Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Methods Mol Biol ; 2033: 1-14, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31332743

RESUMO

Antibody-drug conjugates (ADCs) have been proven to be a successful therapeutic concept, allowing targeted delivery of highly potent active pharmaceutical ingredients (HPAPIs) selectively to tumor tissue. So far, HPAPIs have been mainly attached to the antibody via a chemical reaction of the payload with lysine or cysteine side chains of the antibody backbone. However, these conventional conjugation technologies result in formation of rather heterogeneous products with undesired properties. To overcome the limitations of heterogeneous ADC mixtures, several site-specific conjugation technologies have been developed over the last years. Originally pioneered by scientist from Genentech with their work on THIOMABs, several engineered cysteine mAb ADCs (ECM-ADCs) are now investigated in clinical trials. Here, we describe in detail how to engineer additional cysteines into antibodies and efficiently use them as highly site-specific conjugation sites for HPAPIs.


Assuntos
Anticorpos Monoclonais Humanizados/genética , Citotoxinas/genética , Imunoconjugados/genética , Engenharia de Proteínas , Animais , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos Imunológicos , Proliferação de Células/efeitos dos fármacos , Cisteína/química , Cisteína/genética , Citotoxinas/química , Citotoxinas/imunologia , Citotoxinas/farmacologia , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/farmacologia , Camundongos , Mutagênese Sítio-Dirigida , Compostos de Sulfidrila/química , Trastuzumab/química , Trastuzumab/genética , Trastuzumab/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Sci Rep ; 8(1): 6458, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29691463

RESUMO

Necrotizing soft tissue infections are lethal polymicrobial infections. Two key microbes that cause necrotizing soft tissue infections are Streptococcus pyogenes and Clostridium perfringens. These pathogens evade innate immunity using multiple virulence factors, including cholesterol-dependent cytolysins (CDCs). CDCs are resisted by mammalian cells through the sequestration and shedding of pores during intrinsic membrane repair. One hypothesis is that vesicle shedding promotes immune evasion by concomitantly eliminating key signaling proteins present in cholesterol-rich microdomains. To test this hypothesis, murine macrophages were challenged with sublytic CDC doses. CDCs suppressed LPS or IFNγ-stimulated TNFα production and CD69 and CD86 surface expression. This suppression was cell intrinsic. Two membrane repair pathways, patch repair and intrinsic repair, might mediate TNFα suppression. However, patch repair did not correlate with TNFα suppression. Intrinsic repair partially contributed to macrophage dysfunction because TLR4 and the IFNγR were partially shed following CDC challenge. Intrinsic repair was not sufficient for suppression, because pore formation was also required. These findings suggest that even when CDCs fail to kill cells, they may impair innate immune signaling responses dependent on cholesterol-rich microdomains. This is one potential mechanism to explain the lethality of S. pyogenes and C. perfringens during necrotizing soft tissue infections.


Assuntos
Citotoxinas/metabolismo , Citotoxinas/farmacologia , Macrófagos/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígeno B7-2/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Clostridium perfringens/metabolismo , Clostridium perfringens/patogenicidade , Citotoxinas/imunologia , Feminino , Proteínas Hemolisinas/metabolismo , Imunidade Inata/imunologia , Imunidade Inata/fisiologia , Lectinas Tipo C/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidade , Fator de Necrose Tumoral alfa/efeitos dos fármacos
3.
J Biomed Sci ; 24(1): 58, 2017 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-28822352

RESUMO

BACKGROUND: Vibrio vulnificus is a marine bacterial species that causes opportunistic infections manifested by serious skin lesions and fulminant septicemia in humans. We have previously shown that the multifunctional autoprocessing repeats in toxin (MARTXVv1) of a biotype 1 V. vulnificus strain promotes survival of this organism in the host by preventing it from engulfment by the phagocytes. The purpose of this study was to further explore how MARTXVv1 inhibits phagocytosis of this microorganism by the macrophage. METHODS: We compared between a wild-type V. vulnificus strain and its MARTXVv1-deficient mutant for a variety of phagocytosis-related responses, including morphological change and activation of signaling molecules, they induced in the macrophage. We also characterized a set of MARTXVv1 domain-deletion mutants to define the regions associated with antiphagocytosis activity. RESULTS: The RAW 264.7 cells and mouse peritoneal exudate macrophages underwent cell rounding accompanied by F-actin disorganization in the presence of MARTXVv1. In addition, phosphorylation of some F-actin rearrangement-associated signaling molecules, including Lyn, Fgr and Hck of the Src family kinases (SFKs), focal adhesion kinase (FAK), proline-rich tyrosine kinase 2 (Pyk2), phosphoinositide 3-kinase (PI3K) and Akt, but not p38, was decreased. By using specific inhibitors, we found that these kinases were all involved in the phagocytosis of MARTXVv1-deficient mutant in an order of SFKs-FAK/Pyk2-PI3K-Akt. Deletion of the effector domains in the central region of MARTXVv1 could lead to reduced cytotoxicity, depending on the region and size of deletion, but did not affect the antiphagocytosis activity and ability to cause rounding of macrophage. Reduced phosphorylation of Akt was closely associated with inhibition of phagocytosis by the wild-type strain and MARTXVv1 domain-deletion mutants, and expression of the constitutively active Akt, myr-Akt, enhanced the engulfment of these strains by macrophage. CONCLUSIONS: MARTXVv1 could inactivate the SFKs-FAK/Pyk2-PI3K-Akt signaling pathway in the macrophages. This might lead to impaired phagocytosis of the V. vulnificus-infected macrophage. The majority of the central region of MARTXVv1 is not associated with the antiphagocytosis activity.


Assuntos
Toxinas Bacterianas/imunologia , Fagocitose/imunologia , Vibrioses/microbiologia , Vibrio vulnificus/imunologia , Vibrio vulnificus/patogenicidade , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Citotoxinas/imunologia , Citotoxinas/metabolismo , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Vibrioses/patologia , Vibrio vulnificus/genética
4.
Immunity ; 45(6): 1258-1269, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27939674

RESUMO

Programmed death and shedding of epithelial cells is a powerful defense mechanism to reduce bacterial burden during infection but this activity cannot be indiscriminate because of the critical barrier function of the epithelium. We report that during cystitis, shedding of infected bladder epithelial cells (BECs) was preceded by the recruitment of mast cells (MCs) directly underneath the superficial epithelium where they docked and extruded their granules. MCs were responding to interleukin-1ß (IL-1ß) secreted by BECs after inflammasome and caspase-1 signaling. Upon uptake of granule-associated chymase (mouse MC protease 4 [mMCPT4]), BECs underwent caspase-1-associated cytolysis and exfoliation. Thus, infected epithelial cells require a specific cue for cytolysis from recruited sentinel inflammatory cells before shedding.


Assuntos
Quimases/imunologia , Citotoxinas/imunologia , Células Epiteliais/microbiologia , Mastócitos/imunologia , Infecções Urinárias/imunologia , Animais , Degranulação Celular/imunologia , Linhagem Celular , Grânulos Citoplasmáticos/química , Feminino , Imunofluorescência , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
J Cyst Fibros ; 15(5): 597-604, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26821814

RESUMO

BACKGROUND: Staphylococcus aureus is one of the earliest bacterial pathogens to colonize the lungs of children with cystic fibrosis and is an important contributor to pulmonary exacerbations. The adaptive host response to S. aureus in cystic fibrosis remains inadequately defined and has important implications for pathogenesis and potential interventions. The objectives of this study were to determine the functional antibody response to select staphylococcal exotoxins (LukAB, alpha-hemolysin, and PVL) in children with cystic fibrosis and to evaluate the relationship of this response with pulmonary exacerbations. METHODS: Fifty children with cystic fibrosis were enrolled and followed prospectively for 12months. Clinical characteristics and serologic profiles were assessed at routine visits and during pulmonary exacerbations, and functional antibody assessments were performed to measure neutralization of LukAB-mediated cytotoxicity. RESULTS: For each antigen, geometric mean titers were significantly higher if S. aureus was detected at the time of exacerbation. For LukAB, geometric mean titers were significantly higher at exacerbation follow-up compared to titers during the exacerbation, consistent with expression during human disease, and the humoral response capably neutralized LukAB-mediated cytotoxicity. Moreover, the presence of a positive S. aureus culture during a pulmonary exacerbation was associated with 31-fold higher odds of having a LukA titer ≥1:160, suggesting potential diagnostic capability of this assay. CONCLUSIONS: The leukotoxin LukAB is expressed by S. aureus and recognized by the human adaptive immune response in the setting of pulmonary infection in cystic fibrosis. Anti-LukAB antibodies were not only predictive of positive staphylococcal culture during exacerbation, but also functional in the neutralization of this toxin.


Assuntos
Proteínas de Bactérias/imunologia , Fibrose Cística , Leucocidinas/imunologia , Infecções Estafilocócicas , Staphylococcus aureus , Imunidade Adaptativa/imunologia , Adolescente , Formação de Anticorpos/imunologia , Criança , Pré-Escolar , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Fibrose Cística/fisiopatologia , Fibrose Cística/terapia , Citotoxinas/imunologia , Feminino , Humanos , Pneumopatias/diagnóstico , Pneumopatias/imunologia , Pneumopatias/fisiopatologia , Masculino , Estudos Prospectivos , Infecções Estafilocócicas/diagnóstico , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/fisiopatologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/isolamento & purificação , Exacerbação dos Sintomas , Estados Unidos
6.
J Immunol ; 195(7): 3190-7, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26297758

RESUMO

The discovery of new vaccines against infectious diseases and cancer requires the development of novel adjuvants with well-defined activities. The TLR4 agonist adjuvant GLA-SE elicits robust Th1 responses to a variety of vaccine Ags and is in clinical development for both infectious diseases and cancer. We demonstrate that immunization with a recombinant protein Ag and GLA-SE also induces granzyme A expression in CD4 T cells and produces cytolytic cells that can be detected in vivo. Surprisingly, these in vivo CTLs were CD4 T cells, not CD8 T cells, and this cytolytic activity was not dependent on granzyme A/B or perforin. Unlike previously reported CD4 CTLs, the transcription factors Tbet and Eomes were not necessary for their development. CTL activity was also independent of the Fas ligand-Fas, TRAIL-DR5, and canonical death pathways, indicating a novel mechanism of CTL activity. Rather, the in vivo CD4 CTL activity induced by vaccination required T cell expression of CD154 (CD40L) and target cell expression of CD40. Thus, vaccination with a TLR4 agonist adjuvant induces CD4 CTLs, which kill through a previously unknown CD154-dependent mechanism.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos CD40/biossíntese , Ligante de CD40/biossíntese , Citotoxinas/imunologia , Proteína Ligante Fas/imunologia , Granzimas/biossíntese , Granzimas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Proteínas com Domínio T/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Células Th1/imunologia , Vacinação
7.
Nat Rev Drug Discov ; 14(7): 487-98, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26000725

RESUMO

Scientific insights into the human immune system have recently led to unprecedented breakthroughs in immunotherapy. In the twenty-first century, drugs and cell-based therapies developed to bolster humoral and T cell immunity represent an established and growing component of cancer therapeutics. Although natural killer (NK) cells have long been known to have advantages over T cells in terms of their capacity to induce antigen-independent host immune responses against malignancies, their therapeutic potential in the clinic has been largely unexplored. A growing number of scientific discoveries into pathways that both activate and suppress NK cell function, as well as methods to sensitize tumours to NK cell cytotoxicity, have led to the development of numerous pharmacological and genetic methods to enhance NK cell antitumour immunity. These findings, as well as advances in our ability to expand NK cells ex vivo and manipulate their capacity to home to the tumour, have now provided investigators with a variety of new methods and strategies to harness the full potential of NK cell-based cancer immunotherapy in the clinic.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Produtos Biológicos/imunologia , Produtos Biológicos/uso terapêutico , Citotoxinas/imunologia , Citotoxinas/uso terapêutico , Humanos , Imunoterapia/tendências
8.
Curr Top Med Chem ; 14(24): 2822-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25487009

RESUMO

Antibody-drug conjugates (ADCs) are an emerging area of study within medicinal chemistry and are thought of as sophisticated drug delivery systems due to their specificity to a disease-targeted antigen. ADCs have been actively utilized as therapeutics for hematological and solid tumor cancers due to their capability to deliver a cytotoxic compound to a specific cancer cell without affecting normal cells. An antibody drug conjugate has three major constituents: a monoclonal antibody (mAb), a chemical linker, and a potent cytotoxic payload. There has been a continuing effort to optimize antibody-drug conjugates, with the primary focus of design and development directed at either the mAb or the chemical linker, with little effort devoted to the optimization of payload compounds. In fact, among the 114 ongoing or recently completed clinical trials, there is generally a lack of diversity in the cytotoxic payloads that are utilized, with only seven payload compounds reported (four additional trials are ongoing with structures that have not been reported). Six of these seven payload compounds are derived from natural product sources, highlighting the importance of natural products as cytotoxic payloads for ADC.


Assuntos
Antígenos de Neoplasias/metabolismo , Antineoplásicos/química , Produtos Biológicos/química , Citotoxinas/química , Neoplasias Hematológicas/tratamento farmacológico , Imunoconjugados/química , Aminoglicosídeos/química , Aminoglicosídeos/imunologia , Aminoglicosídeos/uso terapêutico , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos de Neoplasias/imunologia , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Brentuximab Vedotin , Ensaios Clínicos como Assunto , Citotoxinas/imunologia , Citotoxinas/uso terapêutico , Sistemas de Liberação de Medicamentos , Gemtuzumab , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Imunoconjugados/imunologia , Imunoconjugados/uso terapêutico , Terapia de Alvo Molecular , Ligação Proteica , Trastuzumab
9.
J Pharmacokinet Pharmacodyn ; 41(5): 461-78, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25281420

RESUMO

Cancer therapies that harness the actions of the immune response, such as targeted monoclonal antibody treatments and therapeutic vaccines, are relatively new and promising in the landscape of cancer treatment options. Mathematical modeling and simulation of immune-modifying therapies can help to offset the costs of drug discovery and development, and encourage progress toward new immunotherapies. Despite advances in cancer immunology research, questions such as how the immune system interacts with a growing tumor, and which components of the immune system play significant roles in responding to immunotherapy are still not well understood. Mathematical modeling and simulation are powerful tools that provide an analytical framework in which to address such questions. A quantitative understanding of the kinetics of the immune response to treatment is crucial in designing treatment strategies, such as dosing, timing, and predicting the response to a specific treatment. These models can be used both descriptively and predictively. In this chapter, various mathematical models that address different cancer treatments, including cytotoxic chemotherapy, immunotherapy, and combinations of both treatments, are presented. The aim of this chapter is to highlight the importance of mathematical modeling and simulation in the design of immunotherapy protocols for cancer treatment. The results demonstrate the power of these approaches in explaining determinants that are fundamental to cancer-immune dynamics, therapeutic success, and the development of efficient therapies.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Citotoxinas/imunologia , Citotoxinas/uso terapêutico , Imunoterapia , Modelos Imunológicos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Simulação por Computador , Humanos
10.
Mol Oncol ; 8(2): 378-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24389243

RESUMO

In clinical practice, targeted therapies are usually administered together with chemotherapeutics. However, little is known whether conventional cytotoxic agents enhance the efficacy of targeted compounds, and whether a possible synergy would be dictated by drug-sensitizing genetic alterations. To explore these issues, we leveraged the design of clinical studies in humans to conduct a multi-arm trial in an 'in-cell' format. Using the MET oncogene as a model target and a panel of genetically characterized cell lines as a reference population, we found that two different chemotherapeutic regimens - cisplatin and 5-fluorouracil - exerted widespread cytotoxic activity that was not further enhanced by MET inhibition with a monovalent anti-MET antibody. From a complementary perspective, targeted MET inhibition was successful in a selected complement of cells harboring MET genomic lesions. In this latter setting, addition of chemotherapy did not provide a therapeutic advantage. Mechanistically, chemotherapeutics did not influence the basal activity of MET in cells with normal MET genomic status nor did they contribute to neutralize MET signals in cells with MET amplification. These data suggest that tumors displaying MET aberrations achieve plateau responses by MET monotherapy and do not receive further benefit by addition of cytotoxic treatments.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/farmacologia , Citotoxinas/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Citotoxinas/imunologia , Ensaios de Seleção de Medicamentos Antitumorais , Fluoruracila/farmacologia , Humanos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-met/genética
11.
Mol Cancer Ther ; 12(9): 1774-82, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23832121

RESUMO

Targeted endocytic uptake is a first step toward tissue-specific cytoplasmic macromolecular delivery; however, inefficient escape from the endolysosomal compartment makes this generally impractical at present. We report here a targeted cytolysin approach that dramatically potentiates endosomal release of an independently targeted potent gelonin immunotoxin. Fibronectin domains engineered for affinity to EGF receptor or carcinoembryonic antigen were fused to the plant toxin gelonin or bacterial pore-forming cytolysins. These fusion proteins display synergistic activity in both antigen-specific cytotoxicity in vitro, enhancing potency by several orders of magnitude, and in tumor growth inhibition in vivo. In addition, the number of internalized gelonin molecules required to induce apoptosis is reduced from approximately 5 × 10(6) to less than 10(3). Targeted potentiation shows promise for enhancing cytoplasmic delivery of other macromolecular payloads such as DNA, siRNA, and miRNA.


Assuntos
Antígeno Carcinoembrionário/imunologia , Citotoxinas/imunologia , Endossomos/metabolismo , Receptores ErbB/imunologia , Fibronectinas/metabolismo , Imunotoxinas/imunologia , Proteínas Inativadoras de Ribossomos Tipo 1/administração & dosagem , Proteínas Inativadoras de Ribossomos Tipo 1/imunologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Citoplasma/imunologia , Citotoxinas/farmacologia , Sinergismo Farmacológico , Endossomos/imunologia , Fibronectinas/imunologia , Humanos , Imunotoxinas/farmacologia , Camundongos , Terapia de Alvo Molecular , Especificidade de Órgãos , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia
12.
Am J Rhinol Allergy ; 27(3): 168-76, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23710950

RESUMO

BACKGROUNDS: Diagnosis of allergic fungal rhinosinusitis (AFRS) is complicated because of the presence of fungi on mucosal surfaces of sinonasal passages. The objectives of this study were to define, using immunohistochemistry, lymphocyte populations associated with noninvasive fungal-related chronic rhinosinusitis (CRS; AFRS and FBs [FB]) relative to CRS with nasal polyposis (CRSwNP) and CRS without nasal polyposis (CRSsNP) as a means of diagnosing different forms of CRS. METHODS: Sinus CT scans, nasal endoscopy scores, and the presence of eosinophilic fungal mucin or FBs were used to prospectively define patient groups with CRS who had failed medical treatment and were undergoing endoscopic sinus surgery. Four patient groups were identified: AFRS, FB, CRSwNP, and CRSsNP. Tissue specimens were studied and graded for histopathological changes. Immunophenotyping of mucosal lymphocytes was performed using anti-CD3, -CD20, -CD4, -CD8, -CD56, and -perforin antibodies. RESULTS: Nasal polyposis scores were similar between AFRS and CRSwNP. Radiological changes associated with AFRS can also be present in CRSwNP, e.g., heterogenicity in 9/30 (30%), expansion in 25/30 (83%), and bony attenuation of the ethmoid trabeculae in 19/30 (63%). Different grades of basement membrane thickness, edema, and fibrosis were observed. In both types of noninvasive fungal rhinosinusitis, CD3+ T lymphocytes were most commonly identified. In cases of AFRS, most T cells were CD8+ (p < 0.001). In FB cases, CD4+ lymphocytes were dominant (p < 0.001). In nonfungal CRS cases, CD20+ lymphocytes (B lymphocytes) predominated (p < 0.001). CONCLUSION: Although CT scans and histological examination can assist the diagnosis of rhinosinusitis, tissue immunophenotyping can be used in defining different types of fungal and nonfungal CRS cases.


Assuntos
Antígenos CD/imunologia , Imuno-Histoquímica , Fungos Mitospóricos , Micoses/diagnóstico , Pólipos Nasais/diagnóstico , Rinite/diagnóstico , Sinusite/diagnóstico , Adulto , Antígenos CD20/imunologia , Biomarcadores/metabolismo , Complexo CD3/imunologia , Antígenos CD4/imunologia , Antígeno CD56/imunologia , Antígenos CD8/imunologia , Doença Crônica , Citotoxinas/imunologia , Diagnóstico Diferencial , Eosinófilos/imunologia , Feminino , Humanos , Imuno-Histoquímica/métodos , Imunofenotipagem/métodos , Masculino , Pessoa de Meia-Idade , Fungos Mitospóricos/isolamento & purificação , Micoses/complicações , Micoses/imunologia , Micoses/terapia , Pólipos Nasais/imunologia , Pólipos Nasais/microbiologia , Pólipos Nasais/terapia , Perforina/imunologia , Estudos Prospectivos , Rinite/imunologia , Rinite/microbiologia , Rinite/terapia , Rinite Alérgica , Rinite Alérgica Perene/diagnóstico , Sinusite/imunologia , Sinusite/microbiologia , Sinusite/terapia
13.
Vaccine ; 30(34): 5099-109, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22691432

RESUMO

Staphylococcus aureus causes significant illnesses throughout the world, including toxic shock syndrome (TSS), pneumonia, and infective endocarditis. Major contributors to S. aureus illnesses are secreted virulence factors it produces, including superantigens and cytolysins. This study investigates the use of superantigens and cytolysins as staphylococcal vaccine candidates. Importantly, 20% of humans and 50% of rabbits in our TSS model cannot generate antibody responses to native superantigens. We generated three TSST-1 mutants; G31S/S32P, H135A, and Q136A. All rabbits administered these TSST-1 toxoids generated strong antibody responses (titers>10,000) that neutralized native TSST-1 in TSS models, both in vitro and in vivo. These TSST-1 mutants lacked detectable residual toxicity. Additionally, the TSST-1 mutants exhibited intrinsic adjuvant activity, increasing antibody responses to a second staphylococcal antigen (ß-toxin). This effect may be due to TSST-1 mutants binding to the immune co-stimulatory molecule CD40. The superantigens TSST-1 and SEC and the cytolysin α-toxin are known to contribute to staphylococcal pneumonia. Immunization of rabbits against these secreted toxins provided complete protection from highly lethal challenge with a USA200 S. aureus strain producing all three exotoxins; USA200 strains are common causes of staphylococcal infections. The same three exotoxins plus the cytolysins ß-toxin and γ-toxin contribute to infective endocarditis and sepsis caused by USA200 strains. Immunization against these five exotoxins protected rabbits from infective endocarditis and lethal sepsis. These data suggest that immunization against toxoid proteins of S. aureus exotoxins protects from serious illnesses, and concurrently superantigen toxoid mutants provide endogenous adjuvant activity.


Assuntos
Toxinas Bacterianas/imunologia , Citotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Coelhos/imunologia , Infecções Estafilocócicas/terapia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Formação de Anticorpos , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/administração & dosagem , Antígenos CD40/imunologia , Linhagem Celular , Citotoxinas/administração & dosagem , Endocardite Bacteriana/imunologia , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/terapia , Exotoxinas/imunologia , Feminino , Proteínas Hemolisinas/administração & dosagem , Humanos , Masculino , Testes de Neutralização , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/terapia , Coelhos/microbiologia , Choque Séptico/imunologia , Choque Séptico/microbiologia , Choque Séptico/terapia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Toxoide Estafilocócico/administração & dosagem , Toxoide Estafilocócico/imunologia , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Superantígenos/administração & dosagem , Vacinação
14.
Blood ; 119(16): 3767-78, 2012 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-22271448

RESUMO

We describe the use of novel bispecific hexavalent Abs (HexAbs) to enhance anticancer immunotherapy. Two bispecific HexAbs [IgG-(Fab)(4) constructed from veltuzumab (anti-CD20 IgG) and milatuzumab (anti-CD74 IgG)] show enhanced cytotoxicity in mantle cell lymphoma (MCL) and other lymphoma/leukemia cell lines, as well as patient tumor samples, without a crosslinking Ab, compared with their parental mAb counterparts, alone or in combination. The bispecific HexAbs have different properties from and are more potent than their parental mAbs in vitro. The juxtaposition of CD20 and CD74 on MCL cells by the HexAbs resulted in homotypic adhesion and triggered intracellular changes that include loss of mitochondrial transmembrane potential, production of reactive oxygen species, rapid and sustained phosphorylation of ERKs and JNK, down-regulation of pAkt and Bcl-xL, actin reorganization, and lysosomal membrane permeabilization, culminating in cell death. They also displayed different potencies in depleting lymphoma cells and normal B cells from whole blood ex vivo and significantly extended the survival of nude mice bearing MCL xenografts in a dose-dependent manner, thus indicating stability and antitumor activity in vivo. Such bispecific HexAbs may constitute a new class of therapeutic agents for improved cancer immunotherapy, as shown here for MCL and other CD20(+)/CD74(+) malignancies.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antígenos CD20/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Linfoma de Célula do Manto/tratamento farmacológico , Linfoma de Célula do Manto/imunologia , Citoesqueleto de Actina/imunologia , Citoesqueleto de Actina/metabolismo , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linhagem Celular Tumoral , Citotoxinas/imunologia , Citotoxinas/farmacologia , Desenho de Fármacos , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Imunoterapia/métodos , Lisossomos/imunologia , Lisossomos/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Immunol Res ; 50(2-3): 118-23, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21717083

RESUMO

Toxins secreted by bacteria can impact the host in a number of different ways. In some infections, toxins play a crucial and central role in pathogenesis (i.e., anthrax), while in other bacterial infections, the role of toxins is less understood. The cholesterol-dependent cytolysins (CDCs), of which streptolysin O is a prototype, are a class of pore-forming toxins produced by many gram-positive bacteria and have only been studied in a few experimental infection models. Our laboratory has demonstrated that CDCs have effects on macrophages that are both pro- and anti-inflammatory. Here, we review evidence that CDCs promote inflammation by driving secretion of IL-1ß and HMGB-1 from macrophages in a NLRP3-dependent manner, while also causing shedding of membrane microvesicles from cells that can interact with macrophages and inhibit TNF-α release. CDCs thus impact macrophage function in ways that may be both beneficial and detrimental to the host.


Assuntos
Toxinas Bacterianas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Colesterol/metabolismo , Citotoxinas/imunologia , Citotoxinas/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Toxina Tetânica/metabolismo , Toxina Tetânica/farmacologia
17.
Biochemistry ; 50(33): 7157-67, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21749039

RESUMO

Pulsed-field gel electrophoresis (PFGE) clonal type USA200 is the most widely disseminated Staphylococcus aureus colonizer of the nose and is a major cause of toxic shock syndrome (TSS). Exoproteins derived from these organisms have been suggested to contribute to their colonization and causation of human diseases but have not been well-characterized. Two representative S. aureus USA200 isolates, MNPE (α-toxin positive) and CDC587 (α-toxin mutant), isolated from pulmonary post-influenza TSS and menstrual vaginal TSS, respectively, were evaluated. Biochemical, immunobiological, and cell-based assays, including mass spectrometry, were used to identify key exoproteins derived from the strains that are responsible for proinflammatory and cytotoxic activity on human vaginal epithelial cells. Exoproteins associated with virulence were produced by both strains, and cytolysins (α-toxin and γ-toxin), superantigens, and proteases were identified as the major exoproteins, which caused epithelial cell inflammation and cytotoxicity. Exoprotein fractions from MNPE were more proinflammatory and cytotoxic than those from CDC587 due to high concentrations of α-toxin. CDC587 produced a small amount of α-toxin, despite the presence of a stop codon (TAG) at codon 113. Additional exotoxin identification studies of USA200 strain [S. aureus MN8 (α-toxin mutant)] confirmed that MN8 also produced low levels of α-toxin despite the same stop codon. The differences observed in virulence factor profiles of two USA200 strains provide insight into environmental factors that select for specific virulence factors. Cytolysins, superantigens, and proteases were identified as potential targets, where toxin neutralization may prevent or diminish epithelial damage associated with S. aureus.


Assuntos
Citotoxinas/imunologia , Enterotoxinas/imunologia , Exotoxinas/imunologia , Choque Séptico/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Vagina/imunologia , Animais , Cromatografia Líquida de Alta Pressão , Citotoxinas/metabolismo , Eletroforese em Gel de Campo Pulsado , Enterotoxinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Exotoxinas/metabolismo , Feminino , Humanos , Immunoblotting , Imunoglobulina G/imunologia , Interleucina-8/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Coelhos , Choque Séptico/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/isolamento & purificação , Superantígenos/metabolismo , Suínos , Vagina/metabolismo , Vagina/microbiologia , Fatores de Virulência
18.
J Infect Dis ; 203(12): 1866-74, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21422475

RESUMO

Vibrio vulnificus is a marine bacterium causing serious septicemia and wound infection in humans. It produces an RTX toxin that can lyse a variety of cells and is important for virulence in mice. In this study, we explored the role of RTX in pathogenesis by characterizing an RTX-deficient mutant. This mutant showed an ∼2-log reduction in virulence for mice infected by various routes. Survival of the mutant at the infection site and subsequent spread into the bloodstream were impaired. In mice pretreated with cyclophosphamide to deplete the neutrophils, both the virulence and survival at the infection site of this mutant were enhanced. This mutant was further shown to be more readily cleared from the macrophage-rich mouse peritoneal cavity and phagocytosed by murine macrophages. These findings suggest that the RTX of V. vulnificus is required for bacterial survival during infection by protecting the organism from phagocytosis.


Assuntos
Toxinas Bacterianas/imunologia , Fagocitose/imunologia , Vibrioses/microbiologia , Vibrio vulnificus/imunologia , Vibrio vulnificus/patogenicidade , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Linhagem Celular , Citotoxinas/imunologia , Citotoxinas/metabolismo , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Mutação , Reação em Cadeia da Polimerase , Vibrioses/patologia , Vibrio vulnificus/genética , Virulência
19.
J Biol Chem ; 286(12): 10258-64, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21247889

RESUMO

Heat shock-binding protein HspBP1 is a member of the Hsp70 co-chaperone family. The interaction between HspBP1 and the ATPase domain of the major heat shock protein Hsp70 up-regulates nucleotide exchange and reduces the affinity between Hsp70 and the peptide in its peptide-binding site. Previously we have shown that Tag7 (also known as peptidoglycan recognition protein PGRP-S), an innate immunity protein, interacts with Hsp70 to form a stable Tag7-Hsp70 complex with cytotoxic activity against some tumor cell lines. This complex can be produced in cytotoxic lymphocytes and released during interaction with tumor cells. Here the effect of HspBP1 on the cytotoxic activity of the Tag7-Hsp70 complex was examined. HspBP1 could bind not only to Hsp70, but also to Tag7. This interaction eliminated the cytotoxic activity of Tag7-Hsp70 complex and decreased the ATP concentration required to dissociate Tag7 from the peptide-binding site of Hsp70. Moreover, HspBP1 inhibited the cytotoxic activity of the Tag7-Hsp70 complex secreted by lymphocytes. HspBP1 was detected in cytotoxic CD8+ lymphocytes. This protein was released simultaneously with Tag7-Hsp70 during interaction of these lymphocytes with tumor cells. The simultaneous secretion of the cytotoxic complex with its inhibitor could be a mechanism protecting normal cells from the cytotoxic effect of this complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Citocinas/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Complexos Multiproteicos/metabolismo , Neoplasias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/genética , Citocinas/imunologia , Citotoxinas/genética , Citotoxinas/imunologia , Citotoxinas/metabolismo , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/imunologia , Humanos , Células K562 , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Neoplasias/genética , Neoplasias/imunologia , Ligação Proteica
20.
J Liposome Res ; 21(2): 124-33, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20550461

RESUMO

Vaccine development today encounters a main obstacle, which is the need for effective adjuvants suitable for clinical trials. Aluminum salts, discovered 70 years ago and, very recently, MF59, are the only types of adjuvants currently used in vaccines licensed by the U.S. Food and Drug Administration. Liposomes represent an alternative approach to vaccine adjuvants. In this article, we describe the inflammatory response and biological effect of polymerization and the addition of positive charges in liposome formulations. Nonpolymerized cationic (NP(+)) liposomes significantly reduce metabolism in Vero cells after 24 hours. Correspondingly, both NP(+) and polymerized cationic (P(+)) liposomes reduce cell viability following a 48-hour incubation. Similar results were obtained with cells from the peritoneal cavities of mice. Paradoxically, those liposomes that presented clearly cytostatic or cytotoxic effects in vitro stimulated metabolism and had a mitogenic effect in vivo. Finally, the adjuvant effect was tested by immunization in BALB/c mice. The major effect was obtained with NP(+) liposomes. Accordingly, we also demonstrated that NP(+) liposomes injected into the dermis produced an outstanding inflammatory reaction, showing the histopathological characteristics of an inoculation granuloma. Thus, positive charge would play an important role in the immunoadjuvant effect of liposomes by conferring them cytotoxic capacity.


Assuntos
Adjuvantes Imunológicos/química , Citotoxinas/imunologia , Lipossomos/imunologia , Macrófagos Peritoneais/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Cátions/química , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Citotoxinas/administração & dosagem , Citotoxinas/química , Relação Dose-Resposta a Droga , Imunização , Injeções Intradérmicas , Lipossomos/administração & dosagem , Lipossomos/química , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Polimerização/efeitos da radiação , Eletricidade Estática , Raios Ultravioleta , Vacinas/química , Vacinas/imunologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA