Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(6): 114297, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38824643

RESUMO

The mechanical environment generated through the adhesive interaction of endothelial cells (ECs) with the matrix controls nuclear tension, preventing aberrant gene synthesis and the transition from restrictive to leaky endothelium, a hallmark of acute lung injury (ALI). However, the mechanisms controlling tension transmission to the nucleus and EC-restrictive fate remain elusive. Here, we demonstrate that, in a kinase-independent manner, focal adhesion kinase (FAK) safeguards tension transmission to the nucleus to maintain EC-restrictive fate. In FAK-depleted ECs, robust activation of the RhoA-Rho-kinase pathway increased EC tension and phosphorylation of the nuclear envelope protein, emerin, activating DNMT3a. Activated DNMT3a methylates the KLF2 promoter, impairing the synthesis of KLF2 and its target S1PR1 to induce the leaky EC transcriptome. Repleting FAK (wild type or kinase dead) or inhibiting RhoA-emerin-DNMT3a activities in damaged lung ECs restored KLF2 transcription of the restrictive EC transcriptome. Thus, FAK sensing and control of tension transmission to the nucleus govern restrictive endothelium to maintain lung homeostasis.


Assuntos
Núcleo Celular , Células Endoteliais , Fatores de Transcrição Kruppel-Like , Transcriptoma , Proteína rhoA de Ligação ao GTP , Humanos , Núcleo Celular/metabolismo , Transcriptoma/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Células Endoteliais/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , DNA Metiltransferase 3A , Quinase 1 de Adesão Focal/metabolismo , Quinase 1 de Adesão Focal/genética , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Regiões Promotoras Genéticas/genética , Fosforilação
2.
Adv Sci (Weinh) ; 10(3): e2206014, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36453581

RESUMO

Various signals in tissue microenvironments are often unevenly distributed around cells. Cellular responses to asymmetric cell-matrix adhesion in a 3D space remain generally unclear and are to be studied at the single-cell resolution. Here, the authors developed a droplet-based microfluidic approach to manufacture a pure population of single cells in a microscale layer of compartmentalized 3D hydrogel matrices with a tunable spatial presentation of ligands at the subcellular level. Cells elongate with an asymmetric presentation of the integrin adhesion ligand Arg-Gly-Asp (RGD), while cells expand isotropically with a symmetric presentation of RGD. Membrane tension is higher on the side of single cells interacting with RGD than on the side without RGD. Finite element analysis shows that a non-uniform isotropic cell volume expansion model is sufficient to recapitulate the experimental results. At a longer timescale, asymmetric ligand presentation commits mesenchymal stem cells to the osteogenic lineage. Cdc42 is an essential mediator of cell polarization and lineage specification in response to asymmetric cell-matrix adhesion. This study highlights the utility of precisely controlling 3D ligand presentation around single cells to direct cell polarity for regenerative engineering and medicine.


Assuntos
Encapsulamento de Células , Polaridade Celular , Ligantes , Hidrogéis , Oligopeptídeos
3.
Nat Rev Mater ; 8(6): 390-402, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38463907

RESUMO

The extracellular matrix in microenvironments harbors a variety of signals to control cellular functions and the materiality of tissues. Most efforts to synthetically reconstitute the matrix by biomaterial design have focused on decoupling cell-secreted and polymer-based cues. Cells package molecules into nanoscale lipid membrane-bound extracellular vesicles and secrete them. Thus, extracellular vesicles inherently interact with the meshwork of the extracellular matrix. In this Review, we discuss various aspects of extracellular vesicle-matrix interactions. Cells receive feedback from the extracellular matrix and leverage intracellular processes to control the biogenesis of extracellular vesicles. Once secreted, various biomolecular and biophysical factors determine whether extracellular vesicles are locally incorporated into the matrix or transported out of the matrix to be taken up by other cells or deposited into tissues at a distal location. These insights can be utilized to develop engineered biomaterials where EV release and retention can be precisely controlled in host tissue to elicit various biological and therapeutic outcomes.

4.
ACS Nano ; 15(11): 17439-17452, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34677951

RESUMO

Extracellular vesicles (EVs) are cell-secreted particles with broad potential to treat tissue injuries by delivering cargo to program target cells. However, improving the yield of functional EVs on a per cell basis remains challenging due to an incomplete understanding of how microenvironmental cues regulate EV secretion at the nanoscale. We show that mesenchymal stromal cells (MSCs) seeded on engineered hydrogels that mimic the elasticity of soft tissues with a lower integrin ligand density secrete ∼10-fold more EVs per cell than MSCs seeded on a rigid plastic substrate, without compromising their therapeutic activity or cargo to resolve acute lung injury in mice. Mechanistically, intracellular CD63+ multivesicular bodies (MVBs) transport faster within MSCs on softer hydrogels, leading to an increased frequency of MVB fusion with the plasma membrane to secrete more EVs. Actin-related protein 2/3 complex but not myosin-II limits MVB transport and EV secretion from MSCs on hydrogels. The results provide a rational basis for biomaterial design to improve EV secretion while maintaining their functionality.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Camundongos , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Comunicação Celular , Transporte Biológico , Hidrogéis/farmacologia , Hidrogéis/metabolismo
5.
Acta Biomater ; 133: 126-138, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34365041

RESUMO

Hydrogels have been used to design synthetic matrices that capture salient features of matrix microenvironments to study and control cellular functions. Recent advances in understanding of both extracellular matrix biology and biomaterial design have shown that biophysical cues are powerful mediators of cell biology, especially that of mesenchymal stromal cells (MSCs). MSCs have been tested in many clinical trials because of their ability to modulate immune cells in different pathological conditions. While roles of biophysical cues in MSC biology have been studied in the context of multilineage differentiation, their significance in regulating immunomodulatory functions of MSCs is just beginning to be elucidated. This review first describes design principles behind how biophysical cues in native microenvironments influence the ability of MSCs to regulate immune cell production and functions. We will then discuss how biophysical cues can be leveraged to optimize cell isolation, priming, and delivery, which can help improve the success of MSC therapy for immunomodulation. Finally, a perspective is presented on how implementing biophysical cues in MSC potency assay can be important in predicting clinical outcomes. STATEMENT OF SIGNIFICANCE: Stromal cells of mesenchymal origin are known to direct immune cell functions in vivo by secreting paracrine mediators. This property has been leveraged in developing mesenchymal stromal cell (MSC)-based therapeutics by adoptive transfer to treat immunological rejection and tissue injuries, which have been tested in over one thousand clinical trials to date, but with mixed success. Advances in biomaterial design have enabled precise control of biophysical cues based on how stromal cells interact with the extracellular matrix in microenvironments in situ. Investigators have begun to use this approach to understand how different matrix biophysical parameters, such as fiber orientation, porosity, dimensionality, and viscoelasticity impact stromal cell-mediated immunomodulation. The insights gained from this effort can potentially be used to precisely define the microenvironmental cues for isolation, priming, and delivery of MSCs, which can be tailored based on different disease indications for optimal therapeutic outcomes.


Assuntos
Células-Tronco Mesenquimais , Diferenciação Celular , Sinais (Psicologia) , Matriz Extracelular , Imunomodulação
6.
Adv Sci (Weinh) ; 7(20): 2001066, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33101850

RESUMO

Advances in engineered hydrogels reveal how cells sense and respond to 3D biophysical cues. However, most studies rely on interfacing a population of cells in a tissue-scale bulk hydrogel, an approach that overlooks the heterogeneity of local matrix deposition around individual cells. A droplet microfluidic technique to deposit a defined amount of 3D hydrogel matrices around single cells independently of material composition, elasticity, and stress relaxation times is developed. Mesenchymal stem cells (MSCs) undergo isotropic volume expansion more rapidly in thinner gels that present an Arg-Gly-Asp integrin ligand. Mathematical modeling and experiments show that MSCs experience higher membrane tension as they expand in thinner gels. Furthermore, thinner gels facilitate osteogenic differentiation of MSCs. By modulating ion channels, it is shown that isotropic volume expansion of single cells predicts intracellular tension and stem cell fate. The results suggest the utility of precise microscale gel deposition to control single cell functions.

7.
Adv Biosyst ; 4(11): e2000012, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33053274

RESUMO

The extracellular matrix varies considerably in mechanical properties at the microscale. It remains unclear how cells respond to these properties, in part, due to lack of tools to create precisely defined microenvironments in a discrete manner. Here, freeform stereolithography is leveraged to control the placement and elastic modulus of individual hydrogel microposts that serve as discrete matrix signals to interface with cells. Mesenchymal stromal cells (MSCs) located in the interstitial spaces between microposts above a base layer are analyzed. Cell volume is higher when MSCs interact with more microposts. MSCs show higher strain energy when they interact simultaneously with 4-kPa and 20-kPa microposts than with mechanically homogeneous micropost arrays. MSCs are sensitive to pharmacological inhibition of Rho-associated protein kinase in 4-kPa arrays, but resistant when presented together with 20-kPa arrays. Yes-associated protein (YAP) activity increases with higher cell volume and elastic modulus of microposts. Surprisingly, YAP activity becomes less variable with higher cell volume and decreases with higher average force and strain energy per post when MSCs interact with both 4-kPa and 20-kPa microposts simultaneously. Together, these results describe a material system for systematically investigating how the placement and intrinsic properties of discrete matrix signals impact cell volume and mechanotransduction.


Assuntos
Tamanho Celular , Técnicas Citológicas/instrumentação , Hidrogéis/química , Mecanotransdução Celular/fisiologia , Células Cultivadas , Módulo de Elasticidade , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Estereolitografia
8.
Sci Adv ; 6(15): eaaw0158, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32284989

RESUMO

Mesenchymal stromal cells (MSCs) modulate immune cells to ameliorate multiple inflammatory pathologies. Biophysical signals that regulate this process are poorly defined. By engineering hydrogels with tunable biophysical parameters relevant to bone marrow where MSCs naturally reside, we show that soft extracellular matrix maximizes the ability of MSCs to produce paracrine factors that have been implicated in monocyte production and chemotaxis upon inflammatory stimulation by tumor necrosis factor-α (TNFα). Soft matrix increases clustering of TNF receptors, thereby enhancing NF-κB activation and downstream gene expression. Actin polymerization and lipid rafts, but not myosin-II contractility, regulate mechanosensitive activation of MSCs by TNFα. We functionally demonstrate that human MSCs primed with TNFα in soft matrix enhance production of human monocytes in marrow of xenografted mice and increase trafficking of monocytes via CCL2. The results suggest the importance of biophysical signaling in tuning inflammatory activation of stromal cells to control the innate immune system.


Assuntos
Matriz Extracelular/metabolismo , Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Monócitos/metabolismo , Actinas/metabolismo , Animais , Biomarcadores , Movimento Celular/imunologia , Células Cultivadas , Quimiotaxia , Citocinas/metabolismo , Humanos , Inflamação/etiologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos , Camundongos , Monócitos/imunologia , NF-kappa B/metabolismo , Ligação Proteica , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
9.
Nat Nanotechnol ; 15(3): 217-223, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32066904

RESUMO

Cells release extracellular vesicles (EVs) to communicate over long distances, which requires EVs to traverse the extracellular matrix (ECM). However, given that the size of EVs is usually larger than the mesh size of the ECM, it is not clear how they can travel through the dense ECM. Here we show that, in contrast to synthetic nanoparticles, EVs readily transport through nanoporous ECM. Using engineered hydrogels, we demonstrate that the mechanical properties of the matrix regulate anomalous EV transport under confinement. Matrix stress relaxation allows EVs to overcome the confinement, and a higher crosslinking density facilitates a fluctuating transport motion through the polymer mesh, which leads to free diffusion and fast transport. Furthermore, water permeation through aquaporin-1 mediates the EV deformability, which further supports EV transport in hydrogels and a decellularized matrix. Our results provide evidence for the nature of EV transport within confined environments and demonstrate an unexpected dependence on matrix mechanics and water permeation.


Assuntos
Matriz Extracelular/química , Vesículas Extracelulares/metabolismo , Água/metabolismo , Animais , Aquaporina 1/metabolismo , Transporte Biológico , Fenômenos Biomecânicos , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Hidrogéis/química , Camundongos , Permeabilidade , Alicerces Teciduais/química
10.
Artigo em Inglês | MEDLINE | ID: mdl-31649928

RESUMO

A primary goal in tissue engineering is to develop functional tissues by recapitulating salient features of complex biological systems that exhibit a diverse range of physical forces. Induced pluripotent stem cells (iPSCs) are promising autologous cell sources to execute these developmental programs and their functions; however, cells require an extracellular environment where they will sense and respond to mechanical forces. Thus, understanding the biophysical relationships between stem cells and their extracellular environments will improve the ability to design complex biological systems through tissue engineering. This article first describes how the mechanical properties of the environment are important determinants of developmental processes, and then further details how biomaterials can be designed to precisely control the mechanics of cell-matrix interactions in order to study and define their reprogramming, self-renewal, differentiation, and morphogenesis. Finally, a perspective is presented on how insights from the mechanics of cell-matrix interactions can be leveraged to control pluripotent stem cells for tissue engineering applications.

11.
Methods Mol Biol ; 1740: 109-124, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29388139

RESUMO

Extracellular vesicles (EVs) are secreted nanoscale particles that transfer biomolecular cargo between cells in multicellular organisms. EVs play a variety of roles in intercellular communication and are being explored as potential vehicles for delivery of therapeutic biomolecules. However, EVs are highly heterogeneous in composition and biogenesis route, and this poses substantial challenges for understanding the role of EVs in biology and for harnessing these mechanisms for therapeutic applications, for which purifying therapeutic EVs from mixed EV populations may be necessary. Currently, technologies for isolating EV subsets are limited by overlapping physical properties among EV subsets. To meet this need, here we report an affinity chromatography-based method for enriching a specific EV subset from a heterogeneous EV starting population. By displaying an affinity tagged protein (tag-protein) on the EV surface, tagged EVs may be specifically isolated using simple affinity chromatography. Moreover, recovered EVs are enriched in the tag-protein relative to the starting population of EVs and relative to EVs purified from cell culture supernatant by standard differential centrifugation. Furthermore, chromatographically enriched EVs confer enhanced delivery of a cargo protein to recipient cells (via enhancing the amount of cargo protein per EV) relative to EVs isolated by centrifugation. Altogether, affinity chromatographic enrichment of EV subsets is a viable and facile strategy for investigating EV biology and for harnessing EVs for therapeutic applications.


Assuntos
Cromatografia de Afinidade/métodos , Líquido Extracelular , Vesículas Extracelulares , Animais , Células Cultivadas , Humanos
12.
APL Bioeng ; 2(3): 031802, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31069313

RESUMO

It is increasingly appreciated that physical forces play important roles in cancer biology, in terms of progression, invasiveness, and drug resistance. Clinical progress in treating hematological malignancy and in developing cancer immunotherapy highlights the role of the hematopoietic system as a key model in devising new therapeutic strategies against cancer. Understanding mechanobiology of the hematopoietic system in the context of cancer will thus yield valuable fundamental insights that can information about novel cancer therapeutics. In this perspective, biophysical insights related to blood cancer are defined and detailed. The interactions with immune cells relevant to immunotherapy against cancer are considered and expounded, followed by speculation of potential regulatory roles of mesenchymal stromal cells (MSCs) in this complex network. Finally, a perspective is presented as to how insights from these complex interactions between matrices, blood cancer cells, immune cells, and MSCs can be leveraged to influence and engineer the treatment of blood cancers in the clinic.

13.
Biotechnol Bioeng ; 113(5): 944-52, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26479709

RESUMO

Chemicals with aldehyde moieties are useful in the synthesis of polymerization reagents, pharmaceuticals, pesticides, flavors, and fragrances because of their high reactivity. However, chemical synthesis of aldehydes from carboxylic acids has unfavorable thermodynamics and limited specificity. Enzymatically catalyzed reductive bioaldehyde synthesis is an attractive route that overcomes unfavorable thermodynamics by ATP hydrolysis in ambient, aqueous conditions. Carboxylic acid reductases (Cars) are particularly attractive, as only one enzyme is required. We sought to increase the knowledge base of permitted substrates for four Cars. Additionally, the Lys2 enzyme family was found to be mechanistically the same as Cars and two isozymes were also tested. Our results show that Cars prefer molecules where the carboxylic acid is the only polar/charged group. Using this data and other published data, we develop a support vector classifier (SVC) for predicting Car reactivity and make predictions on all carboxylic acid metabolites in iAF1260 and Model SEED.


Assuntos
Aldeídos/metabolismo , Ácidos Carboxílicos/metabolismo , Mycobacterium/enzimologia , Nocardia/enzimologia , Oxirredutases/metabolismo , Simulação por Computador , Microbiologia Industrial/métodos , Modelos Biológicos , NADP/metabolismo , Oxirredução , Especificidade por Substrato , Máquina de Vetores de Suporte , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...