Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
1.
Cell ; 164(6): 1105-1109, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26967278

RESUMO

While studies of cultured cells have led to new insights into biological control, greater understanding of human pathophysiology requires the development of experimental systems that permit analysis of intercellular communications and tissue-tissue interactions in a more relevant organ context. Human organs-on-chips offer a potentially powerful new approach to confront this long-standing problem.


Assuntos
Técnicas Analíticas Microfluídicas/métodos , Técnicas de Cultura de Órgãos , Engenharia Tecidual/métodos , Barreira Hematoencefálica , Humanos , Neoplasias/fisiopatologia
2.
Nat Rev Genet ; 23(8): 467-491, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35338360

RESUMO

The failure of animal models to predict therapeutic responses in humans is a major problem that also brings into question their use for basic research. Organ-on-a-chip (organ chip) microfluidic devices lined with living cells cultured under fluid flow can recapitulate organ-level physiology and pathophysiology with high fidelity. Here, I review how single and multiple human organ chip systems have been used to model complex diseases and rare genetic disorders, to study host-microbiome interactions, to recapitulate whole-body inter-organ physiology and to reproduce human clinical responses to drugs, radiation, toxins and infectious pathogens. I also address the challenges that must be overcome for organ chips to be accepted by the pharmaceutical industry and regulatory agencies, as well as discuss recent advances in the field. It is evident that the use of human organ chips instead of animal models for drug development and as living avatars for personalized medicine is ever closer to realization.


Assuntos
Dispositivos Lab-On-A-Chip , Medicina de Precisão , Animais , Desenvolvimento de Medicamentos , Humanos , Doenças Raras
3.
Annu Rev Cell Dev Biol ; 29: 27-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24099083

RESUMO

Morphogenesis is the remarkable process by which cells self-assemble into complex tissues and organs that exhibit specialized form and function during embryological development. Many of the genes and chemical cues that mediate tissue and organ formation have been identified; however, these signals alone are not sufficient to explain how tissues and organs are constructed that exhibit their unique material properties and three-dimensional forms. Here, we review work that has revealed the central role that physical forces and extracellular matrix mechanics play in the control of cell fate switching, pattern formation, and tissue development in the embryo and how these same mechanical signals contribute to tissue homeostasis and developmental control throughout adult life.


Assuntos
Fenômenos Biomecânicos , Desenvolvimento Embrionário , Animais , Citoesqueleto , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Morfogênese , Transdução de Sinais
4.
Angiogenesis ; 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38969874

RESUMO

The development of reliable methods for producing functional endothelial cells (ECs) is crucial for progress in vascular biology and regenerative medicine. In this study, we present a streamlined and efficient methodology for the differentiation of human induced pluripotent stem cells (iPSCs) into induced ECs (iECs) that maintain the ability to undergo vasculogenesis in vitro and in vivo using a doxycycline-inducible system for the transient expression of the ETV2 transcription factor. This approach mitigates the limitations of direct transfection methods, such as mRNA-mediated differentiation, by simplifying the protocol and enhancing reproducibility across different stem cell lines. We detail the generation of iPSCs engineered for doxycycline-induced ETV2 expression and their subsequent differentiation into iECs, achieving over 90% efficiency within four days. Through both in vitro and in vivo assays, the functionality and phenotypic stability of the derived iECs were rigorously validated. Notably, these cells exhibit key endothelial markers and capabilities, including the formation of vascular networks in a microphysiological platform in vitro and in a subcutaneous mouse model. Furthermore, our results reveal a close transcriptional and proteomic alignment between the iECs generated via our method and primary ECs, confirming the biological relevance of the differentiated cells. The high efficiency and effectiveness of our induction methodology pave the way for broader application and accessibility of iPSC-derived ECs in scientific research, offering a valuable tool for investigating endothelial biology and for the development of EC-based therapies.

5.
PLoS Comput Biol ; 19(5): e1011050, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37146076

RESUMO

Drug repurposing requires distinguishing established drug class targets from novel molecule-specific mechanisms and rapidly derisking their therapeutic potential in a time-critical manner, particularly in a pandemic scenario. In response to the challenge to rapidly identify treatment options for COVID-19, several studies reported that statins, as a drug class, reduce mortality in these patients. However, it is unknown if different statins exhibit consistent function or may have varying therapeutic benefit. A Bayesian network tool was used to predict drugs that shift the host transcriptomic response to SARS-CoV-2 infection towards a healthy state. Drugs were predicted using 14 RNA-sequencing datasets from 72 autopsy tissues and 465 COVID-19 patient samples or from cultured human cells and organoids infected with SARS-CoV-2. Top drug predictions included statins, which were then assessed using electronic medical records containing over 4,000 COVID-19 patients on statins to determine mortality risk in patients prescribed specific statins versus untreated matched controls. The same drugs were tested in Vero E6 cells infected with SARS-CoV-2 and human endothelial cells infected with a related OC43 coronavirus. Simvastatin was among the most highly predicted compounds (14/14 datasets) and five other statins, including atorvastatin, were predicted to be active in > 50% of analyses. Analysis of the clinical database revealed that reduced mortality risk was only observed in COVID-19 patients prescribed a subset of statins, including simvastatin and atorvastatin. In vitro testing of SARS-CoV-2 infected cells revealed simvastatin to be a potent direct inhibitor whereas most other statins were less effective. Simvastatin also inhibited OC43 infection and reduced cytokine production in endothelial cells. Statins may differ in their ability to sustain the lives of COVID-19 patients despite having a shared drug target and lipid-modifying mechanism of action. These findings highlight the value of target-agnostic drug prediction coupled with patient databases to identify and clinically evaluate non-obvious mechanisms and derisk and accelerate drug repurposing opportunities.


Assuntos
COVID-19 , Inibidores de Hidroximetilglutaril-CoA Redutases , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , SARS-CoV-2 , Atorvastatina/farmacologia , Teorema de Bayes , Células Endoteliais , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Reposicionamento de Medicamentos , Prontuários Médicos
6.
Biochem J ; 480(4): 243-257, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36821520

RESUMO

The field of mechanobiology, which focuses on the key role that physical forces play in control of biological systems, has grown enormously over the past few decades. Here, I provide a brief personal perspective on the development of the tensegrity theory that contributed to the emergence of the mechanobiology field, the key role that crossing disciplines has played in its development, and how it has matured over time. I also describe how pursuing questions relating to mechanochemical transduction and mechanoregulation can lead to the creation of novel technologies and open paths for development of new therapeutic strategies for a broad range of diseases and disorders.


Assuntos
Mecanotransdução Celular , Humanos , Biofísica
7.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33879614

RESUMO

The de novo design of polar protein-protein interactions is challenging because of the thermodynamic cost of stripping water away from the polar groups. Here, we describe a general approach for designing proteins which complement exposed polar backbone groups at the edge of beta sheets with geometrically matched beta strands. We used this approach to computationally design small proteins that bind to an exposed beta sheet on the human transferrin receptor (hTfR), which shuttles interacting proteins across the blood-brain barrier (BBB), opening up avenues for drug delivery into the brain. We describe a design which binds hTfR with a 20 nM Kd, is hyperstable, and crosses an in vitro microfluidic organ-on-a-chip model of the human BBB. Our design approach provides a general strategy for creating binders to protein targets with exposed surface beta edge strands.


Assuntos
Engenharia de Proteínas/métodos , Receptores da Transferrina/metabolismo , Receptores da Transferrina/fisiologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Proteínas/metabolismo , Transferrina/metabolismo
8.
J Infect Dis ; 228(Suppl 5): S337-S354, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37669225

RESUMO

The National Center for Advancing Translational Sciences (NCATS) Assay Guidance Manual (AGM) Workshop on 3D Tissue Models for Antiviral Drug Development, held virtually on 7-8 June 2022, provided comprehensive coverage of critical concepts intended to help scientists establish robust, reproducible, and scalable 3D tissue models to study viruses with pandemic potential. This workshop was organized by NCATS, the National Institute of Allergy and Infectious Diseases, and the Bill and Melinda Gates Foundation. During the workshop, scientific experts from academia, industry, and government provided an overview of 3D tissue models' utility and limitations, use of existing 3D tissue models for antiviral drug development, practical advice, best practices, and case studies about the application of available 3D tissue models to infectious disease modeling. This report includes a summary of each workshop session as well as a discussion of perspectives and challenges related to the use of 3D tissues in antiviral drug discovery.


Assuntos
Antivirais , Descoberta de Drogas , Antivirais/farmacologia , Antivirais/uso terapêutico , Bioensaio
9.
Physiology (Bethesda) ; 37(5): 0, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35658627

RESUMO

The intertwined relationship between structure and function has been key to understanding human organ physiology and disease pathogenesis. An organ-on-a-chip (organ chip) is a bioengineered microfluidic cell culture device lined by living cells and tissues that recapitulates organ-level functions in vitro. This is accomplished by recreating organ-specific tissue-tissue interfaces and microenvironmental biochemical and mechanical cues while providing dynamic perfusion through endothelium-lined vascular channels. In this review, we discuss how this emerging technology has contributed to the understanding of human lung structure-function relationships at the cell, tissue, and organ levels.


Assuntos
Técnicas de Cultura de Células , Dispositivos Lab-On-A-Chip , Células Endoteliais , Humanos , Pulmão
10.
J Cell Sci ; 133(20)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32989042

RESUMO

One of the most rapid (less than 4 ms) transmembrane cellular mechanotransduction events involves activation of transient receptor potential vanilloid 4 (TRPV4) ion channels by mechanical forces transmitted across cell surface ß1 integrin receptors on endothelial cells, and the transmembrane solute carrier family 3 member 2 (herein denoted CD98hc, also known as SLC3A2) protein has been implicated in this response. Here, we show that ß1 integrin, CD98hc and TRPV4 all tightly associate and colocalize in focal adhesions where mechanochemical conversion takes place. CD98hc knockdown inhibits TRPV4-mediated calcium influx induced by mechanical forces, but not by chemical activators, thus confirming the mechanospecificity of this signaling response. Molecular analysis reveals that forces applied to ß1 integrin must be transmitted from its cytoplasmic C terminus via the CD98hc cytoplasmic tail to the ankyrin repeat domain of TRPV4 in order to produce ultrarapid, force-induced channel activation within the focal adhesion.


Assuntos
Integrina beta1 , Mecanotransdução Celular , Adesão Celular , Células Endoteliais/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
11.
Acc Chem Res ; 54(18): 3529-3539, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34478255

RESUMO

The ability to perform multiplexed detection of various biomarkers within complex biological fluids in a robust, rapid, sensitive, and cost-effective manner could transform clinical diagnostics and enable personalized healthcare. Electrochemical (EC) sensor technology has been explored as a way to address this challenge because it does not require optical instrumentation and it is readily compatible with both integrated circuit and microfluidic technologies; yet this approach has had little impact as a viable commercial bioanalytical tool to date. The most critical limitation hindering their clinical application is the fact that EC sensors undergo rapid biofouling when exposed to complex biological samples (e.g., blood, plasma, saliva, urine), leading to the loss of sensitivity and selectivity. Thus, to break through this barrier, we must solve this biofouling problem.In response to this challenge, our group has developed a rapid, robust, and low-cost nanocomposite-based antifouling coating for multiplexed EC sensors that enables unprecedented performance in terms of biomarker signal detection compared to reported literature. The bioinspired antifouling coating that we developed is a nanoporous composite that contains various conductive nanomaterials, including gold nanowires (AuNWs), carbon nanotubes (CNTs), or reduced graphene oxide nanoflakes (rGOx). Each study has progressively evolved this technology to provide increasing performance while simplifying process flow, reducing time, and decreasing cost. For example, after successfully developing a semipermeable nanocomposite coating containing AuNWs cross-linked to bovine serum albumin (BSA) using glutaraldehyde, we replaced the nanomaterials with reduced graphene oxide, reducing the cost by 100-fold while maintaining similar signal transduction and antifouling properties. We, subsequently, developed a localized heat-induced coating method that significantly improved the efficiency of the drop-casting coating process and occurs within the unprecedented time of <1 min (at least 3 orders of magnitude faster than state-of-the-art). Moreover, the resulting coated electrodes can be stored at room temperature for at least 5 months and still maintain full sensitivity and specificity. Importantly, this improved coating showed excellent antifouling activity against various biological fluids, including plasma, serum, whole blood, urine, and saliva.To enable affinity-based sensing of multiple biomarkers simultaneously, we have developed multiplexed EC sensors coated with the improved nanocomposite coating and then employed a sandwich enzyme-linked immunosorbent assay (ELISA) format for signal detection in which the substrate for the enzyme bound to the secondary antibody precipitates locally at the molecular binding site above the electrode surface. Using this improved EC sensor platform, we demonstrated ultrasensitive detection of a wide range of biomarkers from biological fluids, including clinical biomarkers, in both single and multiplex formats (N = 4) with assay times of 37 and 15 min when integrated with a microfluidic system. These biosensors developed demonstrate the vast potential of solving the biofouling problem, and how it can enable potential clinically important diagnostic applications. This Account reviews our antifouling surface chemistry and the multiplexed EC sensor-based biodetection method we developed and places it in context of the various innovative contributions that have been made by other researchers in this field. We are optimistic that future iterations of these systems will change the way diagnostic testing is done, and where it can be carried out, in the future.


Assuntos
Biomarcadores/análise , Técnicas Eletroquímicas/métodos , Anticorpos/análise , Incrustação Biológica/prevenção & controle , Líquidos Corporais/química , Líquidos Corporais/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Microfluídica , Nanocompostos/química , Sistemas Automatizados de Assistência Junto ao Leito
12.
Annu Rev Pharmacol Toxicol ; 58: 37-64, 2018 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-29309256

RESUMO

Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches are beginning to be integrated into drug development and approval processes because they enable key pharmacokinetic (PK) parameters to be predicted from in vitro data. However, these approaches are hampered by many limitations, including an inability to incorporate organ-specific differentials in drug clearance, distribution, and absorption that result from differences in cell uptake, transport, and metabolism. Moreover, such approaches are generally unable to provide insight into pharmacodynamic (PD) parameters. Recent development of microfluidic Organ-on-a-Chip (Organ Chip) cell culture devices that recapitulate tissue-tissue interfaces, vascular perfusion, and organ-level functionality offer the ability to overcome these limitations when multiple Organ Chips are linked via their endothelium-lined vascular channels. Here, we discuss successes and challenges in the use of existing culture models and vascularized Organ Chips for PBPK and PD modeling of human drug responses, as well as in vitro to in vivo extrapolation (IVIVE) of these results, and how these approaches might advance drug development and regulatory review processes in the future.


Assuntos
Desenvolvimento de Medicamentos/métodos , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Simulação por Computador , Aprovação de Drogas/métodos , Humanos , Dispositivos Lab-On-A-Chip , Modelos Biológicos , Farmacocinética
13.
Development ; 145(16)2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29776965

RESUMO

Although initially developed to replace animal testing in drug development, human 'organ on a chip' (organ chip) microfluidic culture technology offers a new tool for studying tissue development and pathophysiology, which has brought us one step closer to carrying out human experimentation in vitro In this Spotlight article, I discuss the central role that developmental biology played in the early stages of organ-chip technology, and how these models have led to new insights into human physiology and disease mechanisms. Advantages and disadvantages of the organ-chip approach relative to organoids and other human cell cultures are also discussed.


Assuntos
Biologia do Desenvolvimento , Dispositivos Lab-On-A-Chip , Microfluídica , Organoides/citologia , Engenharia Tecidual , Alternativas aos Testes com Animais/instrumentação , Alternativas aos Testes com Animais/métodos , Biologia do Desenvolvimento/instrumentação , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências , Doença , Desenvolvimento Embrionário/fisiologia , Humanos , Microfluídica/instrumentação , Microfluídica/métodos , Microfluídica/tendências , Modelos Biológicos , Técnicas de Cultura de Órgãos/métodos , Técnicas de Cultura de Órgãos/tendências , Esferoides Celulares/citologia , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências
14.
Nat Rev Mol Cell Biol ; 10(1): 75-82, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19197334

RESUMO

Research in cellular mechanotransduction often focuses on how extracellular physical forces are converted into chemical signals at the cell surface. However, mechanical forces that are exerted on surface-adhesion receptors, such as integrins and cadherins, are also channelled along cytoskeletal filaments and concentrated at distant sites in the cytoplasm and nucleus. Here, we explore the molecular mechanisms by which forces might act at a distance to induce mechanochemical conversion in the nucleus and alter gene activities.


Assuntos
Núcleo Celular/fisiologia , Matriz Extracelular/fisiologia , Mecanotransdução Celular/fisiologia , Animais , Citoplasma/fisiologia , Humanos , Estresse Mecânico
15.
Proc Natl Acad Sci U S A ; 115(5): E982-E991, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29343648

RESUMO

Stimulation of protease-activated receptor 1 (PAR1) on endothelium by activated protein C (APC) is protective in several animal models of disease, and APC has been used clinically in severe sepsis and wound healing. Clinical use of APC, however, is limited by its immunogenicity and its anticoagulant activity. We show that a class of small molecules termed "parmodulins" that act at the cytosolic face of PAR1 stimulates APC-like cytoprotective signaling in endothelium. Parmodulins block thrombin generation in response to inflammatory mediators and inhibit platelet accumulation on endothelium cultured under flow. Evaluation of the antithrombotic mechanism showed that parmodulins induce cytoprotective signaling through Gßγ, activating a PI3K/Akt pathway and eliciting a genetic program that includes suppression of NF-κB-mediated transcriptional activation and up-regulation of select cytoprotective transcripts. STC1 is among the up-regulated transcripts, and knockdown of stanniocalin-1 blocks the protective effects of both parmodulins and APC. Induction of this signaling pathway in vivo protects against thromboinflammatory injury in blood vessels. Small-molecule activation of endothelial cytoprotection through PAR1 represents an approach for treatment of thromboinflammatory disease and provides proof-of-principle for the strategy of targeting the cytoplasmic surface of GPCRs to achieve pathway selective signaling.


Assuntos
Células Endoteliais/metabolismo , Inflamação/metabolismo , Receptor PAR-1/agonistas , Trombose/metabolismo , Animais , Apoptose , Fator Xa/metabolismo , Técnicas de Silenciamento de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Peptídeo Hidrolases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Transcrição Gênica , Regulação para Cima
16.
J Am Soc Nephrol ; 31(7): 1479-1495, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32540856

RESUMO

BACKGROUND: Genetic mutations in α-actinin-4 (ACTN4)-an important actin crosslinking cytoskeletal protein that provides structural support for kidney podocytes-have been linked to proteinuric glomerulosclerosis in humans. However, the effect of post-translational modifications of ACTN4 on podocyte integrity and kidney function is not known. METHODS: Using mass spectrometry, we found that ACTN4 is phosphorylated at serine (S) 159 in human podocytes. We used phosphomimetic and nonphosphorylatable ACTN4 to comprehensively study the effects of this phosphorylation in vitro and in vivo. We conducted x-ray crystallography, F-actin binding and bundling assays, and immunofluorescence staining to evaluate F-actin alignment. Microfluidic organ-on-a-chip technology was used to assess for detachment of podocytes simultaneously exposed to fluid flow and cyclic strain. We then used CRISPR/Cas9 to generate mouse models and assessed for renal injury by measuring albuminuria and examining kidney histology. We also performed targeted mass spectrometry to determine whether high extracellular glucose or TGF-ß levels increase phosphorylation of ACTN4. RESULTS: Compared with the wild type ACTN4, phosphomimetic ACTN4 demonstrated increased binding and bundling activity with F-actin in vitro. Phosphomimetic Actn4 mouse podocytes exhibited more spatially correlated F-actin alignment and a higher rate of detachment under mechanical stress. Phosphomimetic Actn4 mice developed proteinuria and glomerulosclerosis after subtotal nephrectomy. Moreover, we found that exposure to high extracellular glucose or TGF-ß stimulates phosphorylation of ACTN4 at S159 in podocytes. CONCLUSIONS: These findings suggest that increased phosphorylation of ACTN4 at S159 leads to biochemical, cellular, and renal pathology that is similar to pathology resulting from human disease-causing mutations in ACTN4. ACTN4 may mediate podocyte injury as a consequence of both genetic mutations and signaling events that modulate phosphorylation.


Assuntos
Actinina/metabolismo , Albuminúria/metabolismo , Glomerulosclerose Segmentar e Focal/metabolismo , Podócitos/metabolismo , Processamento de Proteína Pós-Traducional , Actinina/genética , Actinas/metabolismo , Actinas/ultraestrutura , Albuminúria/etiologia , Albuminúria/patologia , Animais , Células Cultivadas , Feminino , Glomerulosclerose Segmentar e Focal/etiologia , Glomerulosclerose Segmentar e Focal/patologia , Glucose/farmacologia , Humanos , Dispositivos Lab-On-A-Chip , Masculino , Camundongos , Nefrectomia/efeitos adversos , Peptidomiméticos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Serina/metabolismo , Fator de Crescimento Transformador beta/farmacologia
17.
Nat Mater ; 18(10): 1071-1077, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31209386

RESUMO

Cell stiffness measurements have led to insights into various physiological and pathological processes1,2. Although many cellular behaviours are influenced by intracellular mechanical forces3-6 that also alter the material properties of the cell, the precise mechanistic relationship between intracellular forces and cell stiffness remains unclear. Here we develop a cell mechanical imaging platform with high spatial resolution that reveals the existence of nanoscale stiffness patterns governed by intracellular forces. On the basis of these findings, we develop and validate a cellular mechanical model that quantitatively relates cell stiffness to intracellular forces. This allows us to determine the magnitude of tension within actin bundles, cell cortex and plasma membrane from the cell stiffness patterns across individual cells. These results expand our knowledge on the mechanical interaction between cells and their environments, and offer an alternative approach to determine physiologically relevant intracellular forces from high-resolution cell stiffness images.


Assuntos
Células , Nanoestruturas , Fenômenos Biomecânicos , Humanos , Microscopia de Força Atômica
18.
Small ; 15(42): e1903087, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31448553

RESUMO

Inhomogeneous microcapsules that can encapsulate various cargo for controlled release triggered by osmotic shock are designed and reported. The microcapsules are fabricated using a microfluidic approach and the inhomogeneity of shell thickness in the microcapsules can be controlled by tuning the flow rate ratio of the middle phase to the inner phase. This study demonstrates the swelling of these inhomogeneous microcapsules begins at the thinnest part of shell and eventually leads to rupture at the weak spot with a low osmotic pressure. Systematic studies indicate the rupture fraction of these microcapsules increases with increasing inhomogeneity, while the rupture osmotic pressure decreases linearly with increasing inhomogeneity. The inhomogeneous microcapsules are demonstrated to be impermeable to small probe molecules, which enables long-term storage. Thus, these microcapsules can be used for long-term storage of enzymes, which can be controllably released through osmotic shock without impairing their biological activity. The study provides a new approach to design effective carriers to encapsulate biomolecules and release them on-demand upon applying osmotic shock.


Assuntos
Cápsulas/química , Microtecnologia/métodos , Pressão Osmótica , Soluções Hipotônicas , Microfluídica , Peso Molecular , Imagem Óptica , Peptídeo Hidrolases/metabolismo
19.
Nat Methods ; 13(2): 151-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26689262

RESUMO

Here we describe the development of a human lung 'small airway-on-a-chip' containing a differentiated, mucociliary bronchiolar epithelium and an underlying microvascular endothelium that experiences fluid flow, which allows for analysis of organ-level lung pathophysiology in vitro. Exposure of the epithelium to interleukin-13 (IL-13) reconstituted the goblet cell hyperplasia, cytokine hypersecretion and decreased ciliary function of asthmatics. Small airway chips lined with epithelial cells from individuals with chronic obstructive pulmonary disease recapitulated features of the disease such as selective cytokine hypersecretion, increased neutrophil recruitment and clinical exacerbation by exposure to viral and bacterial infections. With this robust in vitro method for modeling human lung inflammatory disorders, it is possible to detect synergistic effects of lung endothelium and epithelium on cytokine secretion, identify new biomarkers of disease exacerbation and measure responses to anti-inflammatory compounds that inhibit cytokine-induced recruitment of circulating neutrophils under flow.


Assuntos
Epitélio/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-13/farmacologia , Dispositivos Lab-On-A-Chip , Pneumopatias/tratamento farmacológico , Pneumopatias/metabolismo , Humanos , Inflamação/patologia , Técnicas de Cultura de Tecidos
20.
Proc Natl Acad Sci U S A ; 113(1): E7-15, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26668389

RESUMO

A human gut-on-a-chip microdevice was used to coculture multiple commensal microbes in contact with living human intestinal epithelial cells for more than a week in vitro and to analyze how gut microbiome, inflammatory cells, and peristalsis-associated mechanical deformations independently contribute to intestinal bacterial overgrowth and inflammation. This in vitro model replicated results from past animal and human studies, including demonstration that probiotic and antibiotic therapies can suppress villus injury induced by pathogenic bacteria. By ceasing peristalsis-like motions while maintaining luminal flow, lack of epithelial deformation was shown to trigger bacterial overgrowth similar to that observed in patients with ileus and inflammatory bowel disease. Analysis of intestinal inflammation on-chip revealed that immune cells and lipopolysaccharide endotoxin together stimulate epithelial cells to produce four proinflammatory cytokines (IL-8, IL-6, IL-1ß, and TNF-α) that are necessary and sufficient to induce villus injury and compromise intestinal barrier function. Thus, this human gut-on-a-chip can be used to analyze contributions of microbiome to intestinal pathophysiology and dissect disease mechanisms in a controlled manner that is not possible using existing in vitro systems or animal models.


Assuntos
Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiopatologia , Dispositivos Lab-On-A-Chip , Microbiota/fisiologia , Modelos Biológicos , Peristaltismo/fisiologia , Animais , Antibacterianos/uso terapêutico , Bactérias/crescimento & desenvolvimento , Células CACO-2 , Humanos , Íleus/tratamento farmacológico , Íleus/microbiologia , Íleus/fisiopatologia , Técnicas In Vitro , Doenças Inflamatórias Intestinais/tratamento farmacológico , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Interleucina-8/imunologia , Mucosa Intestinal/efeitos dos fármacos , Peristaltismo/efeitos dos fármacos , Probióticos/uso terapêutico , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA