Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lab Chip ; 23(22): 4804-4820, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37830228

RESUMO

Genetic reprogramming of immune cells to recognize and target tumor cells offers a possibility of long-term cure. Cell therapies, however, lack simple and affordable manufacturing workflows, especially to genetically edit immune cells to more effectively target cancer cells and avoid immune suppression mechanisms. Microfluidics is a pathway to improve the manufacturing precision of gene modified cells. However, to date, it remains to be demonstrated that microfluidic treatment preserves the functionality of T cell products in a complete workflow. In this study, we used microfluidics to perform CRISPR/Cas9 gene editing of CD5, a negative T-cell regulator, followed by the insertion of a chimeric antigen receptor (CAR) transgene via lentiviral vector transduction to generate CAR T cells targeted against the B cell antigen CD19. As part of the workflow, we have optimized a microfluidic device that relies on convective volume exchange between cells and surrounding fluid to deliver guide RNA and Cas9 ribonucleoprotein to primary T cells. We comprehensively tested critical design features of the device to improve the gene-edited product yield. By combining high-speed video and cell mechanics measurements using the atomic force microscope, we validate a model that relates the device design features to cell properties. Our findings showed enhanced performance was obtained by focusing the cells to counteract the flow resistance caused by the ridge constrictions, providing a ridge layout that allows sufficient cycles of compression and time for volume recovery, and including a gutter to clear aggregates that could reduce cell viability. The optimized device was used in a workflow to generate CD5-knockout CD19 CAR T cells. The microfluidics approach resulted in >60% CD5 editing efficiency, ≥80% cell viability, similar memory phenotype composition as unprocessed cells, and superior cell growth. The microfluidics workflow yielded 4-fold increase of edited T cells compared to an electroporation workflow post-expansion. The transduced CAR T cells showed similar transduction efficiency and cytotoxicity against CD19-positive leukemia cells. Moreover, patient-derived T cells showed the ability to be similarly edited, though their distinct biomechanics resulted in slightly lower outcomes. Microfluidics-based manufacturing is a promising path towards more productive clinical manufacturing of gene edited CAR T cells.


Assuntos
Receptores de Antígenos Quiméricos , Linfócitos T , Humanos , Receptores de Antígenos Quiméricos/metabolismo , Microfluídica , Fluxo de Trabalho , Edição de Genes , Transfecção , Imunoterapia Adotiva/métodos
2.
Stem Cells ; 41(11): 1037-1046, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37632456

RESUMO

Inherited retinal degeneration is a term used to describe heritable disorders that result from the death of light sensing photoreceptor cells. Although we and others believe that it will be possible to use gene therapy to halt disease progression early in its course, photoreceptor cell replacement will likely be required for patients who have already lost their sight. While advances in autologous photoreceptor cell manufacturing have been encouraging, development of technologies capable of efficiently delivering genome editing reagents to stem cells using current good manufacturing practices (cGMP) are needed. Gene editing reagents were delivered to induced pluripotent stem cells (iPSCs) using a Zephyr microfluidic transfection platform (CellFE). CRISPR-mediated cutting was quantified using an endonuclease assay. CRISPR correction was confirmed via digital PCR and Sanger sequencing. The resulting corrected cells were also karyotyped and differentiated into retinal organoids. We describe use of a novel microfluidic transfection platform to correct, via CRISPR-mediated homology-dependent repair (HDR), a disease-causing NR2E3 mutation in patient-derived iPSCs using cGMP compatible reagents and approaches. We show that the resulting cell lines have a corrected genotype, exhibit no off-target cutting, retain pluripotency and a normal karyotype and can be differentiated into retinal tissue suitable for transplantation. The ability to codeliver CRISPR/Cas9 and HDR templates to patient-derived iPSCs without using proprietary transfection reagents will streamline manufacturing protocols, increase the safety of resulting cell therapies, and greatly reduce the regulatory burden of clinical trials.


Assuntos
Edição de Genes , Células-Tronco Pluripotentes Induzidas , Humanos , Edição de Genes/métodos , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Microfluídica , Transfecção
3.
iScience ; 26(4): 106393, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37034996

RESUMO

Stiffness has been observed to decrease for many cancer cell types as their metastatic potential increases. Although cell mechanics and metastatic potential are related, the underlying molecular factors associated with these phenotypes remain unknown. Therefore, we have developed a workflow to measure the mechanical properties and gene expression of single cells that is used to generate large linked-datasets. The process combines atomic force microscopy to measure the mechanics of individual cells with multiplexed RT-qPCR gene expression analysis on the same single cells. Surprisingly, the genes that most strongly correlated with mechanical properties were not cytoskeletal, but rather were markers of extracellular matrix remodeling, epithelial-to-mesenchymal transition, cell adhesion, and cancer stemness. In addition, dimensionality reduction analysis showed that cell clustering was improved by combining mechanical and gene expression data types. The single cell genomechanics method demonstrates how single cell studies can identify molecular drivers that could affect the biophysical processes underpinning metastasis.

4.
J Biomech ; 150: 111479, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36871429

RESUMO

Because cells vary in thickness and in biomechanical properties, the use of a constant force trigger during atomic force microscopy (AFM) stiffness mapping produces a varied nominal strain that can obfuscate the comparison of local material properties. In this study, we measured the biomechanical spatial heterogeneity of ovarian and breast cancer cells by using an indentation-dependent pointwise Hertzian method. Force curves and surface topography were used together to determine cell stiffness as a function of nominal strain. By recording stiffness at a particular strain, it may be possible to improve comparison of the material properties of cells and produce higher contrast representations of cell mechanical properties. Defining a linear region of elasticity that corresponds to a modest nominal strain, we were able to clearly distinguish the mechanics of the perinuclear region of cells. We observed that, relative to the lamelopodial stiffness, the perinuclear region was softer for metastatic cancer cells than their nonmetastatic counterparts. Moreover, contrast in the strain-dependent elastography in comparison to conventional force mapping with Hertzian model analysis revealed a significant stiffening phenomenon in the thin lamellipodial region in which the modulus scales inversely and exponentially with cell thickness. The observed exponential stiffening is not affected by relaxation of cytoskeletal tension, but finite element modeling indicates it is affected by substrate adhesion. The novel cell mapping technique explores cancer cell mechanical nonlinearity that results from regional heterogeneity, which could help explain how metastatic cancer cells can show soft phenotypes while simultaneously increasing force generation and invasiveness.


Assuntos
Técnicas de Imagem por Elasticidade , Neoplasias , Humanos , Fenômenos Mecânicos , Elasticidade , Citoesqueleto , Microscopia de Força Atômica/métodos
5.
iScience ; 25(7): 104606, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35800766

RESUMO

The correlation between cardiovascular disease and iron deficiency anemia (IDA) is well documented but poorly understood. Using a multi-disciplinary approach, we explore the hypothesis that the biophysical alterations of red blood cells (RBCs) in IDA, such as variable degrees of microcytosis and decreased deformability may directly induce endothelial dysfunction via mechanobiological mechanisms. Using a combination of atomic force microscopy and microfluidics, we observed that subpopulations of IDA RBCs (idRBCs) are significantly stiffer and smaller than both healthy RBCs and the remaining idRBC population. Furthermore, computational simulations demonstrated that the smaller and stiffer idRBC subpopulations marginate toward the vessel wall causing aberrant shear stresses. This leads to increased vascular inflammation as confirmed with perfusion of idRBCs into our "endothelialized" microfluidic systems. Overall, our multifaceted approach demonstrates that the altered biophysical properties of idRBCs directly lead to vasculopathy, suggesting that the IDA and cardiovascular disease association extends beyond correlation and into causation.

6.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35101971

RESUMO

Metastatic osteosarcoma has a poor prognosis with a 2-y, event-free survival rate of ∼15 to 20%, highlighting the need for the advancement of efficacious therapeutics. Chimeric antigen receptor (CAR) T-cell therapy is a potent strategy for eliminating tumors by harnessing the immune system. However, clinical trials with CAR T cells in solid tumors have encountered significant challenges and have not yet demonstrated convincing evidence of efficacy for a large number of patients. A major bottleneck for the success of CAR T-cell therapy is our inability to monitor the accumulation of the CAR T cells in the tumor with clinical-imaging techniques. To address this, we developed a clinically translatable approach for labeling CAR T cells with iron oxide nanoparticles, which enabled the noninvasive detection of the iron-labeled T cells with magnetic resonance imaging (MRI), photoacoustic imaging (PAT), and magnetic particle imaging (MPI). Using a custom-made microfluidics device for T-cell labeling by mechanoporation, we achieved significant nanoparticle uptake in the CAR T cells, while preserving T-cell proliferation, viability, and function. Multimodal MRI, PAT, and MPI demonstrated homing of the T cells to osteosarcomas and off-target sites in animals administered with T cells labeled with the iron oxide nanoparticles, while T cells were not visualized in animals infused with unlabeled cells. This study details the successful labeling of CAR T cells with ferumoxytol, thereby paving the way for monitoring CAR T cells in solid tumors.


Assuntos
Neoplasias Ósseas , Óxido Ferroso-Férrico/farmacologia , Imunoterapia Adotiva , Imageamento por Ressonância Magnética , Nanopartículas/uso terapêutico , Neoplasias Experimentais , Osteossarcoma , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/terapia , Camundongos , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Osteossarcoma/diagnóstico por imagem , Osteossarcoma/imunologia , Osteossarcoma/terapia
7.
Sci Rep ; 12(1): 2955, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35194089

RESUMO

Regenerative medicine uses the patient own stem cells to regenerate damaged tissues. Molecular imaging techniques are commonly used to image the transplanted cells, either right after surgery or at a later time. However, few techniques are fast or straightforward enough to label cells intraoperatively. Adipose tissue-derived stem cells (ADSCs) were harvested from knee joints of minipigs. The cells were labeled with PET contrast agent by flowing mechanoporation using a microfluidic device. While flowing through a series of microchannels, cells are compressed repeatedly by micro-ridges, which open transient pores in their membranes and induce convective transport, intended to facilitate the transport of 68Ga-labeled and lipid-coated mesoporous nanoparticles (MSNs) into the cells. This process enables cells to be labeled in a matter of seconds. Cells labeled with this approach were then implanted into cartilage defects, and the implant was imaged using positron emission tomography (PET) post-surgery. The microfluidic device can efficiently label millions of cells with 68Ga-labeled MSNs in as little as 15 min. The method achieved labeling efficiency greater than 5 Bq/cell on average, comparable to 30 min-long passive co-incubation with 68Ga-MSNs, but with improved biocompatibility due to the reduced exposure to ionizing radiation. Labeling time could also be accelerated by increasing throughput through more parallel channels. Finally, as a proof of concept, ADSCs were labeled with 68Ga-MSNs and quantitatively assessed using clinical PET/MR in a mock transplant operation in pig knee joints. MSN-assisted mechanoporation is a rapid, effective and straightforward approach to label cells with 68Ga. Given its high efficiency, this labeling method can be used to track small cells populations without significant effects on viability. The system is applicable to a variety of cell tracking studies for cancer therapy, regenerative therapy, and immunotherapy.


Assuntos
Tecido Adiposo/metabolismo , Radioisótopos de Gálio/farmacologia , Nanopartículas , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacologia , Células-Tronco/metabolismo , Animais , Suínos , Porco Miniatura
8.
Sci Rep ; 11(1): 21407, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725429

RESUMO

Messenger RNA (mRNA) delivery provides gene therapy with the potential to achieve transient therapeutic efficacy without risk of insertional mutagenesis. Amongst other applications, mRNA can be employed as a platform to deliver gene editing molecules, to achieve protein expression as an alternative to enzyme replacement therapies, and to express chimeric antigen receptors (CARs) on immune cells for the treatment of cancer. We designed a novel microfluidic device that allows for efficient mRNA delivery via volume exchange for convective transfection (VECT). In the device, cells flow through a ridged channel that enforces a series of ultra-fast and large intensity deformations able to transiently open pores and induce convective transport of mRNA into the cell. Here, we describe efficient delivery of mRNA into T cells, natural killer (NK) cells and hematopoietic stem and progenitor cells (HSPCs), three human primary cell types widely used for ex vivo gene therapy applications. Results demonstrate that the device can operate at a wide range of cell and payload concentrations and that ultra-fast compressions do not have a negative impact on T cell function, making this a novel and competitive platform for the development of ex vivo mRNA-based gene therapies and other cell products engineered with mRNA.


Assuntos
Células-Tronco Hematopoéticas/citologia , Linfócitos/metabolismo , Microfluídica , Células-Tronco/citologia , Transfecção/métodos , Antígenos CD34/biossíntese , Transporte Biológico , Sobrevivência Celular , Eletroporação , Citometria de Fluxo , Terapia Genética , Humanos , Células Matadoras Naturais/citologia , Dispositivos Lab-On-A-Chip , Engenharia de Proteínas , RNA Mensageiro/metabolismo , Linfócitos T/citologia
9.
Sci Rep ; 11(1): 18032, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504124

RESUMO

The isolation of a patient's metastatic cancer cells is the first, enabling step toward treatment of that patient using modern personalized medicine techniques. Whereas traditional standard-of-care approaches select treatments for cancer patients based on the histological classification of cancerous tissue at the time of diagnosis, personalized medicine techniques leverage molecular and functional analysis of a patient's own cancer cells to select treatments with the highest likelihood of being effective. Unfortunately, the pure populations of cancer cells required for these analyses can be difficult to acquire, given that metastatic cancer cells typically reside in fluid containing many different cell populations. Detection and analyses of cancer cells therefore require separation from these contaminating cells. Conventional cell sorting approaches such as Fluorescence Activated Cell Sorting or Magnetic Activated Cell Sorting rely on the presence of distinct surface markers on cells of interest which may not be known nor exist for cancer applications. In this work, we present a microfluidic platform capable of label-free enrichment of tumor cells from the ascites fluid of ovarian cancer patients. This approach sorts cells based on differences in biomechanical properties, and therefore does not require any labeling or other pre-sort interference with the cells. The method is also useful in the cases when specific surface markers do not exist for cells of interest. In model ovarian cancer cell lines, the method was used to separate invasive subtypes from less invasive subtypes with an enrichment of ~ sixfold. In ascites specimens from ovarian cancer patients, we found the enrichment protocol resulted in an improved purity of P53 mutant cells indicative of the presence of ovarian cancer cells. We believe that this technology could enable the application of personalized medicine based on analysis of liquid biopsy patient specimens, such as ascites from ovarian cancer patients, for quick evaluation of metastatic disease progression and determination of patient-specific treatment.


Assuntos
Ascite/diagnóstico , Separação Celular/métodos , Técnicas Analíticas Microfluídicas/instrumentação , Células Neoplásicas Circulantes/metabolismo , Neoplasias Ovarianas/diagnóstico , Proteína Supressora de Tumor p53/genética , Ascite/genética , Ascite/metabolismo , Ascite/patologia , Líquido Ascítico/metabolismo , Líquido Ascítico/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Fenômenos Biomecânicos , Separação Celular/instrumentação , Feminino , Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Biópsia Líquida/métodos , Modelos Biológicos , Reação em Cadeia da Polimerase Multiplex , Mutação , Invasividade Neoplásica , Células Neoplásicas Circulantes/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Medicina de Precisão , Proteína Supressora de Tumor p53/metabolismo
10.
Stem Cells Transl Med ; 10(10): 1384-1393, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34156760

RESUMO

Autologous photoreceptor cell replacement is one of the most promising approaches currently under development for the treatment of inherited retinal degenerative blindness. Unlike endogenous stem cell populations, induced pluripotent stem cells (iPSCs) can be differentiated into both rod and cone photoreceptors in high numbers, making them ideal for this application. That said, in addition to photoreceptor cells, state of the art retinal differentiation protocols give rise to all of the different cell types of the normal retina, the majority of which are not required and may in fact hinder successful photoreceptor cell replacement. As such, following differentiation photoreceptor cell enrichment will likely be required. In addition, to prevent the newly generated photoreceptor cells from suffering the same fate as the patient's original cells, correction of the patient's disease-causing genetic mutations will be necessary. In this review we discuss literature pertaining to the use of different cell sorting and transfection approaches with a focus on the development and use of novel next generation microfluidic devices. We will discuss how gold standard strategies have been used, the advantages and disadvantages of each, and how novel microfluidic platforms can be incorporated into the clinical manufacturing pipeline to reduce the complexity, cost, and regulatory burden associated with clinical grade production of photoreceptor cells for autologous cell replacement.


Assuntos
Células-Tronco Pluripotentes Induzidas , Degeneração Retiniana , Diferenciação Celular , Humanos , Microfluídica , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/terapia
11.
Langmuir ; 37(16): 4783-4792, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33848167

RESUMO

Cytotoxic effector cells are an integral component of the immune response against pathogens and diseases such as cancer and thus of great interest to researchers who wish to enhance the native immune response. Although researchers routinely use particles to stimulate cytotoxic T cells, few studies have comprehensively investigated: (1) beyond initial activation responses (i.e., proliferation and CD25/CD69 expression) to downstream cancer-killing effects and (2) how to drive cytotoxic T-cell responses by adjusting biomolecular and physical properties of particles. In this study, we designed particles displaying an anti-CD3 antibody to activate cytotoxic T cells and study their downstream cytotoxic effects. We evaluated the effect of antibody immobilization, particle size, molecular surface density of an anti-CD3 antibody, and the inclusion of an anti-CD28 antibody on cytolytic granule release by T cells. We found that immobilizing the anti-CD3 antibody onto smaller nanoparticles elicited increased T-cell activation products for an equivalent delivery of the anti-CD3 antibody. We further established that the mechanism behind increased cancer cell death was associated with the proximity of T cells to cancer cells. Functionalizing particles additionally with the anti-CD28 antibody at an optimized antibody density caused increased T-cell proliferation and T-cell binding but we observed no effective increase in cytotoxicity. Meaningfully, our results are discussed within the context of commercially available and widely used anti-CD3/28 Dynabeads. These results showed that T-cell activation and cytotoxicity can be optimized with a molecular presentation on smaller particles and thus, offer exciting new possibilities to engineer T-cell activation responses for effective outcomes.


Assuntos
Anticorpos Monoclonais , Ativação Linfocitária , Linfócitos T Citotóxicos , Antígenos CD28 , Complexo CD3 , Células Cultivadas , Humanos
13.
APL Bioeng ; 4(3): 036101, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32637856

RESUMO

To improve the survival rate of cancer patients, new diagnosis strategies are necessary to detect lower levels of cancer cells before and after treatment regimens. The scarcity of diseased cells, particularly in residual disease after treatment, demands highly sensitive detection approaches or the ability to enrich the diseased cells in relation to normal cells. We report a label-free microfluidic approach to enrich leukemia cells from healthy cells using inherent differences in cell biophysical properties. The microfluidic device consists of a channel with an array of diagonal ridges that recurrently compress and translate flowing cells in proportion to cell stiffness. Using devices optimized for acute T cell leukemia model Jurkat, the stiffer white blood cells were translated orthogonally to the channel length, while softer leukemia cells followed hydrodynamic flow. The device enriched Jurkat leukemia cells from white blood cells with an enrichment factor of over 760. The sensitivity, specificity, and accuracy of the device were found to be > 0.8 . The values of sensitivity and specificity could be adjusted by selecting one or multiple outlets for analysis. We demonstrate that low levels of Jurkat leukemia cells (1 in 10 4 white blood cells) could be more quickly detected using flow cytometry by using the stiffness sorting pre-enrichment. In a second mode of operation, the device was implemented to sort resistive leukemia cells from both drug-sensitive leukemia cells and normal white blood cells. Therefore, microfluidic biomechanical sorting can be a useful tool to enrich leukemia cells that may improve downstream analyses.

14.
Theranostics ; 10(13): 6024-6034, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32483435

RESUMO

Autologous therapeutic cells are typically harvested and transplanted in one single surgery. This makes it impossible to label them with imaging biomarkers through classical transfection techniques in a laboratory. To solve this problem, we developed a novel microfluidic device, which provides highly efficient labeling of therapeutic cells with imaging biomarkers through mechanoporation. Methods: Studies were performed with a new, custom-designed microfluidic device, which contains ridges, which compress adipose tissue-derived stem cells (ADSCs) during their device passage. Cell relaxation after compression leads to cell volume exchange for convective transfer of nanoparticles and nanoparticle uptake into the cell. ADSCs were passed through the microfluidic device doped with iron oxide nanoparticles and 18F-fluorodeoxyglucose (FDG). The cellular nanoparticle and radiotracer uptake was evaluated with DAB-Prussian blue, fluorescent microscopy, and inductively coupled plasma spectrometry (ICP). Labeled and unlabeled ADSCs were imaged in vitro as well as ex vivo in pig knee specimen with magnetic resonance imaging (MRI) and positron emission tomography (PET). T2 relaxation times and radiotracer signal were compared between labeled and unlabeled cell transplants using Student T-test with p<0.05. Results: We report significant labeling of ADSCs with iron oxide nanoparticles and 18F-FDG within 12+/-3 minutes. Mechanoporation of ADSCs with our microfluidic device led to significant nanoparticle (> 1 pg iron per cell) and 18F-FDG uptake (61 mBq/cell), with a labeling efficiency of 95%. The labeled ADSCs could be detected with MRI and PET imaging technologies: Nanoparticle labeled ADSC demonstrated significantly shorter T2 relaxation times (24.2±2.1 ms) compared to unlabeled cells (79.6±0.8 ms) on MRI (p<0.05) and 18F-FDG labeled ADSC showed significantly higher radiotracer uptake (614.3 ± 9.5 Bq / 1×104 cells) compared to controls (0.0 ± 0.0 Bq/ 1×104 cells) on gamma counting (p<0.05). After implantation of dual-labeled ADSCs into pig knee specimen, the labeled ADSCs revealed significantly shorter T2 relaxation times (41±0.6 ms) compared to unlabeled controls (90±1.8 ms) (p<0.05). Conclusion: The labeling of therapeutic cells with our new microfluidic device does not require any chemical intervention, therefore it is broadly and immediately clinically applicable. Cellular labeling using mechanoporation can improve our understanding of in vivo biodistributions of therapeutic cells and ultimately improve long-term outcomes of therapeutic cell transplants.


Assuntos
Fluordesoxiglucose F18/administração & dosagem , Imagem Multimodal/métodos , Células-Tronco/metabolismo , Tecido Adiposo/diagnóstico por imagem , Tecido Adiposo/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita/administração & dosagem , Tomografia por Emissão de Pósitrons/métodos , Coloração e Rotulagem/métodos , Suínos
16.
Spine (Phila Pa 1976) ; 45(8): E417-E424, 2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-31703050

RESUMO

STUDY DESIGN: An in vivo study examining the functional osseointegration of smooth, rough, and porous surface topographies presenting polyether-ether-ketone (PEEK) or titanium surface chemistry. OBJECTIVE: To investigate the effects of surface topography and surface chemistry on implant osseointegration. SUMMARY OF BACKGROUND DATA: Interbody fusion devices have been used for decades to facilitate fusion across the disc space, yet debate continues over their optimal surface topography and chemistry. Though both factors influence osseointegration, the relative effects of each are not fully understood. METHODS: Smooth, rough, and porous implants presenting either a PEEK or titanium surface chemistry were implanted into the proximal tibial metaphyses of 36 skeletally mature male Sprague Dawley rats. At 8 weeks, animals were euthanized and bone-implant interfaces were subjected to micro-computed tomography analysis (n = 12), histology (n = 4), and biomechanical pullout testing (n = 8) to assess functional osseointegration and implant fixation. RESULTS: Micro-computed tomography analysis demonstrated that bone ingrowth was 38.9 ±â€Š2.8% for porous PEEK and 30.7 ±â€Š3.3% for porous titanium (P = 0.07). No differences in fixation strength were detected between porous PEEK and porous titanium despite titanium surfaces exhibiting an overall increase in bone-implant contact compared with PEEK (P < 0.01). Porous surfaces exhibited increased fixation strength compared with smooth and rough surfaces regardless of surface chemistry (P < 0.05). Across all groups both surface topography and chemistry had a significant overall effect on fixation strength (P < 0.05), but topography accounted for 65.3% of the total variance (ω = 0.65), whereas surface chemistry accounted for 5.9% (ω = 0.06). CONCLUSIONS: The effect of surface topography (specifically porosity) dominated the effect of surface chemistry in this study and could lead to further improvements in orthopedic device design. The poor osseointegration of existing smooth PEEK implants may be linked more to their smooth surface topography rather than their material composition. LEVEL OF EVIDENCE: N/A.


Assuntos
Prótese Ancorada no Osso/tendências , Cetonas/química , Osseointegração/efeitos dos fármacos , Osseointegração/fisiologia , Polietilenoglicóis/química , Titânio/química , Animais , Benzofenonas , Cetonas/administração & dosagem , Masculino , Polietilenoglicóis/administração & dosagem , Polímeros , Porosidade , Próteses e Implantes/tendências , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície , Titânio/administração & dosagem , Microtomografia por Raio-X/métodos
17.
Cell Stem Cell ; 24(4): 608-620.e6, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30880025

RESUMO

Hematopoietic stem cell (HSC) quiescence is a tightly regulated process crucial for hematopoietic regeneration, which requires a healthy and supportive microenvironmental niche within the bone marrow (BM). Here, we show that deletion of Ptpn21, a protein tyrosine phosphatase highly expressed in HSCs, induces stem cell egress from the niche due to impaired retention within the BM. Ptpn21-/- HSCs exhibit enhanced mobility, decreased quiescence, increased apoptosis, and defective reconstitution capacity. Ptpn21 deletion also decreased HSC stiffness and increased physical deformability, in part by dephosphorylating Spetin1 (Tyr246), a poorly described component of the cytoskeleton. Elevated phosphorylation of Spetin1 in Ptpn21-/- cells impaired cytoskeletal remodeling, contributed to cortical instability, and decreased cell rigidity. Collectively, these findings show that Ptpn21 maintains cellular mechanics, which is correlated with its important functions in HSC niche retention and preservation of hematopoietic regeneration capacity.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Septinas/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Tirosina Fosfatases não Receptoras/deficiência , Nicho de Células-Tronco
18.
iScience ; 9: 347-358, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30453164

RESUMO

At early stages of organismal development, endothelial cells self-organize into complex networks subsequently giving rise to mature blood vessels. The compromised collective behavior of endothelial cells leads to the development of a number of vascular diseases, many of which can be life-threatening. Cerebral cavernous malformation is an example of vascular diseases caused by abnormal development of blood vessels in the brain. Despite numerous efforts to date, enlarged blood vessels (cavernomas) can be effectively treated only by risky and complex brain surgery. In this work, we use a comprehensive simulation model to dissect the mechanisms contributing to an emergent behavior of the multicellular system. By tightly integrating computational and experimental approaches we gain a systems-level understanding of the basic mechanisms of vascular tubule formation, its destabilization, and pharmacological rescue, which may facilitate the development of new strategies for manipulating collective endothelial cell behavior in the disease context.

19.
JCI Insight ; 3(20)2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30333317

RESUMO

Fibrosis is characterized by persistent deposition of extracellular matrix (ECM) by fibroblasts. Fibroblast mechanosensing of a stiffened ECM is hypothesized to drive the fibrotic program; however, the spatial distribution of ECM mechanics and their derangements in progressive fibrosis are poorly characterized. Importantly, fibrosis presents with significant histopathological heterogeneity at the microscale. Here, we report that fibroblastic foci (FF), the regions of active fibrogenesis in idiopathic pulmonary fibrosis (IPF), are surprisingly of similar modulus as normal lung parenchyma and are nonlinearly elastic. In vitro, provisional ECMs with mechanical properties similar to those of FF activate both normal and IPF patient-derived fibroblasts, whereas type I collagen ECMs with similar mechanical properties do not. This is mediated, in part, by αvß3 integrin engagement and is augmented by loss of expression of Thy-1, which regulates αvß3 integrin avidity for ECM. Thy-1 loss potentiates cell contractility-driven strain stiffening of provisional ECM in vitro and causes elevated αvß3 integrin activation, increased fibrosis, and greater mortality following fibrotic lung injury in vivo. These data suggest a central role for αvß3 integrin and provisional ECM in overriding mechanical cues that normally impose quiescent phenotypes, driving progressive fibrosis through physical stiffening of the fibrotic niche.


Assuntos
Fibroblastos/patologia , Fibrose Pulmonar Idiopática/patologia , Integrina alfaVbeta3/metabolismo , Pulmão/patologia , Animais , Bleomicina/toxicidade , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/patologia , Feminino , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/genética , Pulmão/citologia , Masculino , Camundongos , Camundongos Knockout , Cultura Primária de Células , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo
20.
PLoS One ; 13(3): e0192631, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29518080

RESUMO

The highly proliferative and pluripotent characteristics of embryonic stem cells engender great promise for tissue engineering and regenerative medicine, but the rapid identification and isolation of target cell phenotypes remains challenging. Therefore, the objectives of this study were to characterize cell mechanics as a function of differentiation and to employ differences in cell stiffness to select population subsets with distinct mechanical, morphological, and biological properties. Biomechanical analysis with atomic force microscopy revealed that embryonic stem cells stiffened within one day of differentiation induced by leukemia inhibitory factor removal, with a lagging but pronounced change from spherical to spindle-shaped cell morphology. A microfluidic device was then employed to sort a differentially labeled mixture of pluripotent and differentiating cells based on stiffness, resulting in pluripotent cell enrichment in the soft device outlet. Furthermore, sorting an unlabeled population of partially differentiated cells produced a subset of "soft" cells that was enriched for the pluripotent phenotype, as assessed by post-sort characterization of cell mechanics, morphology, and gene expression. The results of this study indicate that intrinsic cell mechanical properties might serve as a basis for efficient, high-throughput, and label-free isolation of pluripotent stem cells, which will facilitate a greater biological understanding of pluripotency and advance the potential of pluripotent stem cell differentiated progeny as cell sources for tissue engineering and regenerative medicine.


Assuntos
Fibroblastos/citologia , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Fenômenos Biomecânicos , Fenômenos Biofísicos , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Células Cultivadas , Fibroblastos/metabolismo , Expressão Gênica , Camundongos , Microscopia de Força Atômica , Células-Tronco Embrionárias Murinas/metabolismo , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA