Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.944
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancer Discov ; 14(8): 1375-1388, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39091205

RESUMEN

The extracellular matrix (ECM) is an abundant noncellular component of most solid tumors known to support tumor progression and metastasis. The interplay between the ECM and cancer therapeutics opens up new avenues in understanding cancer biology. While the ECM is known to protect the tumor from anticancer agents by serving as a biomechanical barrier, emerging studies show that various cancer therapies induce ECM remodeling, resulting in therapy resistance and tumor progression. This review discusses critical issues in this field including how the ECM influences treatment outcome, how cancer therapies affect ECM remodeling, and the challenges associated with targeting the ECM. Significance: The intricate relationship between the extracellular matrix (ECM) and cancer therapeutics reveals novel insights into tumor biology and its effective treatment. While the ECM may protect tumors from anti-cancer agents, recent research highlights the paradoxical role of therapy-induced ECM remodeling in promoting treatment resistance and tumor progression. This review explores the key aspects of the interplay between ECM and cancer therapeutics.


Asunto(s)
Matriz Extracelular , Neoplasias , Humanos , Matriz Extracelular/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Microambiente Tumoral , Animales
2.
Methods Mol Biol ; 2805: 113-124, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39008177

RESUMEN

The extracellular matrix (ECM) provides dynamic structural and molecular signals that affect the form and function of developing tissues. In order to parse how the individual features of the ECM impact cell- and tissue-level behavior during development, engineered culture models should reproduce key structural and molecular features of native ECM. Here, we describe a protocol for bioprinting epithelial cell aggregates embedded within a collagen-Matrigel ink in order to study the dynamic interplay between epithelial tissues and aligned networks of type I collagen fibers. Collagen fiber alignment and geometry can be spatially controlled by modulating the printing speed, nozzle geometry, surface chemistry, and degree of molecular crowding in the printing ink. We provide detailed procedures for generating epithelial cell aggregates, microextrusion printing collagen-Matrigel bioinks, culturing the three-dimensional (3D)-printed tissues, and imaging 3D-printed collagen-Matrigel constructs.


Asunto(s)
Bioimpresión , Colágeno , Células Epiteliales , Matriz Extracelular , Hidrogeles , Impresión Tridimensional , Ingeniería de Tejidos , Bioimpresión/métodos , Hidrogeles/química , Colágeno/química , Colágeno/metabolismo , Ingeniería de Tejidos/métodos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Animales , Morfogénesis , Humanos , Proteoglicanos/química , Proteoglicanos/metabolismo , Andamios del Tejido/química , Laminina/química , Combinación de Medicamentos , Perros , Epitelio/metabolismo , Epitelio/crecimiento & desarrollo
3.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 663-671, 2024 Jul 09.
Artículo en Chino | MEDLINE | ID: mdl-38949134

RESUMEN

Objective: To investigate the characteristics of extracellular matrix vesicle mimetics prepared by mechanical extrusion and their effects on the cell viability and osteogenic differentiation potential of human periodontal ligament stem cells (PDLSC). Methods: PDLSC derived extracellular matrix vesicles were prepared by collagenase digestion, while the cell derived vesicle mimetics were simulated by mechanical extrusion. The obtained extracellular matrix vesicles and parental cell derived vesicle mimetics were divided into 4 groups: matrix vesicles derived from PDLSC cultured in basic medium for 7 days (PDLSC matrix vesicles, MVs), vesicle mimetics derived from PDLSC cultured in basic medium for 7 days (PDLSC vesicle mimetics, CVMs), matrix vesicles derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC matrix vesicles, O-MVs) and vesicle mimetics derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC vesicle mimetics, O-CVMs). Vesicles morphologies and sizes were observed by transmission electron microscopy and nanoparticle tracking analysis. Vesicles uptake was detected by immunofluorescence. With PDLSC as the control group, the effects of vesicles on the viability of PDLSC were detected by cell activity assay (cell counting kit-8), and the effects of vesicles on the osteogenic differentiation potential of PDLSC were detected by alizarin red staining and Western blotting. Results: Vesicles in MVs, O-MVs, CVMs and O-CVMs were all observed with a round structure (size 50-250 nm), and could be taken up by PDLSC without affecting the cell viability. Under osteogenic inducing conditions, PDLSC incubated with O-MVs or O-CVMs could produce more mineralized nodules than those in the control group (PDLSC). MVs, O-MVs, CVMs and O-CVMs could promote the expression of osteogenic-related proteins in PDLSC. PDLSC in group O-CVMs showed significant higher expressions of osteogenic-related proteins, including alkaline phosphatase (ALP) (1.571±0.348), osteopontin (OPN) (1.827±0.627) and osteocalcin (OCN) (1.798±0.537) compared to MVs (ALP: 1.156±0.170, OPN: 1.260±0.293, OCN: 1.286±0.302) (P<0.05). Compared to CMVs-incubated PDLSC, O-CVMs-incubated PDLSC expressed more Runt-related transcription factor 2 (1.632±0.455 vs 1.176±0.128) and OPN (1.827±0.627 vs 1.428±0.427) (P<0.05). Moreover, there was no significant difference in the expression levels of osteoblast-related proteins in PDLSC cultured with MVs, O-MVs and CVMs (P>0.05). Conclusions: The vesicle mimetics prepared by mechanical extrusion method are similar in shape and size to the extracellular matrix vesicles. MVs, O-MVs, CVMs and O-CVMs do not affect the cell viability of PDLSC, and can promote the osteogenic differentiation potential of PDLSC to a certain extent.


Asunto(s)
Diferenciación Celular , Matriz Extracelular , Vesículas Extracelulares , Osteogénesis , Humanos , Matriz Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre/citología , Fosfatasa Alcalina/metabolismo , Ligamento Periodontal/citología , Ligamento Periodontal/metabolismo , Osteocalcina/metabolismo , Osteopontina/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo
4.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000302

RESUMEN

Dupuytren's disease (DD) is a prevalent fibroproliferative disorder of the hand, shaped by genetic, epigenetic, and environmental influences. The extracellular matrix (ECM) is a complex assembly of diverse macromolecules. Alterations in the ECM's content, structure and organization can impact both normal physiological functions and pathological conditions. This study explored the content and organization of glycosaminoglycans, proteoglycans, and collagen in the ECM of patients at various stages of DD, assessing their potential as prognostic indicators. This research reveals, for the first time, relevant changes in the complexity of chondroitin/dermatan sulfate structures, specifically an increase of disaccharides containing iduronic acid residues covalently linked to either N-acetylgalactosamine 6-O-sulfated or N-acetylgalactosamine 4-O-sulfated, correlating with the disease's severity. Additionally, we noted an increase in versican expression, a high molecular weight proteoglycan, across stages I to IV, while decorin, a small leucine-rich proteoglycan, significantly diminishes as DD progresses, both confirmed by mRNA analysis and protein detection via confocal microscopy. Coherent anti-Stokes Raman scattering (CARS) microscopy further demonstrated that collagen fibril architecture in DD varies importantly with disease stages. Moreover, the urinary excretion of both hyaluronic and sulfated glycosaminoglycans markedly decreased among DD patients.Our findings indicate that specific proteoglycans with galactosaminoglycan chains and collagen arrangements could serve as biomarkers for DD progression. The reduction in glycosaminoglycan excretion suggests a systemic manifestation of the disease.


Asunto(s)
Colágeno , Decorina , Contractura de Dupuytren , Proteoglicanos , Humanos , Contractura de Dupuytren/metabolismo , Contractura de Dupuytren/patología , Colágeno/metabolismo , Proteoglicanos/metabolismo , Decorina/metabolismo , Matriz Extracelular/metabolismo , Masculino , Progresión de la Enfermedad , Femenino , Dermatán Sulfato/metabolismo , Persona de Mediana Edad , Anciano , Versicanos/metabolismo , Versicanos/genética , Glicosaminoglicanos/metabolismo , Sulfatos de Condroitina/metabolismo , Polisacáridos
5.
Int J Mol Sci ; 25(13)2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-39000333

RESUMEN

Breast cancer represents the most prevalent form of cancer and the leading cause of cancer-related mortality among females worldwide. It has been reported that several risk factors contribute to the appearance and progression of this disease. Despite the advancements in breast cancer treatment, a significant portion of patients with distant metastases still experiences no cure. The extracellular matrix represents a potential target for enhanced serum biomarkers in breast cancer. Furthermore, extracellular matrix degradation and epithelial-mesenchymal transition constitute the primary stages of local invasion during tumorigenesis. Additionally, the microbiome has a potential influence on diverse physiological processes. It is emerging that microbial dysbiosis is a significant element in the development and progression of various cancers, including breast cancer. Thus, a better understanding of extracellular matrix and microbiome interactions could provide novel alternatives to breast cancer treatment and management. In this review, we summarize the current evidence regarding the intricate relationship between breast cancer with the extracellular matrix and the microbiome. We discuss the arising associations and future perspectives in this field.


Asunto(s)
Neoplasias de la Mama , Matriz Extracelular , Microbiota , Humanos , Neoplasias de la Mama/microbiología , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Matriz Extracelular/metabolismo , Femenino , Transición Epitelial-Mesenquimal , Animales , Disbiosis/microbiología , Microambiente Tumoral
6.
Nat Commun ; 15(1): 5795, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987250

RESUMEN

Animals protect themself from microbial attacks by robust skins or a cuticle as in Caenorhabditis elegans. Nematode-trapping fungi, like Arthrobotrys flagrans, overcome the cuticle barrier and colonize the nematode body. While lytic enzymes are important for infection, small-secreted proteins (SSPs) without enzymatic activity, emerge as crucial virulence factors. Here, we characterized NipA (nematode induced protein) which A. flagrans secretes at the penetration site. In the absence of NipA, A. flagrans required more time to penetrate C. elegans. Heterologous expression of the fungal protein in the epidermis of C. elegans led to blister formation. NipA contains 13 cysteines, 12 of which are likely to form disulfide bridges, and the remaining cysteine was crucial for blister formation. We hypothesize that NipA interferes with cuticle integrity to facilitate fungal entry. Genome-wide expression analyses of C. elegans expressing NipA revealed mis-regulation of genes associated with extracellular matrix (ECM) maintenance and innate immunity.


Asunto(s)
Ascomicetos , Caenorhabditis elegans , Cisteína , Proteínas Fúngicas , Factores de Virulencia , Animales , Caenorhabditis elegans/microbiología , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Cisteína/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Ascomicetos/patogenicidad , Ascomicetos/genética , Ascomicetos/metabolismo , Inmunidad Innata , Matriz Extracelular/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Epidermis/metabolismo , Epidermis/microbiología
7.
Nat Commun ; 15(1): 5791, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987295

RESUMEN

Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.


Asunto(s)
Matriz Extracelular , Células Madre Hematopoyéticas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Nestina , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Nestina/metabolismo , Nestina/genética , Matriz Extracelular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Nicho de Células Madre , Hidrogeles/química , Bioingeniería/métodos , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Trasplante de Células Madre Hematopoyéticas , Antígenos CD34/metabolismo , Colágeno Tipo I/metabolismo , Diferenciación Celular , Ratones Endogámicos C57BL
8.
Cell Death Dis ; 15(7): 493, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987529

RESUMEN

Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.


Asunto(s)
Adenocarcinoma del Pulmón , Colágeno Tipo V , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares , Miofibroblastos , Estrés Mecánico , Humanos , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/tratamiento farmacológico , Miofibroblastos/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Colágeno Tipo V/metabolismo , Colágeno Tipo V/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Línea Celular Tumoral , Animales , Movimiento Celular/efectos de los fármacos , Metástasis de la Neoplasia , Ratones , Microambiente Tumoral , Sorafenib/farmacología , Sorafenib/uso terapéutico , Matriz Extracelular/metabolismo
9.
Discov Med ; 36(186): 1441-1452, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39054715

RESUMEN

BACKGROUND: Synovial inflammation plays a crucial role in osteoarthritis (OA). Gastrodin (GAS), an active ingredient derived from the Gastrodia elata Blume rhizome, possesses antioxidant and anti-inflammatory pharmacological effects. This research aimed to evaluate the function and molecular mechanism of GAS on human fibroblast-like synoviocytes of osteoarthritis (HFLS-OA) induced by interleukin (IL)-1ß. METHODS: The impact of GAS on the viability of IL-1ß-treated HFLS-OA cells was assessed using the cell counting kit-8 (CCK-8). Quantitative real-time reverse transcription PCR (qRT-PCR) was employed to detect changes in IL-8, IL-6, monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and Gremlin-1 mRNA expression in each group. Corresponding kits were utilized to measure the catalase (CAT) and superoxide dismutase (SOD) activities, as well as the nitric oxide (NO) level. Western blot analysis was conducted to examine the expression of extracellular matrix degradation-associated proteins and nuclear factor kappa-B (NF-κB) pathway-correlated proteins in each group. RESULTS: GAS significantly promoted the proliferation of IL-1ß-induced HFLS-OA cells and concurrently down-regulated Gremlin-1 mRNA expression (p < 0.05). Through the down-regulation of Gremlin-1 expression, GAS exhibited the following effects: decreased IL-8, IL-6, and TNF-α mRNA expression, as well as NO levels (p < 0.05); increased SOD and CAT activities (p < 0.05); down-regulated matrix metallopeptidase 13 (MMP-13) and MMP-1 protein expression levels (p < 0.01); and up-regulated collagen II protein expression level (p < 0.01) in IL-1ß-treated HFLS-OA cells. Additionally, GAS decreased phospho-inhibitory kappa B (p-IκB)/IκB, phospho-inhibitory kappa B kinase (p-IKK)/IKK, and p-p65/p65 ratios in IL-1ß-induced HFLS-OA cells by inhibiting Gremlin-1 expression (p < 0.01). CONCLUSION: GAS demonstrates a positive impact on inflammation, oxidative stress, and extracellular matrix degradation in IL-1ß-mediated HFLS-OA cells. This effect is achieved by suppressing Gremlin-1 expression and reducing NF-κB pathway activity.


Asunto(s)
Alcoholes Bencílicos , Matriz Extracelular , Glucósidos , Inflamación , Interleucina-1beta , FN-kappa B , Estrés Oxidativo , Sinoviocitos , Humanos , Glucósidos/farmacología , Interleucina-1beta/metabolismo , Alcoholes Bencílicos/farmacología , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Sinoviocitos/patología , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Osteoartritis/patología , Osteoartritis/metabolismo , Osteoartritis/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intercelular
10.
J Nanobiotechnology ; 22(1): 438, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39061089

RESUMEN

Decellularized extracellular matrix hydrogel (ECM hydrogel), a natural material derived from normal tissue with unique biocompatibility properties, is widely used for tissue repair. However, there are still problems such as poor biological activity and insufficient antimicrobial property. To overcome these drawbacks, fibroblast growth factor 2 (FGF 2) containing exosome (exoFGF 2) was prepared to increase the biological activity. Furthermore, the antimicrobial capacity of ECM hydrogel was optimised by using copper ions as a ligand-bonded cross-linking agent. The decellularized extracellular matrix hydrogel, intricately cross-linked with copper ions through ligand bonds and loaded with FGF 2 containing exosome (exoFGF 2@ECM/Cu2+ hydrogel), has demonstrated exceptional biocompatibility and antimicrobial properties. In vitro, exoFGF 2@ECM/Cu2+ hydrogel effectively promoted cell proliferation, migration, antioxidant and inhibited bacterial growth. In vivo, the wound area of rat treated with exoFGF 2@ECM/Cu2+ hydrogels were significantly smaller than that of other groups at Day 5 (45.24% ± 3.15%), Day 10 (92.20% ± 2.31%) and Day 15 (95.22% ± 1.28%). Histological examination showed that exoFGF 2@ECM/Cu2+ hydrogels promoted angiogenesis and collagen deposition. Overall, this hydrogel has the potential to inhibit bacterial growth and effectively promote wound healing in a variety of clinical applications.


Asunto(s)
Proliferación Celular , Exosomas , Matriz Extracelular , Factor 2 de Crecimiento de Fibroblastos , Hidrogeles , Piel , Cicatrización de Heridas , Hidrogeles/química , Hidrogeles/farmacología , Animales , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/química , Exosomas/química , Exosomas/metabolismo , Ratas , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Piel/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratas Sprague-Dawley , Humanos , Cobre/química , Cobre/farmacología , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Masculino , Ratones , Movimiento Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos
11.
Int J Mol Sci ; 25(14)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39062846

RESUMEN

Hyaluronan (HA) has gained significant attention in cancer research for its role in modulating chemoresistance. This review aims to elucidate the mechanisms by which HA contributes to chemoresistance, focusing on its interactions within the tumor microenvironment. HA is abundantly present in the extracellular matrix (ECM) and binds to cell-surface receptors such as CD44 and RHAMM. These interactions activate various signaling pathways, including PI3K/Akt, MAPK, and NF-κB, which are implicated in cell survival, proliferation, and drug resistance. HA also influences the physical properties of the tumor stroma, enhancing its density and reducing drug penetration. Additionally, HA-mediated signaling contributes to the epithelial-mesenchymal transition (EMT), a process associated with increased metastatic potential and resistance to apoptosis. Emerging therapeutic strategies aim to counteract HA-induced chemoresistance by targeting HA synthesis, degradation, metabolism, or its binding to CD44. This review underscores the complexity of HA's role in chemoresistance and highlights the potential for HA-targeted therapies to improve the efficacy of conventional chemotherapeutics.


Asunto(s)
Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Matriz Extracelular , Ácido Hialurónico , Neoplasias , Transducción de Señal , Microambiente Tumoral , Humanos , Ácido Hialurónico/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Matriz Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Animales
12.
Biomed Pharmacother ; 177: 117051, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38959608

RESUMEN

Due to the limitations of the current skin wound treatments, it is highly valuable to have a wound healing formulation that mimics the extracellular matrix (ECM) and mechanical properties of natural skin tissue. Here, a novel biomimetic hydrogel formulation has been developed based on a mixture of Agarose-Collagen Type I (AC) combined with skin ECM-related components: Dermatan sulfate (DS), Hyaluronic acid (HA), and Elastin (EL) for its application in skin tissue engineering (TE). Different formulations were designed by combining AC hydrogels with DS, HA, and EL. Cell viability, hemocompatibility, physicochemical, mechanical, and wound healing properties were investigated. Finally, a bilayered hydrogel loaded with fibroblasts and mesenchymal stromal cells was developed using the Ag-Col I-DS-HA-EL (ACDHE) formulation. The ACDHE hydrogel displayed the best in vitro results and acceptable physicochemical properties. Also, it behaved mechanically close to human native skin and exhibited good cytocompatibility. Environmental scanning electron microscopy (ESEM) analysis revealed a porous microstructure that allows the maintenance of cell growth and ECM-like structure production. These findings demonstrate the potential of the ACDHE hydrogel formulation for applications such as an injectable hydrogel or a bioink to create cell-laden structures for skin TE.


Asunto(s)
Materiales Biomiméticos , Hidrogeles , Ingeniería de Tejidos , Hidrogeles/química , Humanos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Ingeniería de Tejidos/métodos , Supervivencia Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Cicatrización de Heridas/efectos de los fármacos , Colágeno Tipo I/metabolismo , Piel/efectos de los fármacos , Piel/metabolismo , Dermatán Sulfato/química , Dermatán Sulfato/farmacología , Fibroblastos/efectos de los fármacos , Elastina/química , Matriz Extracelular/metabolismo , Biomimética/métodos , Sefarosa/química , Dermis/efectos de los fármacos , Dermis/metabolismo , Dermis/citología , Animales
13.
Ann Clin Lab Sci ; 54(3): 363-370, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39048163

RESUMEN

OBJECTIVE: During the progression of chronic idiopathic pulmonary fibrosis (IPF), maladaptive tissue remodeling including excessive extracellular matrix (ECM) deposition occurs, which eventually leads to architectural distortion and loss of organ function in organ fibrosis. ADAM15, which is highly expressed in the developing lungs and kidneys, is a transmembrane-anchored multidomain protein belonging to the family of metalloproteinases. Compared to the extensive studies about functions of matrix metalloproteinases (MMPs), less are discussed about ADAM15, particularly in function and mechanism involving fibrogenesis. Our study aims to fill in this gap. METHODS: We identified ADAM15 as a novel antifibrotic mediator in lung fibrosis. We found that ADAM15 has cross-talks with transforming growth factor-ß1 (TGF-ß1), which is the most potent profibrotic mediator. We provided molecular and translational evidence that knockdown of ADAM15 accelerated fibrogenic response induced by TGF-ß1 and upregulation of ADAM15 rescued TGF-ß1-induced myofibroblast activation in part. RESULTS: Overexpression of ADAM15 ameliorates fibrotic changes and ADAM15 deficiency exacerbates changes from fibroblast to myofibroblast in NIH/3T3. Results were also presented and identified by the intuitive immunofluorescence staining. CONCLUSION: In this study, we uncover a new molecular mechanism of tissue fibrogenesis and identify ADAM15 as a potential therapeutic target in the treatment of fibrotic diseases.


Asunto(s)
Proteínas ADAM , Matriz Extracelular , Fibroblastos , Proteínas de la Membrana , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Ratones , Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas ADAM/metabolismo , Proteínas ADAM/genética , Matriz Extracelular/metabolismo , Humanos , Células 3T3 NIH , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/metabolismo
14.
Carbohydr Polym ; 342: 122363, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39048221

RESUMEN

The tumor microenvironment (TME) orchestrates cellular and extracellular matrix (ECM) interactions, playing a key role in tumorigenesis, tumor growth, and metastization. Investigating the interplay between stromal-epithelial cells within the TME is paramount for understanding cancer mechanisms but demands reliable biological models. 3D-models have emerged as powerful in vitro tools, but many fall short in replicating cell-cell/cell-matrix interactions. This study introduces a novel hybrid 3D-model of the breast TME, combining epithelial cells, cancer-associated fibroblasts (CAFs), and their ECM. To build the stromal compartment, porous 3D-printed alginate scaffolds were seeded with CAFs, which proliferated and produced ECM. The pores were infused with oxidized peptide-modified alginate hydrogel laden with MCF10A cells, forming the parenchymal compartment. The hybrid system supported epithelial morphogenesis into acini surrounded by fibroblasts and ECM, and could be readily solubilized to recover cells, their matrix, and sequestered soluble factors. Proteome profiling of the retrieved ECM showed upregulation of proteins associated with matrix assembly/remodeling, epithelial-to-mesenchymal transition (EMT), and cancer. The TME-like microenvironment induced a partial EMT in MCF10A cells, generating a hybrid population with epithelial and mesenchymal features, characteristic of aggressive phenotypes. Our model provided new insights into epithelial-stromal interactions within the TME, offering a valuable tool for cancer research in a physiologically-relevant 3D setting.


Asunto(s)
Alginatos , Neoplasias de la Mama , Células Epiteliales , Matriz Extracelular , Microambiente Tumoral , Humanos , Alginatos/química , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Matriz Extracelular/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/metabolismo , Hidrogeles/química , Impresión Tridimensional , Andamios del Tejido/química , Técnicas de Cultivo Tridimensional de Células/métodos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos
15.
Physiol Res ; 73(3): 415-426, 2024 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-39027958

RESUMEN

Recent research has identified that miR-539-3p impedes chondrogenic differentiation, yet its specific role and underlying mechanisms in childhood-onset osteoarthritis (OA) remain unclear. This study found that miR-539-3p levels were considerably lower in cartilage samples derived from childhood-onset OA patients compared to the control group. Enhancing miR-539-3p expression or suppressing RUNX2 expression notably reduced apoptosis, inflammation, and extracellular matrix (ECM) degradation in OA chondrocytes. In contrast, reducing miR-539-3p or increasing RUNX2 had the opposite effects. RUNX2 was confirmed as a direct target of miR-539-3p. Further experiments demonstrated that miR-539-3p targeting RUNX2 effectively lessened apoptosis, inflammation, and ECM degradation in OA chondrocytes, accompanied by changes in key molecular markers like reduced caspase-3 and matrix etallopeptidase 13 (MMP-13) levels, and increased B-cell lymphoma 2 (Bcl-2) and collagen type X alpha 1 chain (COL2A1). This study underscores the pivotal role of miR-539-3p in alleviating inflammation and ECM degradation in childhood-onset OA through targeting RUNX2, offering new insights for potential therapeutic strategies against this disease.


Asunto(s)
Apoptosis , Condrocitos , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Matriz Extracelular , MicroARNs , Osteoartritis , Humanos , MicroARNs/metabolismo , MicroARNs/genética , Condrocitos/metabolismo , Condrocitos/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/genética , Niño , Masculino , Femenino , Células Cultivadas , Adolescente
16.
Sci Rep ; 14(1): 16897, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043893

RESUMEN

The chemokine (CCL)-chemokine receptor (CCR2) interaction, importantly CCL2-CCR2, involved in the intrahepatic recruitment of monocytes upon liver injury promotes liver fibrosis. CCL2-CCR2 antagonism using Cenicriviroc (CVC) showed promising results in several preclinical studies. Unfortunately, CVC failed in phase III clinical trials due to lack of efficacy to treat liver fibrosis. Lack of efficacy could be attributed to the fact that macrophages are also involved in disease resolution by secreting matrix metalloproteinases (MMPs) to degrade extracellular matrix (ECM), thereby inhibiting hepatic stellate cells (HSCs) activation. HSCs are the key pathogenic cell types in liver fibrosis that secrete excessive amounts of ECM causing liver stiffening and liver dysfunction. Knowing the detrimental role of intrahepatic monocyte recruitment, ECM, and HSCs activation during liver injury, we hypothesize that combining CVC and MMP (MMP1) could reverse liver fibrosis. We evaluated the effects of CVC, MMP1 and CVC + MMP1 in vitro and in vivo in CCl4-induced liver injury mouse model. We observed that CVC + MMP1 inhibited macrophage migration, and TGF-ß induced collagen-I expression in fibroblasts in vitro. In vivo, MMP1 + CVC significantly inhibited normalized liver weights, and improved liver function without any adverse effects. Moreover, MMP1 + CVC inhibited monocyte infiltration and liver inflammation as confirmed by F4/80 and CD11b staining, and TNFα gene expression. MMP1 + CVC also ameliorated liver fibrogenesis via inhibiting HSCs activation as assessed by collagen-I staining and collagen-I and α-SMA mRNA expression. In conclusion, we demonstrated that a combination therapeutic approach by combining CVC and MMP1 to inhibit intrahepatic monocyte recruitment and increasing collagen degradation respectively ameliorate liver inflammation and fibrosis.


Asunto(s)
Matriz Extracelular , Células Estrelladas Hepáticas , Cirrosis Hepática , Metaloproteinasa 1 de la Matriz , Monocitos , Animales , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 1 de la Matriz/genética , Ratones , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Matriz Extracelular/metabolismo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hígado/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Tetracloruro de Carbono , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Humanos , Movimiento Celular/efectos de los fármacos , Sinergismo Farmacológico , Imidazoles , Sulfóxidos
17.
Mol Biol Cell ; 35(8): ar114, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38985514

RESUMEN

Idiopathic pulmonary fibrosis (IPF), one of the most common forms of interstitial lung disease, is a poorly understood, chronic, and often fatal fibroproliferative condition with only two FDA-approved medications. Understanding the pathobiology of the fibroblast in IPF is critical to evaluating and discovering novel therapeutics. Using a decellularized lung matrix derived from patients with IPF, we generate three-dimensional hydrogels as in vitro models of lung physiology and characterize the phenotype of fibroblasts seeded into the hydrogels. When cultured in IPF extracellular matrix hydrogels, IPF fibroblasts display differential contractility compared with their normal counterparts, lose the classical myofibroblast marker α-smooth muscle actin, and increase expression of proinflammatory cytokines compared with fibroblasts seeded two-dimensionally on tissue culture dishes. We validate this proinflammatory state in fibroblast-conditioned media studies with monocytes and monocyte-derived macrophages. These findings add to a growing understanding of the lung microenvironment effect on fibroblast phenotypes, shed light on the potential role of fibroblasts as immune signaling hubs during lung fibrosis, and suggest intervention in fibroblast-immune cell cross-talk as a possible novel therapeutic avenue.


Asunto(s)
Matriz Extracelular , Fibroblastos , Hidrogeles , Fibrosis Pulmonar Idiopática , Pulmón , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibroblastos/metabolismo , Pulmón/patología , Pulmón/metabolismo , Matriz Extracelular/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Miofibroblastos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Actinas/metabolismo , Monocitos/metabolismo
18.
Methods Mol Biol ; 2811: 207-220, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39037661

RESUMEN

Tumor cells often leave the primary tumor mass and get settled in a foreign tissue years before the development of overt metastases, exhibiting the highly inefficient nature of metastatic colony formation. In fact, the tumor cells that disseminate into distant organs and subsequently invade the parenchyma of these organs rarely proceed to found actively growing metastatic colonies. Instead, the majority of these tumor cells undergo prolonged proliferative arrest unless they are swiftly eliminated by the immune system. Together, these observations indicate that the proliferative capacity of the disseminated tumor cells (DTCs) serves as a key determinant of the efficiency of metastasis, highlighting the need to better understand the mechanism governing the proliferation of these cells. Recent studies are unveiling the importance of the interactions between DTCs and the microenvironment of the host tissue in regulating the proliferation of DTCs. However, the details of such interactions remain to be fully delineated. Here I describe the methods for visualizing and analyzing the interactions between DTCs and the extracellular matrix (ECM) components of the host tissue as well as the cytoskeleton of the DTCs that support these interactions. The methods described here will facilitate the study of how DTCs interact with the ECM of their host tissue, which will be crucial for elucidating the mechanism that underlies the regulation of DTC proliferation by the DTC-ECM interactions.


Asunto(s)
Citoesqueleto , Matriz Extracelular , Citoesqueleto/metabolismo , Humanos , Matriz Extracelular/metabolismo , Animales , Línea Celular Tumoral , Microambiente Tumoral , Ratones , Células Neoplásicas Circulantes/patología , Células Neoplásicas Circulantes/metabolismo , Proliferación Celular , Neoplasias/patología , Neoplasias/metabolismo , Metástasis de la Neoplasia , Uniones Célula-Matriz/metabolismo
19.
Sci Rep ; 14(1): 15022, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951570

RESUMEN

Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/ß-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-ß3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-ß3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Condrogénesis , Hidrogeles , Células Madre Mesenquimatosas , Ingeniería de Tejidos , Proteína Wnt3A , Proteína Wnt3A/metabolismo , Condrogénesis/efectos de los fármacos , Ingeniería de Tejidos/métodos , Proliferación Celular/efectos de los fármacos , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Humanos , Cartílago/metabolismo , Gelatina/química , Andamios del Tejido/química , Factor de Crecimiento Transformador beta3/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , Línea Celular , Matriz Extracelular/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/citología , Animales
20.
Lab Chip ; 24(15): 3718-3727, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38953554

RESUMEN

The in vitro recapitulation of tumor microenvironment is of great interest to preclinical screening of drugs. Compared with culture of cell lines, tumor organ slices can better preserve the complex tumor architecture and phenotypic activity of native cells, but are limited by their exposure to fluid shear and gradual degradation under perfusion culture. Here, we established a decellularized liver matrix (DLM)-GelMA "sandwich" structure and a perfusion-based microfluidic platform to support long-term culture of tumor slices with excellent structural integrity and cell viability over 7 days. The DLM-GelMA was able to secrete cytokines and growth factors while providing shear protection to the tumor slice via the sandwich structure, leading to the preservation of the tumor microenvironment where immune cells (CD3, CD8, CD68), tumor-associated fibroblasts (α-SMA), and extracellular matrix components (collagen I, fibronectin) were well maintained. Furthermore, this chip presented anti-tumor efficacy at cisplatin (20 µM) on tumor patients, demonstrating our platform's efficacy to design patient-specific treatment regimens. Taken together, the successful development of this DLM-GelMA sandwich structure on the chip could faithfully reflect the tumor microenvironment and immune response, accelerating the screening process of drug molecules and providing insights for practical medicine.


Asunto(s)
Dispositivos Laboratorio en un Chip , Microambiente Tumoral , Humanos , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Cisplatino/farmacología , Cisplatino/química , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Animales , Hígado/metabolismo , Hígado/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA