Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(10): e2115217119, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235449

RESUMO

The cytoskeleton of eukaryotic cells is primarily composed of networks of filamentous proteins, F-actin, microtubules, and intermediate filaments. Interactions among the cytoskeletal components are important in determining cell structure and in regulating cell functions. For example, F-actin and microtubules work together to control cell shape and polarity, while the subcellular organization and transport of vimentin intermediate filament (VIF) networks depend on their interactions with microtubules. However, it is generally thought that F-actin and VIFs form two coexisting but separate networks that are independent due to observed differences in their spatial distribution and functions. In this paper, we present a closer investigation of both the structural and functional interplay between the F-actin and VIF cytoskeletal networks. We characterize the structure of VIFs and F-actin networks within the cell cortex using structured illumination microscopy and cryo-electron tomography. We find that VIFs and F-actin form an interpenetrating network (IPN) with interactions at multiple length scales, and VIFs are integral components of F-actin stress fibers. From measurements of recovery of cell contractility after transient stretching, we find that the IPN structure results in enhanced contractile forces and contributes to cell resilience. Studies of reconstituted networks and dynamic measurements in cells suggest direct and specific associations between VIFs and F-actin. From these results, we conclude that VIFs and F-actin work synergistically, both in their structure and in their function. These results profoundly alter our understanding of the contributions of the components of the cytoskeleton, particularly the interactions between intermediate filaments and F-actin.


Assuntos
Citoplasma/metabolismo , Filamentos Intermediários/metabolismo , Vimentina/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Animais , Biopolímeros/metabolismo , Células Cultivadas , Tomografia com Microscopia Eletrônica/métodos , Filamentos Intermediários/química , Camundongos , Vimentina/química
2.
Mol Ther ; 31(5): 1480-1495, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-36932674

RESUMO

Optogenetic techniques permit non-invasive, spatiotemporal, and reversible modulation of cellular activities. Here, we report a novel optogenetic regulatory system for insulin secretion in human pluripotent stem cell (hPSC)-derived pancreatic islet-like organoids using monSTIM1 (monster-opto-Stromal interaction molecule 1), an ultra-light-sensitive OptoSTIM1 variant. The monSTIM1 transgene was incorporated at the AAVS1 locus in human embryonic stem cells (hESCs) by CRISPR-Cas9-mediated genome editing. Not only were we able to elicit light-induced intracellular Ca2+ concentration ([Ca2+]i) transients from the resulting homozygous monSTIM1+/+-hESCs, but we also successfully differentiated them into pancreatic islet-like organoids (PIOs). Upon light stimulation, the ß-cells in these monSTIM1+/+-PIOs displayed reversible and reproducible [Ca2+]i transient dynamics. Furthermore, in response to photoexcitation, they secreted human insulin. Light-responsive insulin secretion was similarly observed in monSTIM1+/+-PIOs produced from neonatal diabetes (ND) patient-derived induced pluripotent stem cells (iPSCs). Under LED illumination, monSTIM1+/+-PIO-transplanted diabetic mice produced human c-peptide. Collectively, we developed a cellular model for the optogenetic control of insulin secretion using hPSCs, with the potential to be applied to the amelioration of hyperglycemic disorders.


Assuntos
Diabetes Mellitus Experimental , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Humanos , Camundongos , Animais , Secreção de Insulina , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Organoides , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular
3.
Biophys J ; 122(9): 1691-1700, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36987391

RESUMO

The brain cells are affected by continuous fluid shear stress that is driven by varying hydrostatic and osmotic pressure conditions, depending on the brain's pathophysiological conditions. Although all brain cells are sensitive to the subtle changes in various physicochemical factors in the microenvironment, microglia, the resident brain immune cells, exhibit the most significant morphodynamic transformation. However, little is known about the phenotypic alterations in microglia in response to changes in fluid shear stress. In this study, we established a flow-controlled microenvironment to investigate the effects of shear flow on microglial phenotypes, including morphology, motility, and activation states. We observed two distinct morphologies of microglia in a static condition: bipolar cells that oscillate along their long axis and unipolar cells that migrate persistently. When exposed to flow, a significant fraction of bipolar cells showed unstable oscillation with an increased amplitude of oscillation and a decreased frequency, which consequently led to the phenotypic transformation of oscillating cells into migrating cells. Furthermore, we observed that the level of proinflammatory genes increased in response to shear stress, although there were no significant changes in the level of antiinflammatory genes. Our findings suggest that an interstitial fluid-level stimulus can cause a dramatic phenotypic shift in microglia toward proinflammatory states, shedding light on the pathological outbreaks of severe brain diseases. Given that the fluidic environment in the brain can be locally disrupted in pathological circumstances, the mechanical stimulus by fluid flow should also be considered a crucial element in regulating the immune activities of the microglia in brain diseases.


Assuntos
Encefalopatias , Microglia , Humanos , Microglia/patologia , Microglia/fisiologia , Encéfalo , Encefalopatias/patologia , Anti-Inflamatórios
4.
J Craniofac Surg ; 34(3): 904-909, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-36730874

RESUMO

BACKGROUND: Transforming growth factor-beta (TGF-ß) plays an instrumental role in forming scars and keloids. TGF-ß isoforms exhibit differential expression, indicating distinct wound healing and scar formation functions. However, the role of TGF-ß1 and TGF-ß3 in wound healing and scar formation remains unclear. This study aimed to compare the specific roles of TGF-ß1 and TGF-ß3 in wound healing and scar formation by biomolecular analysis. MATERIALS AND METHODS: The study was conducted by cell isolation and culture cells from a total of 20 human samples. Normal human fibroblasts (NHF) were isolated from normal human samples and myofibroblasts from the different scar types, namely hypertrophic (HT) and keloid (K) scars. NHF and cells from the HT, and K scar, each of which were divided into 3 sample groups: the untreated control, TGF-ß1 (10 µg/mL)-treated group, and TGF-ß3 (10 µg/mL)-treated group. The results of confocal microscopy and fluorescence-activated cell sorting experiments were compared. RESULTS: Both the HT and K groups had higher α-smooth muscle actin (α-SMA) expression than the NHF group in the untreated control group. In comparison with the untreated group, NHFs showed a significant increase in α-SMA expression in the TGF-ß1-treated group. HT showed a high α-SMA level, which was statistically significant compared with the normal fibroblasts. In the TGF-ß3-treated group, α-SMA expression was slightly increased in NHF as compared with the untreated group. TGF-ß3 treated HT exhibited a greater reduction in α-SMA expression than in the TGF-ß1 treated HT. K, on the other hand, had only a minimal effect on the treatment of TGF-ß1 and TGF-ß3. CONCLUSIONS: The findings suggest that TGF-ß3 may play a regulatory role in the wound repair process, which could be useful in the development of scar-reducing therapies for patients with scar-related cosmetic concerns.


Assuntos
Cicatriz Hipertrófica , Queloide , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Fator de Crescimento Transformador beta , Fibroblastos , Hipertrofia , Fatores de Crescimento Transformadores/metabolismo
5.
J Neurosci Res ; 99(7): 1864-1884, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33772860

RESUMO

Traumatic brain injury (TBI) by an external physical impact results in compromised brain function via undesired neuronal death. Following the injury, resident and peripheral immune cells, astrocytes, and neural stem cells (NSCs) cooperatively contribute to the recovery of the neuronal function after TBI. However, excessive pro-inflammatory responses of immune cells, and the disappearance of endogenous NSCs at the injury site during the acute phase of TBI, can exacerbate TBI progression leading to incomplete healing. Therefore, positive outcomes may depend on early interventions to control the injury-associated cellular milieu in the early phase of injury. Here, we explore electrical stimulation (ES) of the injury site in a rodent model (male Sprague-Dawley rats) to investigate its overall effect on the constituent brain cell phenotype and composition during the acute phase of TBI. Our data showed that a brief ES for 1 hr on day 2 of TBI promoted anti-inflammatory phenotypes of microglia as assessed by CD206 expression and increased the population of NSCs and Nestin+ astrocytes at 7 days post-TBI. Also, ES effectively increased the number of viable neurons when compared to the unstimulated control group. Given the salience of microglia and neural stem cells for healing after TBI, our results strongly support the potential benefit of the therapeutic use of ES during the acute phase of TBI to regulate neuroinflammation and to enhance neuroregeneration.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Estimulação Elétrica/métodos , Regeneração Nervosa/fisiologia , Células-Tronco Neurais , Neuroglia , Animais , Masculino , Fenótipo , Ratos , Ratos Sprague-Dawley
6.
Toxicol Appl Pharmacol ; 424: 115589, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34029620

RESUMO

Changes in the physical state of the cells can serve as important indicators of stress responses because they are closely linked with the changes in the pathophysiological functions of the cells. Physical traits can be conveniently assessed by analyzing the morphological features and the stresses at the cell-matrix and cell-cell adhesions in both single-cell and monolayer model systems in 2D. In this study, we investigated the mechano-stress responses of human bronchial epithelial cells, BEAS-2B, to two functionally distinct groups of biocides identified during the humidifier disinfectant accident, namely, guanidine (PHMG) and isothiazolinone (CMIT/MIT). We analyzed the physical traits, including cell area, nuclear area, and nuclear shape. While the results showed inconsistent average responses to the biocides, the degree of dispersion in the data set, measured by standard deviation, was remarkably higher in CMIT/MIT treated cells for all traits. As mechano-stress endpoints, traction and intercellular stresses were also measured, and the cytoskeletal actin structures were analyzed using immunofluorescence. This study demonstrates the versatility of the real-time imaging-based biomechanical analysis, which will contribute to identifying the temporally sensitive cellular behaviors as well as the emergence of heterogeneity in response to exogenously imposed stress factors. This study will also shed light on a comparative understanding of less studied substance, CMIT/MIT, in relation to a more studied substance, PHMG, which will further contribute to more strategic planning for proper risk management of the ingredients involved in toxicological accidents.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Desinfetantes/toxicidade , Guanidina/toxicidade , Tiazóis/toxicidade , Linhagem Celular , Células Epiteliais , Humanos
7.
Aesthetic Plast Surg ; 43(5): 1371-1380, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31346713

RESUMO

Myofibroblasts play a central role in matrix formation and wound contraction during wound healing and undergo apoptosis at the end of the healing. Hypertrophic scarring is a pathologic condition in which myofibroblasts persist in the tissue. It has been hypothesized that abnormalities in epidermal-dermal crosstalk underlie this pathology. Therefore, in this study, we investigated whether myofibroblasts are affected by keratinocytes. Transforming growth factor beta-induced myofibroblasts (Imyo) and myofibroblasts from hypertrophic scar tissue (Hmyo) were characterized using microarrays. Keratinocytes were co-cultured with myofibroblasts, and quantitative PCR analysis was performed. We found that numerous extracellular matrix- and smooth muscle cell-associated genes were upregulated in Imyo and Hmyo respectively, and these findings suggest that Hmyo are fully differentiated myofibroblasts and that Imyo are less differentiated than Hmyo. Decreased collagen type 1 gene expression was found in keratinocytes co-cultured with Imyo and Hmyo; further, α-smooth muscle actin expression in Imyo increased in the presence of keratinocytes. These observations indicate that keratinocytes play a role in the development of pathological fibrosis in hypertrophic scar tissue by regulating the behavior of dermal fibroblasts and myofibroblasts. We believe that this study provides the basis for understanding the pathophysiology of hypertrophic scarring and identifying new therapeutic approaches for this dysfunction.No Level Assigned This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors - www.springer.com/00266 .


Assuntos
Cicatriz Hipertrófica/patologia , Colágeno Tipo I/genética , Miofibroblastos/patologia , Fator de Crescimento Transformador beta1/farmacologia , Cicatrização/genética , Apoptose/genética , Diferenciação Celular/genética , Células Cultivadas , Cicatriz Hipertrófica/genética , Técnicas de Cocultura , Estudos de Coortes , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Regulação da Expressão Gênica , Hospitais Universitários , Humanos , Queratinócitos/citologia , Queratinócitos/patologia , Miofibroblastos/citologia , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Regulação para Cima , Cicatrização/fisiologia
8.
Int J Mol Sci ; 19(2)2018 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-29370101

RESUMO

Human mesenchymal stem cells (hMSCs) have been widely studied for therapeutic development in tissue engineering and regenerative medicine. They can be harvested from human donors via tissue biopsies, such as bone marrow aspiration, and cultured to reach clinically relevant cell numbers. However, an unmet issue lies in the fact that the hMSC donors for regenerative therapies are more likely to be of advanced age. Their stem cells are not as potent compared to those of young donors, and continue to lose healthy, stemness-related activities when the hMSCs are serially passaged in tissue culture plates. Here, we have developed a cheap, scalable, and effective copolymer film to culture hMSCs obtained from aged human donors over several passages without loss of reactive oxygen species (ROS) handling or differentiation capacity. Assays of cell morphology, reactive oxygen species load, and differentiation potential demonstrate the effectiveness of copolymer culture on reduction in senescence-related activities of aging donor-derived hMSCs that could hinder the therapeutic potential of autologous stem cell therapies.


Assuntos
Envelhecimento/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Cultura Primária de Células/métodos , Espécies Reativas de Oxigênio/metabolismo , Materiais Biocompatíveis/química , Proliferação de Células , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Poliésteres , Polietilenoglicóis
9.
J Cell Physiol ; 231(8): 1822-31, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26661891

RESUMO

The expansion of autologous chondrocytes in vitro is used to generate sufficient populations for cell-based therapies. However, during monolayer culture, chondrocytes lose inherent characteristics and shift to fibroblast-like cells as passage number increase. Here, we investigated passage-dependent changes in cellular physiology, including cellular morphology, motility, and gene and protein expression, as well as the role of focal adhesion and cytoskeletal regulation in the dedifferentiation process. We found that the gene and protein expression levels of both the focal adhesion complex and small Rho GTPases are upregulated with increasing passage number and are closely linked to chondrocyte dedifferentiation. The inhibition of focal adhesion kinase (FAK) but not small Rho GTPases induced the loss of fibroblastic traits and the recovery of collagen type II, aggrecan, and SOX9 expression levels in dedifferentiated chondrocytes. Based on these findings, we propose a strategy to suppress chondrogenic dedifferentiation by inhibiting the identified FAK or Src pathways while maintaining the expansion capability of chondrocytes in a 2D environment. These results highlight a potential therapeutic target for the treatment of skeletal diseases and the generation of cartilage in tissue-engineering approaches. J. Cell. Physiol. 231: 1822-1831, 2016. © 2015 Wiley Periodicals, Inc.


Assuntos
Desdiferenciação Celular , Condrócitos/patologia , Condrogênese , Adesões Focais/patologia , Agrecanas/metabolismo , Animais , Desdiferenciação Celular/efeitos dos fármacos , Proliferação de Células , Forma Celular , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Colágeno Tipo II/metabolismo , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Masculino , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos Sprague-Dawley , Fatores de Transcrição SOX9/metabolismo , Fatores de Tempo , Transfecção , Proteínas rho de Ligação ao GTP/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
10.
Arch Biochem Biophys ; 605: 67-75, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27345896

RESUMO

Atmospheric pressure plasma (APP) has been shown effective in sterilization by reducing the number of viable microbes during surface cleaning, food processing, or human tissue treatment. For safe conduct, the majority of previous research focused on complete abolition of microbes, which may require severe treatments. Our aim is to investigate the minimal treatment conditions necessary for effective inactivation of bacteria in such a manner that the APP treated bacteria would not be able to harm the host cells. For this, we ought to identify the objective criteria to make the bacteria dysfunctional. We choose the motile properties and the host-cell invasion capability as two measures to quantify the pathogenic state of bacteria. In this paper, we investigated how the APP treatment in a minimal dosage affects the activity of Salmonella Typhimurium. At 100 W and 15 kHz for 20 s, the APP treatment effectively suppressed active "run and tumble" type motility and induced formation of abnormally long structures. With 20 s exposure, the bacterial cells failed to cause pyroptosis in the host cells with >90% survival after 12 h of co-incubation. Our results suggest novel measures to evaluate the functional pathogenic state for identifying safe APP treatment conditions.


Assuntos
Desinfecção/métodos , Manipulação de Alimentos , Gases em Plasma/química , Salmonella typhimurium/citologia , Pressão Atmosférica , Linhagem Celular Tumoral/microbiologia , Proliferação de Células , Sobrevivência Celular , Humanos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Nitrogênio/química , Fenótipo , Salmonella typhimurium/patogenicidade , Virulência
11.
Soft Matter ; 12(6): 1892-7, 2016 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-26688321

RESUMO

The swimming behavior of a nematode Caenorhabditis elegans (C. elegans) is investigated in a non-Newtonian shear thinning colloidal suspension. At the onset value (ϕ∼ 8%), the suspension begins to exhibit shear thinning behavior, and the average swimming speed of worms jumps by approximately 12% more than that measured in a Newtonian solution exhibiting no shear dependent viscosity. In the shear thinning regime, we observe a gradual yet significant improvement in swimming efficiency with an increase in ϕ while the swimming speed remains nearly constant. We postulate that this enhanced swimming can be explained by the temporal change in the stroke form of the nematode that is uniquely observed in a shear thinning colloidal suspension: the nematode features a fast and large stroke in its head to overcome the temporally high drag imposed by the viscous medium, whose effective viscosity (ηs) is shown to drop drastically, inversely proportional to the strength of its stroke. Our results suggest new insights into how nematodes efficiently maneuver through the complex fluid environment in their natural habitat.


Assuntos
Caenorhabditis elegans/fisiologia , Coloides/química , Natação , Animais , Fenômenos Biomecânicos , Viscosidade
12.
Blood ; 120(13): 2733-44, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22896004

RESUMO

Successful differentiation and expansion of endothelial cells (ECs) from embryonic stem cell (ESC)-derived Flk1(+) mesodermal precursor cells (MPCs) requires supplementation of vascular endothelial growth factor-A (VEGF-A). While analyzing VEGF-A/VEGFR2 downstream signaling pathway that underlies the VEGF-A-induced differentiation and expansion of ECs, we fortuitously found that Rho-associated protein kinase (ROCK) inhibitor Y27632 profoundly promoted the differentiation and expansion of ECs from Flk1(+) MPCs while reducing the differentiation and expansion of mural cells. The ROCK suppression-induced expansion of ECs appears to have resulted from promotion of proliferation of ECs via activation of PI3-kinase-Akt signaling. The ECs obtained by the combination of ROCK suppression and VEGF-A supplementation faithfully expressed most pan-EC surface makers, and phenotypic analyses revealed that they were differentiated toward arterial EC. Further incubation of the ICAM2(+) ECs with Y27632 and VEGF-A for 2 days promoted expansion of ECs by 6.5-fold compared with those incubated with only VEGF-A. Importantly, the ROCK suppression-induced ECs displayed neovasculogenic abilities in vitro and in vivo. Thus, supplementation of ROCK inhibitor Y27632 along with VEGF-A in 2D Matrigel culture system provides a simple, efficient, and versatile method for obtaining ample amount of ESC-derived ECs at high purity suitable for use in therapeutic neovascularization.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Mesoderma/citologia , Neovascularização Fisiológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Amidas/farmacologia , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Imunofluorescência , Laminina/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos , Proteoglicanas/metabolismo , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases Associadas a rho/metabolismo
13.
Exp Dermatol ; 23(3): 172-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24517137

RESUMO

The elasticity of the cellular microenvironment is a key regulator of cellular physiology in many cell types. To investigate the effects of substrate stiffness on the pigmentation process, we cultured normal human melanocytes (NHM) and MNT1 melanoma cells on laminin-coated polydimethylsiloxane (PDMS) substrates of different stiffness. The dendricity of NHM and MNT1 cells was reduced as the substrate stiffness decreased, and the degree of melanosome transfer from NHM or MNT1 cells to normal human keratinocytes was decreased on softer substrates with the reduced dendricity. Gene and protein expressions of MITF, tyrosinase, TRP2, and gp100/PMEL17 exhibited a consistent decreasing trend with the decreasing stiffness. Because the stiffness sensing is mediated by focal adhesion complex through integrin receptors, we checked laminin specific integrin alpha 6 and p-FAK for MNT1 cells to observe that the substrate adhesion was weakened as the substrate stiffness decreased. Weaker adhesion on a softer substrate was accompanied by dynamic shape changes in MNT1 cells with higher speed and larger scattering. Dendritic MNT1 cells cultured on a stiffer substrate exhibited lower migration with smaller root mean squared displacement. These results demonstrate the possibility that skin pigmentation can be influenced by mechanical properties of the cellular microenvironment and can increase when the skin becomes stiff.


Assuntos
Elasticidade , Melanócitos/fisiologia , Melanoma/fisiopatologia , Neoplasias Cutâneas/fisiopatologia , Pigmentação da Pele/fisiologia , Microambiente Tumoral/fisiologia , Adesão Celular , Linhagem Celular Tumoral , Dendritos , Dimetilpolisiloxanos , Expressão Gênica , Humanos , Integrina alfa6/metabolismo , Melaninas/biossíntese , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Melanossomas/fisiologia , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Antígeno gp100 de Melanoma/genética , Antígeno gp100 de Melanoma/metabolismo
14.
APL Bioeng ; 8(2): 021507, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38855445

RESUMO

The understanding of cancer has evolved significantly, with the tumor microenvironment (TME) now recognized as a critical factor influencing the onset and progression of the disease. This broader perspective challenges the traditional view that cancer is primarily caused by mutations, instead emphasizing the dynamic interaction between different cell types and physicochemical factors within the TME. Among these factors, cancer-associated fibroblasts (CAFs) command attention for their profound influence on tumor behavior and patient prognoses. Despite their recognized importance, the biophysical and mechanical interactions of CAFs within the TME remain elusive. This review examines the distinctive physical characteristics of CAFs, their morphological attributes, and mechanical interactions within the TME. We discuss the impact of mechanotransduction on CAF function and highlight how these cells communicate mechanically with neighboring cancer cells, thereby shaping the path of tumor development and progression. By concentrating on the biomechanical regulation of CAFs, this review aims to deepen our understanding of their role in the TME and to illuminate new biomechanical-based therapeutic strategies.

15.
Commun Biol ; 7(1): 115, 2024 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-38245624

RESUMO

A critical requirement for studying cell mechanics is three-dimensional assessment of cellular shapes and forces with high spatiotemporal resolution. Traction force microscopy with fluorescence imaging enables the measurement of cellular forces, but it is limited by photobleaching and a slow acquisition speed. Here, we present refractive-index traction force microscopy (RI-TFM), which simultaneously quantifies the volumetric morphology and traction force of cells using a high-speed illumination scheme with 0.5-Hz temporal resolution. Without labelling, our method enables quantitative analyses of dry-mass distributions and shear (in-plane) and normal (out-of-plane) tractions of single cells on the extracellular matrix. When combined with a constrained total variation-based deconvolution algorithm, it provides 0.55-Pa shear and 1.59-Pa normal traction sensitivity for a 1-kPa hydrogel substrate. We demonstrate its utility by assessing the effects of compromised intracellular stress and capturing the rapid dynamics of cellular junction formation in the spatiotemporal changes in non-planar traction components.


Assuntos
Fenômenos Mecânicos , Tração , Microscopia de Força Atômica/métodos , Algoritmos
16.
Exp Dermatol ; 22(1): 72-4, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23278900

RESUMO

Human dermal fibroblasts (HDFs) regulate wound-healing processes in human skin, including the regeneration of skin sensory fibres, in response to various mechanical stimuli. Because nerve growth factor (NGF) has an essential role in sensory regeneration, we evaluated the possible association of NGF with mechanical stimulus-dependent cellular responses in HDFs. A cyclic tensile stimulus increased both NGF and transforming growth factor (TGF) ß2 production, yet with different gene transcription and signal desensitization profiles. Neutralizing TGFß with antibodies did not affect the tension-induced NGF upregulation, with significant inhibition of endogenous TGFß2 transcription. The treatment with LY294002, SP600125 or U0126 hindered the tension-induced TGFß2 upregulation, although the increase in NGF was regulated only by SP600125 or U0126, indicating the involvement of three signalling kinase pathways in the upregulation of TGFß2. However, the upregulation of NGF was shown to be independent of PI3K, demonstrating the independent regulation of tension-induced NGF and TGFß production in HDFs.


Assuntos
Fibroblastos/metabolismo , Fator de Crescimento Neural/genética , Fator de Crescimento Transformador beta/metabolismo , Antracenos/farmacologia , Anticorpos Neutralizantes/farmacologia , Butadienos/farmacologia , Cromonas/farmacologia , Fibroblastos/efeitos dos fármacos , Humanos , Morfolinas/farmacologia , Fator de Crescimento Neural/metabolismo , Nitrilas/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais , Pele , Estresse Mecânico , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta2/genética , Regulação para Cima/efeitos dos fármacos
17.
ACS Biomater Sci Eng ; 9(1): 280-291, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36573928

RESUMO

Cancer-associated fibroblasts (CAFs) are one of the most prevalent cell types within the tumor microenvironment (TME). While several physicochemical cues from the TME, including growth factors, cytokines, and ECM specificity, have been identified as essential factors for CAF activation, the precise mechanism of how the ECM architecture regulates CAF initiation remains elusive. Using a gelatin-based electrospun fiber mesh, we examined the effect of matrix fiber density on CAF activation induced by MCF-7 conditioned media (CM). A less dense (3D) gelatin mesh matrix facilitated better activation of dermal fibroblasts into a CAF-like phenotype in the CM than a highly dense (3D) gelatin mesh matrix. In addition, it was discovered that CAF activation on the less dense (LD) matrix is dependent on the cell size-related AKT/mTOR signaling cascade, accompanied by an increase in intracellular tension within the well-spread fibroblasts.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Fibroblastos Associados a Câncer/patologia , Gelatina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Transdução de Sinais , Fenótipo
18.
Biomed Mater ; 18(5)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37611612

RESUMO

The field of tissue engineering has been long seeking to develop functional muscle tissue that closely resembles natural muscle. This study used a bio-inspired assembly based on the surface tension mechanism to develop a novel method for engineering muscle tissue. This approach enabled uniaxially ordered electrospun fibers to naturally collide into an aligned bundle without the need for manual handling, thereby reducing cell damage during the cell culture procedure. During the assembly procedure, C2C12 myoblasts were cultured in a viscous collagen hydrogel that caused wetting while providing adequate structural stability for the cell-fiber construct. In addition, gene expression analysis of the resulting muscle-like fibril bundle revealed improved myogenic differentiation. These findings highlight the potential of using a collagen hydrogel and the surface tension mechanism to construct biologically relevant muscle tissue, offering a promising strategy that may outperform existing approaches. Overall, this study contributes to the development of advanced tissue engineering methods and brings us a step closer to creating functional muscle tissue for therapeutic and regenerative medicine applications.


Assuntos
Biomimética , Engenharia Tecidual , Tensão Superficial , Músculos , Hidrogéis
19.
ACS Biomater Sci Eng ; 9(2): 680-692, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36580628

RESUMO

Changes in the extracellular matrix (ECM) influence stem cell fate. When hESCs were differentiated on a thin layer of Matrigel coated onto PDMS (Matrigel_PDMS), they exhibited a substantial increase in focal adhesion and focal adhesion-associated proteins compared with those cultured on Matrigel coated onto TCPS (Matrigel_TCPS), resulting in YAP/TEF1 activation and ultimately promoting the transcriptional activities of pancreatic endoderm (PE)-associated genes. Interestingly, YAP activation in PE cells was mediated through integrin α3-FAK-CDC42-PP1A signaling rather than the typical Hippo signaling pathway. Furthermore, pancreatic islet-like organoids (PIOs) generated on Matrigel_PDMS secreted more insulin than those generated from Matrigel_TCPS. Electron micrographs revealed differential Matrigel architectures depending on the underlying substrate, resulting in varying cell-matrix anchorage resistance levels. Accordingly, the high apparent stiffness of the unique mucus-like network structure of Matrigel_PDMS was the critical factor that directly upregulated focal adhesion, thereby leading to better maturation of the pancreatic development of hESCs in vitro.


Assuntos
Células-Tronco Embrionárias Humanas , Humanos , Diferenciação Celular , Matriz Extracelular/metabolismo , Células-Tronco
20.
Mol Biol Cell ; 33(13): ar129, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36129759

RESUMO

In tissue development and regeneration, the establishment of sharp boundaries between heterotypic cells is essential for the differentiation of tissue functions. During the dynamic rearrangements of constituent cells that result from cell division and collective migration, the segregation boundary encounters various challenges. Several studies have suggested that cortical actomyosin structures play a crucial role in the maintenance of the boundary interface of segregated cell populations, implicating actin-mediated stresses. Examining physical cellular properties such as motility, traction, and intercellular stress, we investigated the formation and maintenance of the stable segregation between epithelial and mesenchymal cell populations devoid of heterotypic adhesions. At the contact boundary, the homotypic adhesion-mediated epithelial aggregates exerted collision-mediated compression against the surrounding mesenchymal cells. Our results demonstrated that heterotypic cell populations established a robust interfacial boundary by accumulating stress from active collisions and repulsions between two dissimilar cell types. Furthermore, the moment of the heterotypic collisions was identified by the existence of a sharp rise in maximum shear stress within the cell cluster.


Assuntos
Actinas , Actomiosina , Separação Celular , Estresse Mecânico , Diferenciação Celular , Adesão Celular , Movimento Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA