/imunologia , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/imunologia , Ehrlichia chaffeensis/patogenicidade , Ehrlichiose/genética , Ehrlichiose/imunologia , Ehrlichiose/patologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Anticorpos de Domínio Único/imunologia , Sistemas de Secreção Tipo IV/antagonistas & inibidores , Sistemas de Secreção Tipo IV/imunologia , Fatores de Virulência
4.
Mol Biol Rep ; 47(12): 9939-9949, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33185828

RESUMO

The outbreak of a novel coronavirus namely SARS-CoV-2, which first emerged from Wuhan, China, has wreaked havoc not only in China but the whole world that now has been engulfed in its wrath. In a short lapse of time, this virus was successful in spreading at a blistering pace throughout the globe, hence raising the flag of pandemic status. The mounting number of deaths with each elapsing day has summoned researchers from all around the world to play their part in driving this SARS-CoV-2 pandemic to an end. As of now, multiple research teams are immersed in either scrutinizing various antiviral drugs for their efficacy or developing different types of vaccines that will be capable of providing long-term immunity against this deadly virus. The mini-review sheds light on the possible approaches that can be undertaken to curb the COVID-19 spread. Possible strategies comprise viral vector-based, nucleic acid-based, protein-based, inactivated and weakened virus vaccines; COVID-19 vaccine being developed by deploying Hyleukin-7 technology; plant-based chimeric protein and subunit vaccines; humanized nano-bodies and human antibodies; intravenous immunoglobulin (IVIG) infusion therapy; inhibitors for ACE-2, Angiotensin 1 receptor (AT1R), complement system, viral proteins, host cell protease and endocytosis; shield immunity; IL-6R, NKG2A and hACE2-SARS-CoV-2-RBD interaction blocking monoclonal antibodies; SARS-CoV RdRp-based drugs, traditional Chinese medicine, repositioned and anti-viral drugs. These vaccines and drugs are currently being screened in the clinical trials as several of them have manifested positive results, hence increasing the probability of becoming one of the potential treatments for this disease.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Vacinas contra COVID-19/farmacologia , COVID-19/prevenção & controle , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Ensaios Clínicos como Assunto , Reposicionamento de Medicamentos , Humanos , Vírus da Bronquite Infecciosa/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , RNA Mensageiro/imunologia , Proteínas Recombinantes/genética , Anticorpos de Domínio Único/farmacologia , Vacinas Atenuadas/farmacologia , Vacinas de Subunidades Antigênicas/farmacologia , Vacinas Sintéticas/farmacologia
5.
J Biomol NMR ; 73(6-7): 375-384, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31073665

RESUMO

The insertase BamA is an essential protein of the bacterial outer membrane. Its 16-stranded transmembrane ß-barrel contains a lateral gate as a key functional element. This gate is formed by the C-terminal half of the last ß-strand. The BamA barrel was previously found to sample different conformations in aqueous solution, as well as different gate-open, gate-closed, and collapsed conformations in X-ray crystallography and cryo-electron microscopy structures. Here, we report the successful identification of conformation-selective nanobodies that stabilize BamA in specific conformations. While the initial candidate generation and selection protocol was based on established alpaca immunization and phage display selection procedures, the final selection of nanobodies was enhanced by a solution NMR-based screening step to shortlist the targets for crystallization. In this way, three crystal structures of BamA-nanobody complexes were efficiently obtained, showing two types of nanobodies that indeed stabilized BamA in two different conformations, i.e., with open and closed lateral gate, respectively. Then, by correlating the structural data with high resolution NMR spectra, we could for the first time assign the BamA conformational solution ensemble to defined structural states. The new nanobodies will be valuable tools towards understanding the client insertion mechanism of BamA and towards developing improved antibiotics.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Modelos Moleculares , Conformação Proteica , Anticorpos de Domínio Único/química , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Anticorpos de Domínio Único/farmacologia , Soluções
6.
Methods Mol Biol ; 1836: 139-158, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30151572

RESUMO

Our understanding of infection biology is based on experiments in which pathogen or host proteins are perturbed by small compound inhibitors, mutation, or depletion. This approach has been remarkably successful, as, for example, demonstrated by the independent identification of the endosomal membrane protein Niemann-Pick C1 as an essential factor for Ebola virus infection in both small compound and insertional mutagenesis screens (Côté, Nature 477:344-348, 2011; Carette et al., Nature 477:340-343, 2011). However, many aspects of host-pathogen interactions are poorly understood because we cannot target all of the involved molecules with small molecules, or because we cannot deplete essential proteins. Single domain antibody fragments expressed in the cytosol or other organelles constitute a versatile alternative to perturb the function of any given protein by masking protein-protein interaction interfaces, by stabilizing distinct conformations, or by directly interfering with enzymatic activities. The variable domains of heavy chain-only antibodies (VHHs) from camelid species can be cloned from blood samples of animals immunized with the desired target molecules. We can thus exploit the ability of the camelid immune system to generate affinity-matured single domain antibody fragments to obtain highly specific tools. Interesting VHH candidates are typically identified based on their affinity toward immobilized antigens using techniques such as phage display.The phenotypical screening approach described here allows the direct identification of VHHs that prevent infection of cells with influenza A virus (IAV) or other pathogens. The VHH repertoire is cloned into a lentiviral vector, which is used to generate pseudo-typed lentivirus particles. Target cells are transduced with the lentivirus, so that every cell inducibly expresses a different VHH. This cell collection is then challenged with a lethal dose of virus. Only the cells which express a VHH that prevents infection by targeting virus proteins or host cell components essential for infection will survive. We can thus identify critical target molecules including vulnerable epitopes and conformations, render target molecules accessible to informative perturbation studies, and stabilize intermediates of virus entry for detailed analysis.


Assuntos
Antirretrovirais/farmacologia , Lentivirus/efeitos dos fármacos , Fenótipo , Anticorpos de Domínio Único/farmacologia , Sequência de Aminoácidos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Biblioteca Gênica , Vetores Genéticos/genética , Humanos , Vírus da Influenza A/genética , Lentivirus/genética , Infecções por Lentivirus/tratamento farmacológico , Infecções por Lentivirus/virologia , Testes de Sensibilidade Microbiana , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/genética
7.
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
8.
Sci Rep ; 6: 32256, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27558409

RESUMO

Candida albicans (C. albicans) is an important human commensal and opportunistic fungal pathogen. Secreted aspartyl proteinases (Saps) are a major virulence trait of C. albicans, and among these proteases Sap2 has the highest expression levels. It is possible that antibodies against Sap2 could provide an antifungal effect. In this study, two phages displaying anti-rSap2 single chain variable fragments (scFvs) were screened from human single fold scFv libraries, and their potential therapeutic roles were evaluated using a murine model infected by C. albicans. The in vivo efficacies were assessed by mortality rates, fungal burden and histological examination. Overall survival rates were significantly increased while the colony counts and infectious foci were significantly decreased after treatment with the scFv-phages relative to the control groups. In order to investigate the immune response provoked by scFv-phages, three kinds of cytokines (Th1, Th2 and Th17 types) were measured and a clear immune response was observed. These findings suggest that anti-rSap2 scFv-phages have potential in the therapy of systemic infection caused by C. albicans.


Assuntos
Anticorpos Antifúngicos/farmacologia , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Candida albicans/imunologia , Candidíase/tratamento farmacológico , Proteínas Fúngicas/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , Animais , Anticorpos Antifúngicos/química , Anticorpos Antifúngicos/genética , Anticorpos Antifúngicos/imunologia , Ácido Aspártico Endopeptidases/imunologia , Bacteriófago M13 , Candidíase/genética , Candidíase/imunologia , Candidíase/patologia , Modelos Animais de Doenças , Feminino , Proteínas Fúngicas/imunologia , Humanos , Camundongos Endogâmicos BALB C , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , ômega-Cloroacetofenona
9.
Eur J Cancer ; 50(4): 713-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22918079

RESUMO

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is over-expressed in pancreatic cancer cells, and it is associated with the progression of pancreatic cancer. We tested a single domain antibody (sdAb) targeting CEACAM6, 2A3, which was isolated previously from a llama immune library, and an Fc conjugated version of this sdAb, to determine how they affect the pancreatic cancer cell line BxPC3. We also compared the effects of the antibodies to gemcitabine. Gemcitabine and 2A3 slowed down cancer cell proliferation. However, only 2A3 retarded cancer cell invasion, angiogenesis within the cancer mass and BxPC3 cell MMP-9 activity, three features important for tumour growth and metastasis. The IC50s for 2A3, 2A3-Fc and gemcitabine were determined as 6.5µM, 8µM and 12nM, respectively. While the 2A3 antibody inhibited MMP-9 activity by 33% compared to non-treated control cells, gemcitabine failed to inhibit MMP-9 activity. Moreover, 2A3 and 2A3-Fc inhibited invasion of BxPC3 by 73% compared to non-treated cells. When conditioned media that were produced using 2A3- or 2A3-Fc-treated BxPC3 cells were used in a capillary formation assay, the capillary length was reduced by 21% and 49%, respectively. Therefore 2A3 is an ideal candidate for treating tumours that over-express CEACAM6.


Assuntos
Antígenos CD/imunologia , Carcinoma Ductal Pancreático/patologia , Moléculas de Adesão Celular/imunologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Neoplasias Pancreáticas/patologia , Anticorpos de Domínio Único/farmacologia , Animais , Camelídeos Americanos , Carcinoma Ductal Pancreático/irrigação sanguínea , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Proteínas Ligadas por GPI/imunologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Invasividade Neoplásica , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas
10.
FEBS J ; 280(15): 3543-50, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23627412

RESUMO

In 1901, the first Nobel Prize in Physiology or Medicine was awarded to Emil von Behring for his ground-breaking discovery of serum therapy: serum from horses vaccinated with toxin-containing culture medium of Corynebacterium diphtheriae contained life-saving 'antitoxins'. The molecular nature of the ADP-ribosylating toxin and the neutralizing antibodies were unraveled only 50 years later. Today, von Behring's antibody therapy is being refined with a new generation of recombinant antibodies and antibody fragments. Nanobodies, which are single-domain antibodies derived from the peculiar heavy-chain antibodies of llamas and other camelids, are emerging as a promising new class of highly specific enzyme inhibitors. In this review, we illustrate the potential of nanobodies as tools to block extracellular and intracellular ADP-ribosyltransferases (ARTs), using the toxin-related membrane-bound mammalian ecto-enzyme ARTC2 and the actin-ADP-ribosylating Salmonella virulence plasmid factor B toxin of Salmonella enterica as examples.


Assuntos
ADP Ribose Transferases/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , ADP Ribose Transferases/metabolismo , Sequência de Aminoácidos , Animais , Infecções Bacterianas/tratamento farmacológico , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/metabolismo , Camelídeos Americanos , Sistemas de Liberação de Medicamentos , Humanos , Dados de Sequência Molecular , Anticorpos de Domínio Único/química , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA