Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 392
Filtrar
1.
Nat Rev Mol Cell Biol ; 18(12): 728-742, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29115301

RESUMO

Stem cells and their local microenvironment, or niche, communicate through mechanical cues to regulate cell fate and cell behaviour and to guide developmental processes. During embryonic development, mechanical forces are involved in patterning and organogenesis. The physical environment of pluripotent stem cells regulates their self-renewal and differentiation. Mechanical and physical cues are also important in adult tissues, where adult stem cells require physical interactions with the extracellular matrix to maintain their potency. In vitro, synthetic models of the stem cell niche can be used to precisely control and manipulate the biophysical and biochemical properties of the stem cell microenvironment and to examine how the mode and magnitude of mechanical cues, such as matrix stiffness or applied forces, direct stem cell differentiation and function. Fundamental insights into the mechanobiology of stem cells also inform the design of artificial niches to support stem cells for regenerative therapies.


Assuntos
Células-Tronco Adultas/fisiologia , Matriz Extracelular/fisiologia , Organogênese/fisiologia , Regeneração/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco Adultas/citologia , Animais , Fenômenos Biomecânicos/fisiologia , Humanos
2.
Nature ; 606(7916): 992-998, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614223

RESUMO

Most cancer vaccines target peptide antigens, necessitating personalization owing to the vast inter-individual diversity in major histocompatibility complex (MHC) molecules that present peptides to T cells. Furthermore, tumours frequently escape T cell-mediated immunity through mechanisms that interfere with peptide presentation1. Here we report a cancer vaccine that induces a coordinated attack by diverse T cell and natural killer (NK) cell populations. The vaccine targets the MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor on T cells and NK cells, but tumours evade immune recognition by proteolytic MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance presentation of tumour antigens by dendritic cells to T cells and augment the cytotoxic function of NK cells. Notably, this vaccine maintains efficacy against MHC class I-deficient tumours resistant to cytotoxic T cells through the coordinated action of NK cells and CD4+ T cells. The vaccine is also efficacious in a clinically important setting: immunization following surgical removal of primary, highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine design enables protective immunity even against tumours with common escape mutations.


Assuntos
Síndromes Mielodisplásicas , Neoplasias , Dermatopatias Genéticas , Vacinas , Antígenos de Histocompatibilidade Classe I , Humanos , Células Matadoras Naturais , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias/prevenção & controle
3.
Nat Immunol ; 16(1): 75-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25347465

RESUMO

In lymph nodes, fibroblastic reticular cells (FRCs) form a collagen-based reticular network that supports migratory dendritic cells (DCs) and T cells and transports lymph. A hallmark of FRCs is their propensity to contract collagen, yet this function is poorly understood. Here we demonstrate that podoplanin (PDPN) regulates actomyosin contractility in FRCs. Under resting conditions, when FRCs are unlikely to encounter mature DCs expressing the PDPN receptor CLEC-2, PDPN endowed FRCs with contractile function and exerted tension within the reticulum. Upon inflammation, CLEC-2 on mature DCs potently attenuated PDPN-mediated contractility, which resulted in FRC relaxation and reduced tissue stiffness. Disrupting PDPN function altered the homeostasis and spacing of FRCs and T cells, which resulted in an expanded reticular network and enhanced immunity.


Assuntos
Colágeno/metabolismo , Fibroblastos/citologia , Lectinas Tipo C/metabolismo , Linfonodos/citologia , Glicoproteínas de Membrana/metabolismo , Amidas/farmacologia , Animais , Sobrevivência Celular/imunologia , Colágeno/imunologia , Citoesqueleto/imunologia , Citoesqueleto/ultraestrutura , Inibidores Enzimáticos/farmacologia , Feminino , Fibroblastos/imunologia , Fibroblastos/ultraestrutura , Lectinas Tipo C/imunologia , Linfonodos/imunologia , Linfonodos/ultraestrutura , Masculino , Glicoproteínas de Membrana/imunologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Fosforilação , Piridinas/farmacologia , Organismos Livres de Patógenos Específicos
4.
Proc Natl Acad Sci U S A ; 121(9): e2304643121, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38377210

RESUMO

Generating strong rapid adhesion between hydrogels has the potential to advance the capabilities of modern medicine and surgery. Current hydrogel adhesion technologies rely primarily on liquid-based diffusion mechanisms and the formation of covalent bonds, requiring prolonged time to generate adhesion. Here, we present a simple and versatile strategy using dry chitosan polymer films to generate instant adhesion between hydrogel-hydrogel and hydrogel-elastomer surfaces. Using this approach we can achieve extremely high adhesive energies (>3,000 J/m2), which are governed by pH change and non-covalent interactions including H-bonding, Van der Waals forces, and bridging polymer entanglement. Potential examples of biomedical applications are presented, including local tissue cooling, vascular sealing, prevention of surgical adhesions, and prevention of hydrogel dehydration. We expect these findings and the simplicity of this approach to have broad implications for adhesion strategies and hydrogel design.


Assuntos
Adesivos , Polímeros , Humanos , Aderências Teciduais/prevenção & controle , Adesivos/química , Elastômeros , Hidrogéis/química
5.
J Immunol ; 212(2): 179-187, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38166245

RESUMO

Therapeutic cancer vaccines offer the promise of stimulating the immune system to specifically eradicate tumor cells and establish long-term memory to prevent tumor recurrence. However, despite showing benign safety profiles and the ability to generate Ag-specific cellular responses, cancer vaccines have been hampered by modest clinical efficacy. Lessons learned from these studies have led to the emergence of innovative materials-based strategies that aim to boost the clinical activity of cancer vaccines. In this Brief Review, we provide an overview of the key elements needed for an effective vaccine-induced antitumor response, categorize current approaches to therapeutic cancer vaccination, and explore recent advances in materials-based strategies to potentiate cancer vaccines.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Neoplasias/prevenção & controle , Neoplasias/tratamento farmacológico , Vacinação
6.
Nature ; 584(7822): 535-546, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32848221

RESUMO

Substantial research over the past two decades has established that extracellular matrix (ECM) elasticity, or stiffness, affects fundamental cellular processes, including spreading, growth, proliferation, migration, differentiation and organoid formation. Linearly elastic polyacrylamide hydrogels and polydimethylsiloxane (PDMS) elastomers coated with ECM proteins are widely used to assess the role of stiffness, and results from such experiments are often assumed to reproduce the effect of the mechanical environment experienced by cells in vivo. However, tissues and ECMs are not linearly elastic materials-they exhibit far more complex mechanical behaviours, including viscoelasticity (a time-dependent response to loading or deformation), as well as mechanical plasticity and nonlinear elasticity. Here we review the complex mechanical behaviours of tissues and ECMs, discuss the effect of ECM viscoelasticity on cells, and describe the potential use of viscoelastic biomaterials in regenerative medicine. Recent work has revealed that matrix viscoelasticity regulates these same fundamental cell processes, and can promote behaviours that are not observed with elastic hydrogels in both two- and three-dimensional culture microenvironments. These findings have provided insights into cell-matrix interactions and how these interactions differentially modulate mechano-sensitive molecular pathways in cells. Moreover, these results suggest design guidelines for the next generation of biomaterials, with the goal of matching tissue and ECM mechanics for in vitro tissue models and applications in regenerative medicine.


Assuntos
Elasticidade , Matriz Extracelular/metabolismo , Substâncias Viscoelásticas , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Técnicas de Cultura de Células , Forma Celular , Matriz Extracelular/química , Humanos , Mecanotransdução Celular , Células-Tronco Mesenquimais/citologia , Modelos Biológicos , Medicina Regenerativa
7.
Proc Natl Acad Sci U S A ; 120(1): e2213222120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36577059

RESUMO

Adoptive T cell transfer (ACT) therapies suffer from a number of limitations (e.g., poor control of solid tumors), and while combining ACT with cytokine therapy can enhance effectiveness, this also results in significant side effects. Here, we describe a nanotechnology approach to improve the efficacy of ACT therapies by metabolically labeling T cells with unnatural sugar nanoparticles, allowing direct conjugation of antitumor cytokines onto the T cell surface during the manufacturing process. This allows local, concentrated activity of otherwise toxic cytokines. This approach increases T cell infiltration into solid tumors, activates the host immune system toward a Type 1 response, encourages antigen spreading, and improves control of aggressive solid tumors and achieves complete blood cancer regression with otherwise noncurative doses of CAR-T cells. Overall, this method provides an effective and easily integrated approach to the current ACT manufacturing process to increase efficacy in various settings.


Assuntos
Citocinas , Neoplasias , Humanos , Citocinas/metabolismo , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Neoplasias/patologia , Terapia Baseada em Transplante de Células e Tecidos
8.
FASEB J ; 38(1): e23321, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38031974

RESUMO

Bypass graft failure occurs in 20%-50% of coronary and lower extremity bypasses within the first-year due to intimal hyperplasia (IH). TSP-2 is a key regulatory protein that has been implicated in the development of IH following vessel injury. In this study, we developed a biodegradable CLICK-chemistry gelatin-based hydrogel to achieve sustained perivascular delivery of TSP-2 siRNA to rat carotid arteries following endothelial denudation injury. At 21 days, perivascular application of TSP-2 siRNA embedded hydrogels significantly downregulated TSP-2 gene expression, cellular proliferation, as well as other associated mediators of IH including MMP-9 and VEGF-R2, ultimately resulting in a significant decrease in IH. Our data illustrates the ability of perivascular CLICK-gelatin delivery of TSP-2 siRNA to mitigate IH following arterial injury.


Assuntos
Gelatina , Lesões do Sistema Vascular , Ratos , Animais , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Hiperplasia , Trombospondinas/genética , Proliferação de Células
9.
Chem Rev ; 123(2): 834-873, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35930422

RESUMO

Biomaterials with the ability to self-heal and recover their structural integrity offer many advantages for applications in biomedicine. The past decade has witnessed the rapid emergence of a new class of self-healing biomaterials commonly termed injectable, or printable in the context of 3D printing. These self-healing injectable biomaterials, mostly hydrogels and other soft condensed matter based on reversible chemistry, are able to temporarily fluidize under shear stress and subsequently recover their original mechanical properties. Self-healing injectable hydrogels offer distinct advantages compared to traditional biomaterials. Most notably, they can be administered in a locally targeted and minimally invasive manner through a narrow syringe without the need for invasive surgery. Their moldability allows for a patient-specific intervention and shows great prospects for personalized medicine. Injected hydrogels can facilitate tissue regeneration in multiple ways owing to their viscoelastic and diffusive nature, ranging from simple mechanical support, spatiotemporally controlled delivery of cells or therapeutics, to local recruitment and modulation of host cells to promote tissue regeneration. Consequently, self-healing injectable hydrogels have been at the forefront of many cutting-edge tissue regeneration strategies. This study provides a critical review of the current state of self-healing injectable hydrogels for tissue regeneration. As key challenges toward further maturation of this exciting research field, we identify (i) the trade-off between the self-healing and injectability of hydrogels vs their physical stability, (ii) the lack of consensus on rheological characterization and quantitative benchmarks for self-healing injectable hydrogels, particularly regarding the capillary flow in syringes, and (iii) practical limitations regarding translation toward therapeutically effective formulations for regeneration of specific tissues. Hence, here we (i) review chemical and physical design strategies for self-healing injectable hydrogels, (ii) provide a practical guide for their rheological analysis, and (iii) showcase their applicability for regeneration of various tissues and 3D printing of complex tissues and organoids.


Assuntos
Materiais Biocompatíveis , Hidrogéis , Humanos , Hidrogéis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/química , Engenharia Tecidual
10.
Proc Natl Acad Sci U S A ; 119(28): e2111003119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35787058

RESUMO

Immunotherapy has had a tremendous impact on cancer treatment in the past decade, with hitherto unseen responses at advanced and metastatic stages of the disease. However, the aggressive brain tumor glioblastoma (GBM) is highly immunosuppressive and remains largely refractory to current immunotherapeutic approaches. The stimulator of interferon genes (STING) DNA sensing pathway has emerged as a next-generation immunotherapy target with potent local immune stimulatory properties. Here, we investigated the status of the STING pathway in GBM and the modulation of the brain tumor microenvironment (TME) with the STING agonist ADU-S100. Our data reveal the presence of STING in human GBM specimens, where it stains strongly in the tumor vasculature. We show that human GBM explants can respond to STING agonist treatment by secretion of inflammatory cytokines. In murine GBM models, we show a profound shift in the tumor immune landscape after STING agonist treatment, with massive infiltration of the tumor-bearing hemisphere with innate immune cells including inflammatory macrophages, neutrophils, and natural killer (NK) populations. Treatment of established murine intracranial GL261 and CT-2A tumors by biodegradable ADU-S100-loaded intracranial implants demonstrated a significant increase in survival in both models and long-term survival with immune memory in GL261. Responses to treatment were abolished by NK cell depletion. This study reveals therapeutic potential and deep remodeling of the TME by STING activation in GBM and warrants further examination of STING agonists alone or in combination with other immunotherapies such as cancer vaccines, chimeric antigen receptor T cells, NK therapies, and immune checkpoint blockade.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Células Matadoras Naturais , Animais , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Humanos , Imunidade , Imunoterapia , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Microambiente Tumoral
11.
Nat Mater ; 22(2): 249-259, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36357687

RESUMO

While mechanical stimulation is known to regulate a wide range of biological processes at the cellular and tissue levels, its medical use for tissue regeneration and rehabilitation has been limited by the availability of suitable devices. Here we present a mechanically active gel-elastomer-nitinol tissue adhesive (MAGENTA) that generates and delivers muscle-contraction-mimicking stimulation to a target tissue with programmed strength and frequency. MAGENTA consists of a shape memory alloy spring that enables actuation up to 40% strain, and an adhesive that efficiently transmits the actuation to the underlying tissue. MAGENTA activates mechanosensing pathways involving yes-associated protein and myocardin-related transcription factor A, and increases the rate of muscle protein synthesis. Disuse muscles treated with MAGENTA exhibit greater size and weight, and generate higher forces compared to untreated muscles, demonstrating the prevention of atrophy. MAGENTA thus has promising applications in the treatment of muscle atrophy and regenerative medicine.


Assuntos
Músculo Esquelético , Adesivos Teciduais , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Adesivos Teciduais/metabolismo , Corantes de Rosanilina/metabolismo , Atrofia Muscular/prevenção & controle , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Contração Muscular
12.
Nat Mater ; 22(1): 117-127, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36456871

RESUMO

Biomolecular and physical cues of the extracellular matrix environment regulate collective cell dynamics and tissue patterning. Nonetheless, how the viscoelastic properties of the matrix regulate collective cell spatial and temporal organization is not fully understood. Here we show that the passive viscoelastic properties of the matrix encapsulating a spheroidal tissue of breast epithelial cells guide tissue proliferation in space and in time. Matrix viscoelasticity prompts symmetry breaking of the spheroid, leading to the formation of invading finger-like protrusions, YAP nuclear translocation and epithelial-to-mesenchymal transition both in vitro and in vivo in a Arp2/3-complex-dependent manner. Computational modelling of these observations allows us to establish a phase diagram relating morphological stability with matrix viscoelasticity, tissue viscosity, cell motility and cell division rate, which is experimentally validated by biochemical assays and in vitro experiments with an intestinal organoid. Altogether, this work highlights the role of stress relaxation mechanisms in tissue growth dynamics, a fundamental process in morphogenesis and oncogenesis.


Assuntos
Células Epiteliais , Matriz Extracelular , Viscosidade , Elasticidade
13.
PLoS Pathog ; 18(2): e1010240, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35143595

RESUMO

Staphylococcus aureus bacteremia (SAB) remains a clinically challenging infection despite extensive investigation. Repurposing medications approved for other indications is appealing as clinical safety profiles have already been established. Ticagrelor, a reversible adenosine diphosphate receptor antagonist that prevents platelet aggregation, is indicated for patients suffering from acute coronary syndrome (ACS). However, some clinical data suggest that patients treated with ticagrelor are less likely to have poor outcomes due to S. aureus infection. There are several potential mechanisms by which ticagrelor may affect S. aureus virulence. These include direct antibacterial activity, up-regulation of the innate immune system through boosting platelet-mediated S. aureus killing, and prevention of S. aureus adhesion to host tissues. In this Pearl, we review the clinical data surrounding ticagrelor and infection as well as explore the evidence surrounding these proposed mechanisms of action. While more evidence is needed before antiplatelet medications formally become part of the arsenal against S. aureus infection, these potential mechanisms represent exciting pathways to target in the host/pathogen interface.


Assuntos
Bacteriemia/tratamento farmacológico , Plaquetas/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Ticagrelor/uso terapêutico , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Inibidores da Agregação Plaquetária/uso terapêutico , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia
14.
Nat Immunol ; 13(5): 499-510, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22466668

RESUMO

Lymph node stromal cells (LNSCs) closely regulate immunity and self-tolerance, yet key aspects of their biology remain poorly elucidated. Here, comparative transcriptomic analyses of mouse LNSC subsets demonstrated the expression of important immune mediators, growth factors and previously unknown structural components. Pairwise analyses of ligands and cognate receptors across hematopoietic and stromal subsets suggested a complex web of crosstalk. Fibroblastic reticular cells (FRCs) showed enrichment for higher expression of genes relevant to cytokine signaling, relative to their expression in skin and thymic fibroblasts. LNSCs from inflamed lymph nodes upregulated expression of genes encoding chemokines and molecules involved in the acute-phase response and the antigen-processing and antigen-presentation machinery. Poorly studied podoplanin (gp38)-negative CD31(-) LNSCs showed similarities to FRCs but lacked expression of interleukin 7 (IL-7) and were identified as myofibroblastic pericytes that expressed integrin α(7). Together our data comprehensively describe the transcriptional characteristics of LNSC subsets.


Assuntos
Expressão Gênica/imunologia , Inflamação/imunologia , Linfonodos/imunologia , Células Estromais/imunologia , Células Estromais/metabolismo , Transcriptoma , Reação de Fase Aguda/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Fibroblastos/imunologia , Fibroblastos/metabolismo , Homeostase/imunologia , Inflamação/genética , Cadeias alfa de Integrinas/imunologia , Cadeias alfa de Integrinas/metabolismo , Interleucina-7/imunologia , Interleucina-7/metabolismo , Linfonodos/citologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/imunologia , Pericitos/metabolismo , Tolerância a Antígenos Próprios/imunologia , Análise Serial de Tecidos/métodos
15.
Nat Mater ; 21(8): 939-950, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35817965

RESUMO

Myelofibrosis is a progressive bone marrow malignancy associated with monocytosis, and is believed to promote the pathological remodelling of the extracellular matrix. Here we show that the mechanical properties of myelofibrosis, namely the liquid-to-solid properties (viscoelasticity) of the bone marrow, contribute to aberrant differentiation of monocytes. Human monocytes cultured in stiff, elastic hydrogels show proinflammatory polarization and differentiation towards dendritic cells, as opposed to those cultured in a viscoelastic matrix. This mechanically induced cell differentiation is blocked by inhibiting a myeloid-specific isoform of phosphoinositide 3-kinase, PI3K-γ. We further show that murine bone marrow with myelofibrosis has a significantly increased stiffness and unveil a positive correlation between myelofibrosis grading and viscoelasticity. Treatment with a PI3K-γ inhibitor in vivo reduced frequencies of monocyte and dendritic cell populations in murine bone marrow with myelofibrosis. Moreover, transcriptional changes driven by viscoelasticity are consistent with transcriptional profiles of myeloid cells in other human fibrotic diseases. These results demonstrate that a fibrotic bone marrow niche can physically promote a proinflammatory microenvironment.


Assuntos
Mielofibrose Primária , Animais , Medula Óssea/patologia , Diferenciação Celular , Fibrose , Humanos , Camundongos , Monócitos , Fosfatidilinositol 3-Quinases , Mielofibrose Primária/patologia
16.
Proc Natl Acad Sci U S A ; 117(42): 25999-26007, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33020289

RESUMO

Mammalian cell morphology has been linked to the viscoelastic properties of the adhesion substrate, which is particularly relevant in biological processes such as wound repair and embryonic development where cell spreading and migration are critical. Plastic deformation, degradation, and relaxation of stress are typically coupled in biomaterial systems used to explore these effects, making it unclear which variable drives cell behavior. Here we present a nondegradable polymer architecture that specifically decouples irreversible creep from stress relaxation and modulus. We demonstrate that network plasticity independently controls mesenchymal stem cell spreading through a biphasic relationship dependent on cell-intrinsic forces, and this relationship can be shifted by inhibiting actomyosin contractility. Kinetic Monte Carlo simulations also show strong correlation with experimental cell spreading data as a function of the extracellular matrix (ECM) plasticity. Furthermore, plasticity regulates many ECM adhesion and remodeling genes. Altogether, these findings confirm a key role for matrix plasticity in stem cell biophysics, and we anticipate this will have ramifications in the design of biomaterials to enhance therapeutic applications of stem cells.


Assuntos
Plasticidade Celular , Matriz Extracelular/química , Hidrogéis/química , Mecanotransdução Celular , Células-Tronco Mesenquimais/fisiologia , Polímeros/química , Estresse Mecânico , Alginatos/química , Adesão Celular , Técnicas de Cultura de Células , Humanos , Células-Tronco Mesenquimais/citologia , Reologia , Substâncias Viscoelásticas
17.
Cancer Immunol Immunother ; 71(12): 2957-2968, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35524791

RESUMO

Disruption of the tumor extracellular matrix (ECM) may alter immune cell infiltration into the tumor and antitumor T cell priming in the tumor-draining lymph nodes (tdLNs). Here, we explore how intratumoral enzyme treatment (ET) of B16 melanoma tumors with ECM-depleting enzyme hyaluronidase alters adaptive and innate immune populations, including T cells, DCs, and macrophages, in the tumors and tdLNs. ET increased CD103+ DC abundance in the tdLNs, as well as antigen presentation of a model tumor antigen ovalbumin (OVA), eliciting local OVA-specific CD8+ T cell responses. Delivered in combination with a distant cryogel-based cancer vaccine, ET increased the systemic antigen-specific CD8+ T cell response. By enhancing activity within the tdLN, ET may broadly support immunotherapies in generating tumor-specific immunity.


Assuntos
Vacinas Anticâncer , Melanoma Experimental , Animais , Humanos , Ovalbumina , Células Dendríticas , Hialuronoglucosaminidase , Criogéis , Antígenos de Neoplasias , Linfonodos , Matriz Extracelular
18.
Nat Mater ; 20(11): 1469-1479, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34226688

RESUMO

Cancer nanomedicines were initially envisioned as magic bullets, travelling through the circulation to target tumours while sparing healthy tissues the toxicity of classic chemotherapy. While a limited number of nanomedicine therapies have resulted, the disappointing news is that major obstacles were overlooked in the nanoparticle's journey. However, some of these challenges may be turned into opportunities. Here, we discuss biological barriers to cancer nanomedicines and elaborate on two directions that the field is currently exploring to meet its initial expectations. The first strategy entails re-engineering cancer nanomedicines to prevent undesired interactions en route to the tumour. The second aims instead to leverage these obstacles into out-of-the-box diagnostic and therapeutic applications of nanomedicines, for cancer and beyond. Both paths require, among other developments, a deeper understanding of nano-bio interactions. We offer a forward look at how classic cancer nanomedicine may overcome its limitations while contributing to other areas of research.


Assuntos
Nanopartículas , Neoplasias , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Nanomedicina/métodos , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico
19.
Langmuir ; 38(18): 5603-5616, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35446569

RESUMO

Nanoparticle-based delivery of therapeutics to the brain has had limited clinical impact due to challenges crossing the blood-brain barrier (BBB). Certain cells, such as monocytes, possess the ability to migrate across the BBB, making them attractive candidates for cell-based brain delivery strategies. In this work, we explore nanoparticle design parameters that impact both monocyte association and monocyte-mediated BBB transport. We use electrohydrodynamic jetting to prepare nanoparticles of varying sizes, compositions, and elasticity to address their impact on uptake by THP-1 monocytes and permeation across the BBB. An in vitro human BBB model is developed using human cerebral microvascular endothelial cells (hCMEC/D3) for the assessment of migration. We compare monocyte uptake of both polymeric and synthetic protein nanoparticles (SPNPs) of various sizes, as well as their effect on cell migration. SPNPs (human serum albumin/HSA or human transferrin/TF) are shown to promote increased monocyte-mediated transport across the BBB over polymeric nanoparticles. TF SPNPs (200 nm) associate readily, with an average uptake of 138 particles/cell. Nanoparticle loading is shown to influence the migration of THP-1 monocytes. The migration of monocytes loaded with 200 nm TF and 200 nm HSA SPNPs was 2.3-fold and 2.1-fold higher than that of an untreated control. RNA-seq analysis after TF SPNP treatment suggests that the upregulation of several migration genes may be implicated in increased monocyte migration (ex. integrin subunits α M and α L). Integrin ß 2 chain combines with either integrin subunit α M chain or integrin subunit α L chain to form macrophage antigen 1 and lymphocyte function-associated antigen 1 integrins. Both products play a pivotal role in the transendothelial migration cascade. Our findings highlight the potential of SPNPs as drug and/or gene delivery platforms for monocyte-mediated BBB transport, especially where conventional polymer nanoparticles are ineffective or otherwise not desirable.


Assuntos
Monócitos , Nanopartículas , Células Endoteliais/metabolismo , Humanos , Integrinas/metabolismo , Migração Transendotelial e Transepitelial , Transferrina/metabolismo
20.
Proc Natl Acad Sci U S A ; 116(31): 15392-15397, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31311862

RESUMO

Mesenchymal stem cell (MSC) therapies demonstrate particular promise in ameliorating diseases of immune dysregulation but are hampered by short in vivo cell persistence and inconsistencies in phenotype. Here, we demonstrate that biomaterial encapsulation into alginate using a microfluidic device could substantially increase in vivo MSC persistence after intravenous (i.v.) injection. A combination of cell cluster formation and subsequent cross-linking with polylysine led to an increase in injected MSC half-life by more than an order of magnitude. These modifications extended persistence even in the presence of innate and adaptive immunity-mediated clearance. Licensing of encapsulated MSCs with inflammatory cytokine pretransplantation increased expression of immunomodulatory-associated genes, and licensed encapsulates promoted repopulation of recipient blood and bone marrow with allogeneic donor cells after sublethal irradiation by a ∼2-fold increase. The ability of microgel encapsulation to sustain MSC survival and increase overall immunomodulatory capacity may be applicable for improving MSC therapies in general.


Assuntos
Encapsulamento de Células , Imunomodulação , Células-Tronco Mesenquimais/citologia , Alginatos/química , Animais , Células Cultivadas , Regulação da Expressão Gênica , Hematopoese/genética , Imunidade , Imunomodulação/genética , Camundongos Endogâmicos BALB C , Fatores de Tempo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa