Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 10: 601, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001248

RESUMO

Therapeutic antibodies have the potential to induce immunogenicity leading to the development of anti-drug antibodies (ADA) that consequently may result in reduced serum drug concentrations, a loss of efficacy or potential hypersensitivity reactions. Among other factors, aggregated antibodies have been suggested to promote immunogenicity, thus enhancing ADA production. Dendritic cells (DC) are the most efficient antigen-presenting cell population and are crucial for the initiation of T cell responses and the subsequent generation of an adaptive immune response. This work focuses on the development of predictive in vitro assays that can monitor DC maturation, in order to determine whether drug products have direct DC stimulatory capabilities. To this end, four independent laboratories aligned a common protocol to differentiate human monocyte-derived DC (moDC) that were treated with either native or aggregated preparations of infliximab, natalizumab, adalimumab, or rituximab. These drug products were subjected to different forms of physical stress, heat and shear, resulting in aggregation and the formation of subvisible particles. Each partner developed and optimized assays to monitor diverse end-points of moDC maturation: measuring the upregulation of DC activation markers via flow cytometry, analyzing cytokine, and chemokine production via mRNA and protein quantification and identifying cell signaling pathways via quantification of protein phosphorylation. These study results indicated that infliximab, with the highest propensity to form aggregates when heat-stressed, induced a marked activation of moDC as measured by an increase in CD83 and CD86 surface expression, IL-1ß, IL-6, IL-8, IL-12, TNFα, CCL3, and CCL4 transcript upregulation and release of respective proteins, and phosphorylation of the intracellular signaling proteins Syk, ERK1/2, and Akt. In contrast, natalizumab, which does not aggregate under these stress conditions, induced no DC activation in any assay system, whereas adalimumab or rituximab aggregates induced only slight parameter variation. Importantly, the data generated in the different assay systems by each partner site correlated and supported the use of these assays to monitor drug-intrinsic propensities to drive maturation of DC. This moDC assay is also a valuable tool as an in vitro model to assess the intracellular mechanisms that drive DC activation by aggregated therapeutic proteins.


Assuntos
Anticorpos Monoclonais/farmacologia , Células Dendríticas/efeitos dos fármacos , Bioensaio , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos
2.
J Immunol Res ; 2018: 5463879, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29651443

RESUMO

Dendritic cell (DC) immunotherapy has been effective for prevention of type 1 diabetes (T1D) in NOD mice but fails to protect if initiated after active autoimmunity. As autoreactivity expands inter- and intramolecularly during disease progression, we investigated whether DCs unpulsed or pulsed with ß cell antigenic dominant determinants (DD), subdominant determinants (SD), and ignored determinants (ID) could prevent T1D in mice with advanced insulitis. We found that diabetes was significantly delayed by DC therapy. Of interest, DCs pulsed with SD or ID appeared to provide better protection. T lymphocytes from DC-treated mice acquired spontaneous proliferating capability during in vitro culture, which could be largely eliminated by IL-2 neutralizing antibodies. This trend maintained even 29 weeks after discontinuing DC therapy and appeared antigen-independent. Furthermore, CD4+Foxp3+ T regulatory cells (Tregs) from DC-treated mice proliferated more actively in vitro compared to the controls, and Tregs from DC-treated mice showed significantly enhanced immunosuppressive activities in contrast to those from the controls. Our study demonstrates that DC therapy leads to long-lasting immunomodulatory effects in an antigen-dependent and antigen-independent manner and provides evidence for peptide-based intervention during a clinically relevant window to guide DC-based immunotherapy for autoimmune diabetes.


Assuntos
Células Dendríticas/fisiologia , Diabetes Mellitus Tipo 1/terapia , Imunoterapia Adotiva/métodos , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno , Autoantígenos/imunologia , Autoantígenos/metabolismo , Autoimunidade , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Tolerância Imunológica , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/metabolismo , Imunomodulação , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD
3.
Chemistry ; 22(24): 8158-66, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27139508

RESUMO

A new dimeric macrolide xylopyranoside, cocosolide (1), was isolated from the marine cyanobacterium preliminarily identified as Symploca sp. from Guam. The structure was determined by a combination of NMR spectroscopy, HRMS, X-ray diffraction studies and Mosher's analysis of the base hydrolysis product. Its carbon skeleton closely resembles that of clavosolides A-D isolated from the sponge Myriastra clavosa, for which no bioactivity is known. We performed the first total synthesis of cocosolide (1) along with its [α,α]-anomer (26) and macrocyclic core (28), thus leading to the confirmation of the structure of natural 1. The convergent synthesis featured Wadsworth-Emmons cyclopropanation, Sakurai annulation, Yamaguchi macrocyclization/dimerization reaction, α-selective glycosidation and ß-selective glycosidation. Compounds 1 and 26 potently inhibited IL-2 production in both T-cell receptor dependent and independent manners. Full activity requires the presence of the sugar moiety as well as the intact dimeric structure. Cocosolide also suppressed the proliferation of anti-CD3-stimulated T-cells in a dose-dependent manner.


Assuntos
Cianobactérias/química , Glicosídeos/síntese química , Imunossupressores/síntese química , Macrolídeos/química , Animais , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Bacillus cereus/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Cianobactérias/metabolismo , Dimerização , Avaliação Pré-Clínica de Medicamentos , Glicosídeos/química , Glicosilação , Células HCT116 , Humanos , Imunossupressores/química , Imunossupressores/farmacologia , Interleucina-2/metabolismo , Células Jurkat , Lipopolissacarídeos/toxicidade , Macrolídeos/síntese química , Macrolídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Conformação Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Óxido Nítrico/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Células RAW 264.7 , Estereoisomerismo
4.
Bioorg Med Chem ; 20(15): 4774-80, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22748705

RESUMO

The linear depsipeptide grassystatin A, a valuable probe for the study of cathepsin E function, has been synthesized by a [4+6] strategy. It exhibited specific inhibitory activity against cathepsin E with an IC(50) value of 0.8 nM. Our studies indicated that inhibition of cathepsin E did not have an impact on ovalbumin antigen processing and peptide presentation, unique from studies of other aspartic protease inhibitors.


Assuntos
Catepsina E/antagonistas & inibidores , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Animais , Catepsina E/metabolismo , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Relação Estrutura-Atividade
5.
Ann N Y Acad Sci ; 1079: 153-6, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17130547

RESUMO

Our previous work demonstrated peptide-pulsed mature myeloid dendritic cells (DC) presenting beta cell antigens induce tolerance. Here we determine whether immature DC (iDC) presenting dominant (insulin beta9-23 chain, proinsulin C19-A3) or ignored (glutamic acid decarboxylase 65(78-97)) antigen determinants promote tolerance. Nonobese diabetic (NOD) mice were given injections of either unpulsed or peptide-pulsed myeloid iDC beginning at 9 weeks of age for 3 consecutive weeks. Diabetes incidence in recipients of unpulsed iDC was comparable to unmanipulated animals ( approximately 80%), whereas GAD65(78-97) pulsed iDC recipients were protected from the disease (P = 0.05). We also analyzed splenic T cell proliferation responses to the panel of studied peptides in diabetic and nondiabetic recipients. When stimulated with insulin or proinsulin peptide, nondiabetic mice receiving the peptide-pulsed iDC had a 21- to 31-fold or 3.9- to 9.0-fold reduction in T cell response, respectively, as compared to the response of diabetic unpulsed recipients. However, only a 2.6- to 3.1-fold reduction in response to beta chain peptide, and a 1.5- to 3.4-fold reduction in proinsulin response were observed in diabetic mice receiving peptide-pulsed iDC. The reduction was not specific to the immunizing peptide, as reduced proliferation was observed to other diabetes-target peptides. We conclude that protective iDC-based therapies require target antigen presentation, and ignored determinants may be preferable perhaps due to an available naïve T cell repertoire. In addition, iDC presenting peptides induce a nonspecific reduction in T cell responses to beta cell antigens, possibly through the induction of regulatory T cells.


Assuntos
Transferência Adotiva , Antígenos/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Ilhotas Pancreáticas/imunologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Células Cultivadas , Células Dendríticas/transplante , Feminino , Glutamato Descarboxilase/imunologia , Tolerância Imunológica , Insulina/farmacologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Baço/citologia , Baço/metabolismo , Linfócitos T/imunologia , Linfócitos T/fisiologia
6.
Autoimmun Rev ; 5(6): 419-23, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16890897

RESUMO

Type I diabetes (TID) is an autoimmune disease characterized by a T cell-mediated destruction of insulin-producing beta cells. The destructive response is believed to be caused by a Th1-dominant immune attack targeted to several autoantigens including glutamate decarboxylase (GAD) and insulin in the presence of an ineffective regulatory response. The development of both the Th1 biased effector cells as well as regulatory T-cell response can be guided by dendritic cells (DC), professional antigen presenting cells (APC) that efficiently capture and process self antigens, and present them to T-cells. These APC can either prime effector T cells or activate regulatory T cells depending on the function of the DC or perhaps distinct DC subsets. Because DC uniquely orchestrate the delicate balance between T cell immunity and regulation, efforts are being made to investigate the potential of DC therapy for the prevention and/or treatment of autoimmune diseases such as TID through augmentation of regulatory responses. As the subset and functional stage of DC appear to be critical for tolerance induction, several strategies for engineering these cells are emerging. Furthermore, the delineation of T1D-associated target antigens allows for the development of antigen-specific DC-based therapy. Here we review recent advances and considerations for this exciting approach and discuss the selection of the appropriate DC subset, self-peptide, and route of administration for the optimization of immunotherapy using these cells.


Assuntos
Células Dendríticas/fisiologia , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/terapia , Animais , Vias de Administração de Medicamentos , Engenharia Genética , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos NOD
7.
Clin Diagn Lab Immunol ; 10(1): 13-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12522033

RESUMO

Recombinase activation gene 1 (RAG-1) function is essential for V(D)J recombination in T-cell-receptor and immunoglobulin rearrangements whereby the immune system may encode memories of a vast array of antigens. The RAG-1 gene is also localized to neurons in the hippocampal formation and related limbic regions that are involved in spatial learning and memory as well as other parameters of neurobehavioral performance. Since the unique ability to encode memory is shared by the immune system and the brain, we tested the hypothesis that loss of the RAG-1 gene in the brain would influence learning and memory performance and examined several different domains of behavior in RAG-1-knockout and control mice. Compared to control mice, RAG-1-knockout mice exhibited increased locomotor activity in an open field under both dim and bright lighting conditions and decreased habituation (reduction in the expected decline in locomotor activity with increasing familiarity with the novel environment in a 1-h test session) in bright lighting. RAG-1-knockout mice also showed reduced levels of fearfulness for some measures of fear-motivated behavior in both the open-field behavior test and elevated-plus maze test. Contrary to our hypothesis, no differences in spatial learning and memory were found between the groups, although modest differences were observed visible-platform testing in the Morris water maze. Neither prepulse inhibition, a measure of sensorimotor gating, nor reflexive acoustic startle responses differed between the RAG-1-knockout and control mice. It remains to be determined if these changes are due to the loss of RAG-1 gene expression in the brain, are due to the absence of the gene in the immune system (e.g., the loss of cytokines with neuromodulatory activities), or are due to some combination of both effects. Study of the neurobiological actions of RAG-1 in the brain may provide new insights into important processes involved in normal brain function and disease.


Assuntos
Química Encefálica/genética , Deleção de Genes , Genes RAG-1/fisiologia , Animais , Medo , Genes RAG-1/genética , Hipocampo , Sistema Imunitário , Aprendizagem , Memória , Camundongos , Camundongos Knockout , Atividade Motora/genética
8.
J Neuroimmunol ; 134(1-2): 95-103, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12507776

RESUMO

Following facial nerve axotomy in mice, T cells cross the intact blood-brain barrier (BBB), home to nerve cell bodies in the facial motor nucleus (FMN), and augment neuroregenerative processes. The pivotal T cell immunoregulatory cytokine, IL-2, appears to have bidirectional effects on neuronal and microglial cell function, suggesting rival hypotheses that IL-2 could either enhance or disrupt processes associated with regeneration of axotomized facial motor neurons. We tested these competing hypotheses by comparing the effect of facial nerve axotomy on C57BL/6-IL-2(-/-) knockout and C57BL/6-IL-2(+/+) wild-type littermates. Since IL-2 may also be produced endogenously in the brain, we also sought to determine whether differences between the knockout and wild-type mice were attributable to loss of IL-2 gene expression in the CNS, loss of peripheral sources of IL-2 and the associated effects on T cell function, or a combination of these factors. To address this question, we bred novel congenic mice with the SCID mutation (mice lacking T cell derived IL-2) that were homozygous for either the IL-2 knockout or wild-type gene alleles (C57BL/6scid-IL-2(-/-) and C57BL/6scid-IL-2(+/+) littermates, respectively). Groups were assessed for differences in (1) T lymphocytes entering the axotomized FMN; (2) perineuronal CD11b(+) microglial phagocytic clusters, a measure of motor neuron death; and (3) activated microglial cells as measured by MHC-II positivity. C57BL/6-IL-2(-/-) knockout mice had significantly higher numbers of T cells and lower numbers of activated MHC-II-positive microglial cells in the regenerating FMN than wild-type littermates, although the number of CD11b(+) phagocytic microglia clusters did not differ. Thus, despite the significant impairment of T cell function known to be associated with loss of peripheral IL-2, the increased number of T cells entering the axotomized FMN appears to have sufficient activity to support neuroregenerative processes. Congenic C57BL/6scid-IL-2(-/-) knockout mice had lower numbers of CD11b(+) microglial phagocytic clusters than congenic C57BL/6scid-IL-2(+/+) wild-type littermates, suggesting that loss of the IL-2 gene in the CNS (and possibly the loss of other unknown sources of the gene) enhanced neuronal regeneration. Further study of IL-2's complex actions in neuronal injury may provide greater understanding of key variables that determine whether or not immunological processes in the brain are proregenerative.


Assuntos
Quimiotaxia de Leucócito/genética , Traumatismos do Nervo Facial/genética , Gliose/genética , Interleucina-2/deficiência , Microglia/imunologia , Regeneração Nervosa/genética , Linfócitos T/imunologia , Animais , Quimiotaxia de Leucócito/imunologia , Nervo Facial/citologia , Nervo Facial/imunologia , Nervo Facial/metabolismo , Traumatismos do Nervo Facial/imunologia , Traumatismos do Nervo Facial/fisiopatologia , Feminino , Gliose/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Imuno-Histoquímica , Interleucina-2/genética , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Microglia/citologia , Microglia/metabolismo , Neurônios Motores/citologia , Neurônios Motores/imunologia , Neurônios Motores/metabolismo , Mutação/genética , Regeneração Nervosa/imunologia , Degeneração Retrógrada/genética , Degeneração Retrógrada/imunologia , Degeneração Retrógrada/fisiopatologia , Linfócitos T/citologia , Linfócitos T/metabolismo
9.
Neurosci Lett ; 328(3): 304-8, 2002 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-12147331

RESUMO

MRL-lpr mice develop systemic lupus-like autoimmune disease associated with changes in emotional reactivity and spatial learning and memory. Although the major immunological deficit in MRL-lpr mice is uncontrolled lymphoproliferation associated with a Fas gene mutation, these mice have a marked deficit in interleukin-2 (IL-2) production which, when treated, can prevent the development of autoimmune disease. Moreover, both MRL-lpr and IL-2 knockout mice manifest alterations in hippocampal cytoarchitecture and cognitive behavior. We found previously that IL-2 knockout mice have alterations in prepulse inhibition (PPI), a measure of sensorimotor gating. Thus, the present study sought to test the hypothesis that that PPI would be altered in MRL-lpr mice. Compared to MRL(+/+) control mice, MRL-lpr mice exhibited different patterns of PPI during development. Whereas 7 and 12-week MRL-lpr mice with evidence of autoimmune disease (the onset and early stages, respectively) showed increased PPI, 5 week predisease MRL-lpr mice did not. MRL-lpr mice also exhibited increased acoustic startle reactivity that was independent of autoimmune disease. These behavioral changes were not associated with increased brain expression of the proinflammatory cytokines genes, IL-1alpha and IL-6, CD3, or c-myc-associated apoptosis.


Assuntos
Doenças Autoimunes/genética , Interleucina-2/deficiência , Camundongos Endogâmicos MRL lpr/fisiologia , Atividade Motora , Sensação , Estimulação Acústica , Envelhecimento/fisiologia , Animais , Apoptose , Comportamento Animal/fisiologia , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Sistema Nervoso/fisiopatologia , Valores de Referência , Reflexo de Sobressalto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...