Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 198: 27-36, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30041943

RESUMO

Most antigen discovery and vaccine development aimed at driving functional B cell responses rely on mouse immunizations studies. To date, there is no 3D ex vivo immune tissues, which are capable of driving antigen-specific B cell responses to rapidly determine the humoral immunogenicity of antigens, understand the role of extracellular matrix in humoral immunity, and generate high affinity antibody responses. This can be attributed to the complexity of B cell differentiation and affinity maturation process in the germinal center (GC) reaction, which makes these highly specialized cells susceptible to rapid apoptosis ex vivo. We have previously reported immune tissues that show ex vivo GC-like response, however in a non-antigen specific manner. Here, we report a maleimide (MAL)-functionalized polyethylene glycol (PEG)-based designer immune tissues that modulate B cell differentiation and enriches antigen-specific GC B cells in the presence of T-cell like signals. With the 3D synthetic immune tissue platform, we assessed various hydrogel design parameters to control ex vivo GC reaction. Using an Ezh2fl/fl Cγ1-cre transgenic mouse model, we demonstrated ex vivo IgG1 antibody class switching. Using immune tissues developed from a B1-8hi mutant mouse that represents a recombined antibody variable region derived from a 4-hydroxy-3-nitrophenylacetyl (NP) hapten binding antibody (B1-8), we demonstrate antigen specificity and selective enrichment of antigen-specific B cells with high affinity at both cell surface and secreted levels in integrin ligand-dependent manner. The ex vivo antigen-specific platform technology offers use in scientific understanding of immunobiology, matrix immunology, and in biotechnology applications, ranging from the antigen testing, vaccine development, and generation of antibodies against diseases.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Linfócitos T/imunologia , Engenharia Tecidual/métodos , Animais , Antígenos/imunologia , Linfócitos B/citologia , Materiais Biocompatíveis/química , Diferenciação Celular , Células Cultivadas , Feminino , Centro Germinativo/citologia , Imunoglobulina G/imunologia , Masculino , Maleimidas/química , Camundongos Endogâmicos C57BL , Polietilenoglicóis/química , Linfócitos T/citologia
2.
Cell Rep ; 23(2): 499-511, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642007

RESUMO

The role of microenvironment-mediated biophysical forces in human lymphomas remains elusive. Diffuse large B cell lymphomas (DLBCLs) are heterogeneous tumors, which originate from highly proliferative germinal center B cells. These tumors, their associated neo-vessels, and lymphatics presumably expose cells to particular fluid flow and survival signals. Here, we show that fluid flow enhances proliferation and modulates response of DLBCLs to specific therapeutic agents. Fluid flow upregulates surface expression of B cell receptors (BCRs) and integrin receptors in subsets of ABC-DLBCLs with either CD79A/B mutations or WT BCRs, similar to what is observed with xenografted human tumors in mice. Fluid flow differentially upregulates signaling targets, such as SYK and p70S6K, in ABC-DLBCLs. By selective knockdown of CD79B and inhibition of signaling targets, we provide mechanistic insights into how fluid flow mechanomodulates BCRs and integrins in ABC-DLBCLs. These findings redefine microenvironment factors that regulate lymphoma-drug interactions and will be critical for testing targeted therapies.


Assuntos
Linfoma Difuso de Grandes Células B/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Animais , Apoptose/efeitos dos fármacos , Antígenos CD79/antagonistas & inibidores , Antígenos CD79/genética , Antígenos CD79/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Doxorrubicina/farmacologia , Humanos , Integrinas/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Camundongos Endogâmicos NOD , Microfluídica/instrumentação , Microfluídica/métodos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Antígenos de Linfócitos B/genética , Resistência ao Cisalhamento , Transdução de Sinais , Microambiente Tumoral , Regulação para Cima , Quinases da Família src/metabolismo
3.
J Biomed Mater Res A ; 106(7): 2048-2058, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29577576

RESUMO

Nanoparticle shape has emerged as a key regulator of nanoparticle transport across physiological barriers, intracellular uptake, and biodistribution. We report a facile approach to synthesize ellipsoidal nanoparticles through self-assembly of poly(glycerol sebacate)-co-poly(ethylene glycol) (PGS-co-PEG). The PGS-PEG nanoparticle system is highly tunable, and the semiaxis length of the nanoparticles can be modulated by changing PGS-PEG molar ratio and incorporating therapeutics. As both PGS and PEG are highly biocompatible, the PGS-co-PEG nanoparticles show high hemo-, immuno-, and cytocompatibility. Our data suggest that PGS-co-PEG nanoparticles have the potential for use in a wide range of biomedical applications including regenerative medicine, stem cell engineering, immune modulation, and cancer therapeutics. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2048-2058, 2018.


Assuntos
Decanoatos/química , Sistemas de Liberação de Medicamentos/métodos , Glicerol/análogos & derivados , Nanopartículas/química , Polietilenoglicóis/química , Polímeros/química , Animais , Linhagem Celular , Decanoatos/síntese química , Endocitose , Glicerol/síntese química , Glicerol/química , Espaço Intracelular , Camundongos , Nanopartículas/ultraestrutura , Polietilenoglicóis/síntese química , Polímeros/síntese química
4.
Nat Commun ; 8(1): 877, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29026085

RESUMO

The EZH2 histone methyltransferase is required for B cells to form germinal centers (GC). Here we show that EZH2 mediates GC formation through repression of cyclin-dependent kinase inhibitor CDKN1A (p21Cip1). Deletion of Cdkn1a rescues the GC reaction in Ezh2 -/- mice. Using a 3D B cell follicular organoid system that mimics the GC reaction, we show that depletion of EZH2 suppresses G1 to S phase transition of GC B cells in a Cdkn1a-dependent manner. GC B cells of Cdkn1a -/- Ezh2 -/- mice have high levels of phospho-Rb, indicating that loss of Cdkn1a enables progression of cell cycle. Moreover, the transcription factor E2F1 induces EZH2 during the GC reaction. E2f1 -/- mice manifest impaired GC responses, which is rescued by restoring EZH2 expression, thus defining a positive feedback loop in which EZH2 controls GC B cell proliferation by suppressing CDKN1A, enabling cell cycle progression with a concomitant phosphorylation of Rb and release of E2F1.The histone methyltransferase EZH2 silences genes by generating H3K27me3 marks. Here the authors use a 3D GC organoid and show EZH2 mediates germinal centre (GC) formation through epigenetic silencing of CDKN1A and release of cell cycle checkpoints.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/antagonistas & inibidores , Fator de Transcrição E2F1/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Centro Germinativo/metabolismo , Animais , Proliferação de Células , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Retroalimentação Fisiológica , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Inativação Gênica , Camundongos
5.
ACS Biomater Sci Eng ; 3(2): 214-225, 2017 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-33450794

RESUMO

Germinal centers are dynamic structures within lymphoid tissues, which develop once B cells receive activating signals from surrounding immune cells. Germinal center B cells are small in number, heterogeneous, and prone to rapid apoptosis unless selected by the body to form memory B cells. Despite extensive research in the B cell differentiation process, the role of the lymphoid niche, in particular integrin ligands, in the development of early germinal center-like phenotype remains unclear. Here, we report a biomaterials-based modular immune organoid that enables development of early germinal-center phenotype in an integrin ligand-specific manner. We demonstrate the differential role of integrin α4ß1- and αvß3-binding ligands in the induction of GL7+ (GC-like) and GL7- (non-GC-like) phenotype in differentiating B cells while in the presence of CD40 ligand and interleukin-4. We further demonstrate the role of integrin ligand specificities in clustering of ß3 integrin and B cell receptor on the surface of differentiated B cells in 3D organoids as compared to the classic 2D cocultures. The study demonstrates that biomaterials-based immune organoids represent an ex vivo platform technology, which recapitulates certain aspects of GC biology to understand the process of B cell differentiation and induction of immunological responses. This platform is particularly useful in understanding the role of selective biomolecular signals and the temporal dependency of immune responses to these signals.

6.
Nat Protoc ; 12(1): 168-182, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28005068

RESUMO

Induction of B-cell immunity against infection depends on the initiation of the germinal center (GC) reaction in secondary lymphoid organs. Ex vivo recapitulation of the GC reaction in 2D cultures results in transient cell growth, with poor yield and short-term survival. Furthermore, no reported 2D ex vivo system can modulate the kinetics of a GC-like phenotype or the rate of antibody class switching. This protocol describes a methodology for developing immune organoids that partially mimic the B-cell zone of a lymphoid tissue, for efficient and rapid generation of B cells with a GC-like phenotype from naive murine B cells. The organoid is composed of a bioadhesive protein, gelatin, that is transformed into an ionically cross-linked hydrated network using biocompatible silicate nanoparticles (SiNPs). We explain how to establish the immune organoid culture to sustain immune cell proliferation and transformation into a GC-like phenotype. Starting with cell encapsulation in digested lymphoid tissues, clusters of proliferating B cells with a GC-like phenotype can be generated in the organoids at controlled rates, within ∼1 week. The culture methodology described here is currently the only one that allows the accelerated induction of a GC-like phenotype in B cells and supports a controllable immunoglobulin class-switching reaction. This method can be easily implemented in a typical tissue culture room by personnel with standard mammalian cell culture expertise.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/citologia , Linfócitos B/imunologia , Organoides/citologia , Engenharia Tecidual/métodos , Animais , Linfócitos B/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Gelatina/química , Gelatina/farmacologia , Centro Germinativo/citologia , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Organoides/efeitos dos fármacos , Fenótipo , Silicatos/química
7.
Adv Healthc Mater ; 5(12): 1413-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27100566

RESUMO

Soluble antigen-based cancer vaccines have poor retention in tissues along with suboptimal antigen processing by dendritic cells. Multiple booster doses are often needed, leading to dose-limiting systemic toxicity. A versatile, immunomodulatory, self-assembly protein nanogel vaccine is reported that induces robust immune cell response at lower antigen doses than soluble antigens, an important step towards biomaterials-based safer immunotherapy approaches.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Imunoterapia , Nanopartículas/química , Proteínas de Neoplasias , Neoplasias , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Humanos , Camundongos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Neoplasias/terapia
8.
Biomaterials ; 63: 24-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26072995

RESUMO

Ex vivo engineered three-dimensional organotypic cultures have enabled the real-time study and control of biological functioning of mammalian tissues. Organs of broad interest where its architectural, cellular, and molecular complexity has prevented progress in ex vivo engineering are the secondary immune organs. Ex vivo immune organs can enable mechanistic understanding of the immune system and more importantly, accelerate the translation of immunotherapies as well as a deeper understanding of the mechanisms that lead to their malignant transformation into a variety of B and T cell malignancies. However, till date, no modular ex vivo immune organ has been developed with an ability to control the rate of immune reaction through tunable design parameter. Here we describe a B cell follicle organoid made of nanocomposite biomaterials, which recapitulates the anatomical microenvironment of a lymphoid tissue that provides the basis to induce an accelerated germinal center (GC) reaction by continuously providing extracellular matrix (ECM) and cell-cell signals to naïve B cells. Compared to existing co-cultures, immune organoids provide a control over primary B cell proliferation with ∼100-fold higher and rapid differentiation to the GC phenotype with robust antibody class switching.


Assuntos
Linfócitos B/citologia , Materiais Biocompatíveis/química , Centro Germinativo/citologia , Nanocompostos/química , Organoides/citologia , Animais , Linfócitos B/imunologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Centro Germinativo/imunologia , Camundongos Endogâmicos C57BL , Nanocompostos/ultraestrutura , Técnicas de Cultura de Órgãos/métodos , Organoides/imunologia , Engenharia Tecidual/métodos
9.
Matrix Biol ; 41: 36-43, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25448408

RESUMO

The development of atherosclerosis involves phenotypic changes among vascular smooth muscle cells (VSMCs) that correlate with stiffening and remodeling of the extracellular matrix (ECM). VSMCs are highly sensitive to the composition and mechanical state of the surrounding ECM, and ECM remodeling during atherosclerosis likely contributes to pathology. We hypothesized that ECM mechanics and biochemistry are interdependent in their regulation of VSMC behavior and investigated the effect of ligand presentation on certain stiffness-mediated processes. Our findings demonstrate that substrate stiffening is not a unidirectional stimulus-instead, the influence of mechanics on cell behavior is highly conditioned on ligand biochemistry. This "stiffness-by-ligand" effect was evident for VSMC adhesion, spreading, cytoskeletal polymerization, and focal adhesion assembly, where VSMCs cultured on fibronectin (Fn)-modified substrates showed an augmented response to increasing stiffness, whereas cells on laminin (Ln) substrates showed a dampened response. By contrast, cells on Fn substrates showed a decrease in myosin light chain (MLC) phosphorylation and elongation with increasing stiffness, whereas Ln supported an increase in MLC phosphorylation and no change in cell shape with increasing stiffness. Taken together, these findings show that identical cell populations exhibit opposing responses to substrate stiffening depending on ECM presentation. Our results also suggest that the shift in VSMC phenotype in a developing atherosclerotic lesion is jointly regulated by stromal mechanics and biochemistry. This study highlights the complex influence of the blood vessel wall microenvironment on VSMC phenotype and provides insight into how cells may integrate ECM biochemistry and mechanics during normal and pathological tissue function.


Assuntos
Aorta/citologia , Matriz Extracelular/fisiologia , Mecanotransdução Celular , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Animais , Animais Recém-Nascidos , Aorta/fisiologia , Adesão Celular , Proliferação de Células , Células Cultivadas , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/citologia , Cadeias Leves de Miosina/metabolismo , Ratos
10.
Cell Mol Bioeng ; 7(3): 394-408, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25328548

RESUMO

Bioengineered hydrogels have been explored in cell and tissue engineering applications to support cell growth and modulate its behavior. A rationally designed scaffold should allow for encapsulated cells to survive, adhere, proliferate, remodel the niche, and can be used for controlled delivery of biomolecules. Here we report a microarray of composite bioadhesive microgels with modular dimensions, tunable mechanical properties and bulk modified adhesive biomolecule composition. Composite bioadhesive microgels of maleimide functionalized polyethylene glycol (PEG-MAL) with interpenetrating network (IPN) of gelatin ionically cross-linked with silicate nanoparticles were engineered by integrating microfabrication with Michael-type addition chemistry and ionic gelation. By encapsulating clinically relevant anchorage-dependent cervical cancer cells and suspension leukemia cells as cell culture models in these composite microgels, we demonstrate enhanced cell spreading, survival, and metabolic activity compared to control gels. The composite bioadhesive hydrogels represent a platform that could be used to study independent effect of stiffness and adhesive ligand density on cell survival and function. We envision that such microarrays of cell adhesive microenvironments, which do not require harsh chemical and UV crosslinking conditions, will provide a more efficacious cell culture platform that can be used to study cell behavior and survival, function as building blocks to fabricate 3D tissue structures, cell delivery systems, and high throughput drug screening devices.

11.
Acta Biomater ; 10(4): 1728-40, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24373907

RESUMO

Controlled modulation of immune response, especially the balance between immunostimulatory and immunosuppressive responses, is critical for a variety of clinical applications, including immunotherapies against cancer and infectious diseases, treatment of autoimmune disorders, transplant surgeries, regenerative medicine, prosthetic implants, etc. Our ability to precisely modify both innate and adaptive immune responses could provide new therapeutic directions in a variety of diseases. In the context of vaccines and immunotherapies, the interplay between antigen-presenting cells (e.g. dendritic cells and macrophages), B cells, T helper and killer subtypes, and regulatory T- and B-cell responses is critical for generating effective immunity against cancer, infectious diseases and autoimmune diseases. In recent years, immunoengineering has emerged as a new field that uses quantitative engineering tools to understand molecular-, cellular- and system-level interactions of the immune system and to develop design-driven approaches to control and modulate immune responses. Biomaterials are an integral part of this engineering toolbox and can exploit the intrinsic biological and mechanical cues of the immune system to directly modulate and train immune cells and direct their response to a particular phenotype. A large body of literature exists on strategies to evade or suppress the immune response in implants, transplantation and regenerative medicine. This review specifically focuses on the use of biomaterials for immunostimulation and controlled modulation, especially in the context of vaccines and immunotherapies against cancer, infectious diseases and autoimmune disorders. Bioengineering smart systems that can simultaneously deliver multiple bioactive agents in a controlled manner or can work as a niche for in situ priming and modulation of the immune system could significantly enhance the efficacy of next-generation immunotherapeutics. In this review, we describe our perspective on the important design aspects for the development of biomaterials that can actively modulate immune responses by stimulating receptor complexes and cells, and delivering multiple immunomodulatory biomolecules.


Assuntos
Bioengenharia/métodos , Imunomodulação , Vacinas/imunologia , Animais , Materiais Biocompatíveis , Apresentação Cruzada/efeitos dos fármacos , Humanos , Imunoterapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA