Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 321
Filtrar
1.
Nat Commun ; 15(1): 4015, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740766

RESUMEN

Microfibril-associated glycoprotein 4 (MFAP4) is a 36-kDa extracellular matrix glycoprotein with critical roles in organ fibrosis, chronic obstructive pulmonary disease, and cardiovascular disorders, including aortic aneurysms. MFAP4 multimerises and interacts with elastogenic proteins, including fibrillin-1 and tropoelastin, and with cells via integrins. Structural details of MFAP4 and its potential interfaces for these interactions are unknown. Here, we present a cryo-electron microscopy structure of human MFAP4. In the presence of calcium, MFAP4 assembles as an octamer, where two sets of homodimers constitute the top and bottom halves of each octamer. Each homodimer is linked together by an intermolecular disulphide bond. A C34S missense mutation prevents disulphide-bond formation between monomers but does not prevent octamer assembly. The atomic model, built into the 3.55 Å cryo-EM map, suggests that salt-bridge interactions mediate homodimer assembly, while non-polar residues form the interface between octamer halves. In the absence of calcium, an MFAP4 octamer dissociates into two tetramers. Binding studies with fibrillin-1, tropoelastin, LTBP4, and small fibulins show that MFAP4 has multiple surfaces for protein-protein interactions, most of which depend upon MFAP4 octamer assembly. The C34S mutation does not affect these protein interactions or cell interactions. MFAP4 assemblies with fibrillin-1 abrogate MFAP4 interactions with cells.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de la Matriz Extracelular , Fibrilina-1 , Microfibrillas , Tropoelastina , Humanos , Adipoquinas , Calcio/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Fibrilina-1/metabolismo , Fibrilina-1/genética , Fibrilina-1/química , Glicoproteínas , Células HEK293 , Microfibrillas/metabolismo , Microfibrillas/química , Microfibrillas/ultraestructura , Modelos Moleculares , Mutación Missense , Unión Proteica , Multimerización de Proteína , Tropoelastina/metabolismo , Tropoelastina/química , Tropoelastina/genética
2.
Biomater Adv ; 161: 213894, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38796956

RESUMEN

Engineering of scaffolds for bone regeneration is often inspired by the native extracellular matrix mimicking its composite fibrous structure. In the present study, we used low loadings of diatomite earth (DE) biosilica to improve the bone regeneration potential of gelatin electrospun fibrillar microenvironments. We explored the effect of increasing the DE content from 1 % to 3 % and 5 %, respectively, on the physico-chemical properties of the fibrous scaffolds denoted FG_DE1, FG_DE3, FG_DE5, regarding the aqueous media affinity, stability under simulated physiological conditions, morphology characteristics, and local mechanical properties at the surface. The presence of biosilica generated composite structures with lower swelling degrees and higher stiffness when compared to gelatin fibers. Increasing DE content led to higher Young modulus, while the stability of the protein matrix in PBS, at 37 °C, over 21 was significantly decreased by the presence of diatomite loadings. The best preosteoblast response was obtained for FG_DE3, with enhanced mineralization during the osteogenic differentiation when compared to the control sample without diatomite. 5 % DE in FG_DE5 proved to negatively influence cells' metabolic activity and morphology. Hence, the obtained composite microfibrillar scaffolds might find application as osteoblast-responsive materials for bone tissue engineering.


Asunto(s)
Gelatina , Osteoblastos , Ingeniería de Tejidos , Andamios del Tejido , Gelatina/química , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Andamios del Tejido/química , Ingeniería de Tejidos/métodos , Animales , Tierra de Diatomeas/química , Osteogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratones , Regeneración Ósea/efectos de los fármacos , Línea Celular , Microambiente Celular/efectos de los fármacos , Microfibrillas/química , Microfibrillas/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos
3.
J Cell Mol Med ; 27(21): 3235-3246, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37635348

RESUMEN

Liver fibrosis is a chronic inflammatory process characterized by the accumulation of extracellular matrix (ECM), which contributes to cirrhosis and hepatocellular carcinoma. Increasing evidence suggests that the activation of hepatic stellate cells (HSCs) under an inflammatory state leads to the secretion of collagens, which can cause cirrhosis. In this study, we analysed data from the Gene Expression Omnibus (GEO) databases to identify differentially expressed genes (DEGs) between quiescent and fibrotic HSCs. We found that Microfibril Associated Protein 2 (MFAP2) was elevated in carbon tetrachloride (CCl4)-induced liver fibrosis and Transforming Growth Factor-Beta 1 (TGF-ß1)-activated HSCs. Knockdown of MFAP2 inhibited HSC proliferation and partially attenuated TGF-ß-stimulated fibrogenesis markers. Bioinformatics analysis revealed that Fibrillin-1 (FBN1) was correlated with MFAP2, and the expression of FBN1 was significantly upregulated after MFAP2 overexpression. Silencing MFAP2 partially attenuated the activation of HSCs by inhibiting HSC proliferation and decreasing collagen deposits. In vitro results showed that the inhibition of MFAP2 alleviated hepatic fibrosis by inhibiting the activation and inducing the apoptosis of active HSCs in a CCl4-induced mouse model. In conclusion, our results suggest that MFAP2 is a potential target for the clinical treatment of liver fibrosis.


Asunto(s)
Microfibrillas , Factor de Crecimiento Transformador beta , Animales , Ratones , Tetracloruro de Carbono/toxicidad , Fibrilina-1/genética , Fibrilina-1/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Microfibrillas/metabolismo , Microfibrillas/patología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
4.
Oncogene ; 42(25): 2061-2073, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37156839

RESUMEN

Highly desmoplastic and immunosuppressive tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) contributes to tumor progression and resistance to current therapies. Clues targeting the notorious stromal environment have offered hope for improving therapeutic response whereas the underlying mechanism remains unclear. Here, we find that prognostic microfibril associated protein 5 (MFAP5) is involved in activation of cancer-associated fibroblasts (CAFs). Inhibition of MFAP5highCAFs shows synergistic effect with gemcitabine-based chemotherapy and PD-L1-based immunotherapy. Mechanistically, MFAP5 deficiency in CAFs downregulates HAS2 and CXCL10 via MFAP5/RCN2/ERK/STAT1 axis, leading to angiogenesis, hyaluronic acid (HA) and collagens deposition reduction, cytotoxic T cells infiltration, and tumor cells apoptosis. Additionally, in vivo blockade of CXCL10 with AMG487 could partially reverse the pro-tumor effect from MFAP5 overexpression in CAFs and synergize with anti-PD-L1 antibody to enhance the immunotherapeutic effect. Therefore, targeting MFAP5highCAFs might be a potential adjuvant therapy to enhance the immunochemotherapy effect in PDAC via remodeling the desmoplastic and immunosuppressive microenvironment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Microfibrillas/metabolismo , Microfibrillas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Proteínas/metabolismo , Inmunoterapia , Microambiente Tumoral , Proteínas de Unión al Calcio/metabolismo , Neoplasias Pancreáticas
5.
Nat Struct Mol Biol ; 30(5): 608-618, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37081316

RESUMEN

Genetic mutations in fibrillin microfibrils cause serious inherited diseases, such as Marfan syndrome and Weill-Marchesani syndrome (WMS). These diseases typically show major dysregulation of tissue development and growth, particularly in skeletal long bones, but links between the mutations and the diseases are unknown. Here we describe a detailed structural analysis of native fibrillin microfibrils from mammalian tissue by cryogenic electron microscopy. The major bead region showed pseudo eightfold symmetry where the amino and carboxy termini reside. On the basis of this structure, we show that a WMS deletion mutation leads to the induction of a structural rearrangement that blocks interaction with latent TGFß-binding protein-1 at a remote site. Separate deletion of this binding site resulted in the assembly of shorter fibrillin microfibrils with structural alterations. The integrin αvß3-binding site was also mapped onto the microfibril structure. These results establish that in complex extracellular assemblies, such as fibrillin microfibrils, mutations may have long-range structural consequences leading to the disruption of growth factor signaling and the development of disease.


Asunto(s)
Matriz Extracelular , Microfibrillas , Animales , Microfibrillas/metabolismo , Microfibrillas/patología , Fibrilinas/genética , Fibrilinas/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Mutación , Sitios de Unión , Mamíferos/metabolismo
6.
Hum Cell ; 36(2): 822-834, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36527580

RESUMEN

Tongue squamous cell carcinoma (TSCC) represents the most frequent malignancy of the oral cavity, characterized by a high metastasis rate and poor prognosis. Microfibril-associated protein 2 (MFAP2), as an extracellular matrix protein, has been found to drive tumor progression. The function and underlying mechanism of MFAP2 in TSCC remain unknown. The expression levels of MFAP2 were analyzed in tissue samples from 30 TSCC patients by real time-polymerase chain reaction and western blot assays. Our results revealed that the expression of MFAP2 mRNA and protein was upregulated in TSCC tissue samples compared with that in the matched para-carcinoma tissue samples. By performing in vitro gain-of-function or loss-of-function experiments and in vivo mouse xenograft experiments, we found that overexpression of MFAP2 induced proliferation and promoted transition from G1 to S phase of TSCC cells. Stronger invasive and migratory capabilities were observed in MFAP2-overexpressing TSCC cells. In contrast, knockdown of MFAP2 exhibited anti-proliferative, apoptosis-promoting and pro-migratory roles in TSCC cells. Knockdown of MFAP2 significantly inhibited xenograft tumor growth. Mechanistically, POU class 2 homeobox 1 (POU2F1) was recruited to the region of MFAP2 promoter and upregulates the expression of MFAP2. Silencing of MFAP2 effectively blocked the proliferation, migration, and invasion of TSCC cells caused by POU2F1 overexpression. Our results indicate that the role of MFAP2 in TSCC may attribute to transcriptional regulation of POU2F1.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Lengua , Humanos , Animales , Ratones , Carcinoma de Células Escamosas/patología , Neoplasias de la Lengua/patología , Genes Homeobox , Microfibrillas/metabolismo , Microfibrillas/patología , Línea Celular Tumoral , Lengua/metabolismo , Lengua/patología , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
7.
Cancer Med ; 12(7): 8403-8414, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36583532

RESUMEN

BACKGROUND: Distant metastasis is the main cause of mortality in colorectal cancer (CRC) patients. N1-methyladenosine (m1A) is a type of epitranscriptome modification. While its regulatory effect on mRNA and its role in CRC metastasis remain unclear. METHODS: The m1A methylation profile of mRNAs in CRC was revealed by m1A methylated RNA immunoprecipitation sequencing. The expression of MFAP2 in tumor tissues was measured by immunohistochemistry and then correlated with the clinical characteristics and prognosis of CRC patients. The role of MFAP2 in the invasiveness of CRC cells was evaluated by transwell assays and peritoneal metastatic model in nude mice. The downstream targets of MFAP2 was screened by mass spectrometry analysis. Then the role of MFAP2-CLK3 signaling axis was verified by cotransfecting MFAP2 siRNA and CLK3 plasmid in CRC cells. RESULTS: Microfibril associated protein 2 (MFAP2) mRNA was overexpressed and m1A-hypermethylated in CRC. High expression of MFAP2 was closely related to lymph node metastasis and distant metastasis, leading to poor prognosis in patients with CRC. In vivo and in vitro studies showed that silencing of MFAP2 inhibited the migration, invasion and metastasis of CRC cells. CDC Like Kinase 3 (CLK3) was a potential downstream target of MFAP2. Further studies showed that MFAP2 depletion might induce autophagic degradation of CLK3, and the role of MFAP2 in the invasiveness of CRC cells was dependent on CLK3. CONCLUSIONS: Our results uncover a newly identified MFAP2-CLK3 signaling axis, which is a potential therapeutic target for CRC metastasis.


Asunto(s)
Neoplasias Colorrectales , Microfibrillas , Animales , Ratones , Línea Celular Tumoral , Metilación , Ratones Desnudos , Microfibrillas/metabolismo , Microfibrillas/patología , Neoplasias Colorrectales/patología , Proteínas/genética , Proliferación Celular , Invasividad Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
8.
Int J Mol Sci ; 23(24)2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36555638

RESUMEN

Ovarian cancer (OC) is usually diagnosed late due to its nonspecific symptoms and lack of reliable tools for early diagnostics and screening. OC studies concentrate on the search for new biomarkers and therapeutic targets. This study aimed to validate the MFAP5 gene, and its encoded protein, as a potential prognostic biomarker. In our previous study, we found that patients with high-grade serous OC who had higher MFAP5 mRNA levels had shorter survival, as compared with those with lower levels. Here, we used the Kaplan-Meier Plotter and CSIOVDB online tools to analyze possible associations of MFAP5 expression with survival and other clinico-pathological features. In these analyses, higher MFAP5 mRNA expression was observed in the more advanced FIGO stages and high-grade tumors, and was significantly associated with shorter overall and progression-free survival. Next, we analyzed the expression of the MFAP5 protein by immunohistochemistry (IHC) in 108 OC samples and tissue arrays. Stronger MFAP5 expression was associated with stronger desmoplastic reaction and serous vs. non-serous histology. We found no significant correlation between IHC results and survival, although there was a trend toward shorter survival in patients with the highest IHC scores. We searched for co-expressed genes/proteins using cBioPortal and analyzed potential MFAP5 interaction networks with the STRING tool. MFAP5 was shown to interact with many extracellular matrix proteins, and was connected to the Notch signaling pathway. Therefore, although not suitable as a prognostic biomarker for evaluation with a simple diagnostic tool like IHC, MFAP5 is worth further studies as a possible therapeutic target.


Asunto(s)
Proteínas Contráctiles , Neoplasias Ováricas , Humanos , Femenino , Proteínas Contráctiles/genética , Microfibrillas/metabolismo , Pronóstico , Péptidos y Proteínas de Señalización Intercelular , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , ARN Mensajero/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo
9.
Appl Environ Microbiol ; 88(17): e0099522, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35997493

RESUMEN

Mushroom-forming fungi (Agaricomycetes) employ enzymatic and nonenzymatic cellulose degradation mechanisms, the latter presumably relying on Fenton-generated radicals. The effects of the two mechanisms on the cellulose microfibrils structure remain poorly understood. We examined cellulose degradation caused by litter decomposers and wood decomposers, including brown-rot and white-rot fungi and one fungus with uncertain wood decay type, by combining small- and wide-angle X-ray scattering. We also examined the effects of commercial enzymes and Fenton-generated radicals on cellulose using the same method. We detected two main degradation or modification mechanisms. The first characterized the mechanism used by most fungi and resembled enzymatic cellulose degradation, causing simultaneous microfibril thinning and decreased crystalline cellulose. The second mechanism was detected in one brown-rot fungus and one litter decomposer and was characterized by patchy amorphogenesis of crystalline cellulose without substantial thinning of the fibers. This pattern did not resemble the effect of Fenton-generated radicals, suggesting a more complex mechanism is involved in the destruction of cellulose crystallinity by fungi. Furthermore, our results showed a mismatch between decay classifications and cellulose degradation patterns and that even within litter decomposers two degradation mechanisms were found, suggesting higher functional diversity under current ecological classifications of fungi. IMPORTANCE Cellulose degradation by fungi plays a fundamental role in terrestrial carbon cycling, but the mechanisms by which fungi cope with the crystallinity of cellulose are not fully understood. We used X-ray scattering to analyze how fungi, a commercial enzyme mix, and a Fenton reaction-generated radical alter the crystalline structure of cellulose. Our data revealed two mechanisms involved in crystalline cellulose degradation by fungi: one that results in the thinning of the cellulose fibers, resembling the enzymatic degradation of cellulose, and one that involves amorphogenesis of crystalline cellulose by yet-unknown pathways, resulting in a patchy-like degradation pattern. These results pave the way to a deeper understanding of cellulose degradation and the development of novel ways to utilize crystalline cellulose.


Asunto(s)
Agaricales , Basidiomycota , Agaricales/metabolismo , Basidiomycota/metabolismo , Celulosa/metabolismo , Hongos/metabolismo , Lignina/metabolismo , Microfibrillas/metabolismo , Madera/microbiología , Rayos X
10.
Eur Cell Mater ; 43: 277-292, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35730482

RESUMEN

Biochemical and biophysical factors need consideration when modelling in vivo cellular behaviour using in vitro cell culture systems. One underappreciated factor is the high concentration of macromolecules present in vivo, which is typically not simulated under standard cell culture conditions. This disparity is especially relevant when studying biochemical processes that govern extracellular matrix (ECM) deposition, which may be altered due to dilution of secreted macromolecules by the relatively large volumes of culture medium required for cell maintenance in vitro. Macromolecular crowding (MMC) utilises the addition of inert macromolecules to cell culture medium to mimic such high concentration environments found in vivo. The present study induced MMC using the sucrose polymer Ficoll and examined whether fibrillin-1 deposition by human lung fibroblasts could be augmented. Fibrillin-1 forms extracellular microfibrils, which are versatile scaffolds required for elastic fibre formation, deposition of other ECM proteins and growth factor regulation. Pathogenic variants in the fibrillin-1 gene (FBN1) cause Marfan syndrome, where ECM deposition of fibrillin-1 can be compromised. Using immunocytochemistry, significantly enhanced fibrillin-1 deposition was observed when lung fibroblasts were cultured under MMC conditions. MMC also augmented fibrillin-1 deposition in Marfan syndrome patient-derived skin fibroblasts in a cell line- and likely FBN1 variant-specific manner. The ability of MMC to increase fibrillin-1 deposition suggested potential applications for tissue-engineering approaches, e.g. to generate tendon or vascular tissues, where fibrillin-1 microfibrils and elastic fibres are key determinants of their biomechanical properties. Moreover, it suggested the potency of MMC to better mimic in vivo ECM environments in cell culture studies.


Asunto(s)
Síndrome de Marfan , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibrilina-1/análisis , Fibrilina-1/genética , Fibrilina-1/metabolismo , Humanos , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patología , Microfibrillas/genética , Microfibrillas/metabolismo , Microfibrillas/patología
11.
Kaohsiung J Med Sci ; 38(8): 772-780, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35546486

RESUMEN

Ovarian cancer is one of the most common gynecological tumors that seriously endanger the health and quality of life of women. Microfibril-associated protein 2 (MFAP2) has been demonstrated to play crucial roles in the development of multiple tumors. However, the function of MFAP2 in ovarian cancer remains unclear. In this study, we found that MFAP2 was upregulated in ovarian cancer and cells and was positively correlated with FOXM1 and glycolysis-related genes. The results of Cell Count Kit-8, colony formation, and flow cytometry assays indicated that MFAP2 promoted cell proliferation. In addition, MFAP2 promotes cell proliferation, glucose uptake, lactate production; increases ATP levels, extracellular acidification ratio, and oxygen consumption ratio in ovarian cancer cells and increases the expression of glycolytic proteins. Further mechanistic analysis suggests that MFAP2 promotes FOXM1/ß-catenin-mediated glycolysis signaling in ovarian cancer cells. Knockdown of MFAP2 inhibits ovarian cancer xenograft tumor growth and expression of Ki-67, MFAP2, FOXM1, GLUT1, HK2, and ß-catenin in mice. In conclusion, MFAP2 promotes cell proliferation and glycolysis by modulating the FOXM1/ß-catenin signaling pathway in ovarian cancer, which may offer a fresh insight into the treatment of ovarian cancer in the glycolysis pathway.


Asunto(s)
Neoplasias Ováricas , beta Catenina , Animales , Carcinoma Epitelial de Ovario/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Humanos , Ratones , Microfibrillas/metabolismo , Microfibrillas/patología , Neoplasias Ováricas/metabolismo , Calidad de Vida , Factores de Empalme de ARN , beta Catenina/genética , beta Catenina/metabolismo
12.
Elife ; 112022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35503090

RESUMEN

The embryonic extracellular matrix (ECM) undergoes transition to mature ECM as development progresses, yet few mechanisms ensuring ECM proteostasis during this period are known. Fibrillin microfibrils are macromolecular ECM complexes serving structural and regulatory roles. In mice, Fbn1 and Fbn2, encoding the major microfibrillar components, are strongly expressed during embryogenesis, but fibrillin-1 is the major component observed in adult tissue microfibrils. Here, analysis of Adamts6 and Adamts10 mutant mouse embryos, lacking these homologous secreted metalloproteases individually and in combination, along with in vitro analysis of microfibrils, measurement of ADAMTS6-fibrillin affinities and N-terminomics discovery of ADAMTS6-cleaved sites, identifies a proteostatic mechanism contributing to postnatal fibrillin-2 reduction and fibrillin-1 dominance. The lack of ADAMTS6, alone and in combination with ADAMTS10 led to excess fibrillin-2 in perichondrium, with impaired skeletal development defined by a drastic reduction of aggrecan and cartilage link protein, impaired BMP signaling in cartilage, and increased GDF5 sequestration in fibrillin-2-rich tissue. Although ADAMTS6 cleaves fibrillin-1 and fibrillin-2 as well as fibronectin, which provides the initial scaffold for microfibril assembly, primacy of the protease-substrate relationship between ADAMTS6 and fibrillin-2 was unequivocally established by reversal of the defects in Adamts6-/- embryos by genetic reduction of Fbn2, but not Fbn1.


Asunto(s)
Proteínas ADAMTS , Microfibrillas , Proteínas ADAMTS/química , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Animales , Fibrilina-1/genética , Fibrilina-2/metabolismo , Fibrilinas/metabolismo , Ratones , Microfibrillas/metabolismo , Proteolisis
13.
Biophys J ; 121(6): 932-942, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35151632

RESUMEN

Plant cell size and shape are tuned to their function and specified primarily by cellulose microfibril (CMF) patterning of the cell wall. Arabidopsis thaliana leaf trichomes are unicellular structures that act as a physical defense to deter insect feeding. This highly polarized cell type employs a strongly anisotropic cellulose wall to extend and taper, generating sharply pointed branches. During elongation, the mechanisms by which shifts in fiber orientation generate cells with predictable sizes and shapes are unknown. Specifically, the axisymmetric growth of trichome branches is often thought to result from axisymmetric CMF patterning. Here, we analyzed the direction and degree of twist of branches after desiccation to reveal the presence of an asymmetric cell wall organization with a left-hand bias. CMF organization, quantified using computational modeling, suggests a limited reorientation of microfibrils during growth and a maximum branch length limited by the wall axial stiffness. The model provides a mechanism for CMF asymmetry, which occurs after the branch bending stiffness becomes low enough that ambient bending affects the principal stresses. After this stage, the CMF synthesis results in a constant bending stiffness for longer branches. The bending vibration natural frequencies of branches with respect to their length are also discussed.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Pared Celular/metabolismo , Celulosa/análisis , Celulosa/metabolismo , Desecación , Microfibrillas/química , Microfibrillas/metabolismo
14.
Int J Mol Sci ; 23(3)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35163482

RESUMEN

Elastin represents the structural component of the extracellular matrix providing elastic recoil to tissues such as skin, blood vessels and lungs. Elastogenic cells secrete soluble tropoelastin monomers into the extracellular space where these monomers associate with other matrix proteins (e.g., microfibrils and glycoproteins) and are crosslinked by lysyl oxidase to form insoluble fibres. Once elastic fibres are formed, they are very stable, highly resistant to degradation and have an almost negligible turnover. However, there are circumstances, mainly related to inflammatory conditions, where increased proteolytic degradation of elastic fibres may lead to consequences of major clinical relevance. In severely affected COVID-19 patients, for instance, the massive recruitment and activation of neutrophils is responsible for the profuse release of elastases and other proteolytic enzymes which cause the irreversible degradation of elastic fibres. Within the lungs, destruction of the elastic network may lead to the permanent impairment of pulmonary function, thus suggesting that elastases can be a promising target to preserve the elastic component in COVID-19 patients. Moreover, intrinsic and extrinsic factors additionally contributing to damaging the elastic component and to increasing the spread and severity of SARS-CoV-2 infection are reviewed.


Asunto(s)
COVID-19/metabolismo , Elastina/fisiología , Matriz Extracelular/fisiología , Animales , Tejido Elástico/metabolismo , Elastina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Trampas Extracelulares/metabolismo , Fibrilinas/metabolismo , Humanos , Pulmón/patología , Microfibrillas/metabolismo , Proteínas de Microfilamentos/metabolismo , Neutrófilos , Proteína-Lisina 6-Oxidasa/metabolismo , SARS-CoV-2/patogenicidad , Tropoelastina/metabolismo
15.
J Cosmet Dermatol ; 21(10): 4796-4804, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35064622

RESUMEN

INTRODUCTION: The elastic fiber structure becomes shorter, thicker, and curved with age. Nonetheless, the proteins and catabolic enzymes influencing the maintenance of and change in the three-dimensional (3D) structure of elastic fibers remain unknown. This study aimed to identify the proteins involved in the maintenance and degeneration of elastic fiber structures. METHODS: We performed a combined 3D structural analysis using tissue decolorization technology and mRNA abundance and comprehensive protein expression of tissue-derived cells. The relationship between the proteins was evaluated. RESULTS: Elastin microfibril interface-located protein 1 (EMILIN-1) and cathepsin K (CTSK) were implicated in structural changes in elastic fibers with aging. EMILIN-1 and CTSK levels were highly correlated and changed with age. CTSK was identified as the degrading enzyme of EMILIN-1. CTSK fragmented the otherwise linearly existing dermal elastic fiber structure, with more evident changes in oxytalan fibers. EMILIN-1 expression in fibroblasts was increased by co-culturing with keratinocytes. Furthermore, CTSK expression was increased by UV stress in keratinocytes, resulting in decreased EMILIN-1 expression. CONCLUSION: Using our new assessment strategy, we observed that EMILIN-1 and CTSK are highly linked to changes in the elastic fiber structure with aging. These results indicate that suppressing CTSK expression and increasing EMILIN-1 expression might be an effective approach to prevent elastic fiber morphological changes that lead to wrinkles and sagging. Furthermore, EMILIN-1 in the dermis increases due to interaction with the epidermis, which could provide a new target for the therapeutic care of elastic fibers (including preservation of oxytalan fibers) in epidermis-dermis interaction.


Asunto(s)
Tejido Elástico , Elastina , Humanos , Elastina/metabolismo , Microfibrillas/química , Microfibrillas/metabolismo , Catepsina K/metabolismo
16.
Hematol Oncol ; 40(2): 181-190, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34783040

RESUMEN

CD49d, the α4 chain of the VLA-4 integrin, is a negative prognosticator in chronic lymphocytic leukemia (CLL) with a key role in CLL cell-microenvironment interactions mainly occurring via its ligands VCAM-1 and fibronectin. In the present study, we focused on EMILIN-1 (Elastin-MIcrofibriL-INterfacer-1), an alternative VLA-4 ligand whose role has been so far reported only in non-hematological settings, by investigating: i) the distribution of EMILIN-1 in CLL-involved tissues; ii) the capability of EMILIN-1 to operate, via its globular C1q (gC1q) domain, as additional adhesion ligand in CLL; iii) the functional meaning of EMILIN-1 gC1q/VLA-4 interactions in CLL. EMILIN-1 is widely present in the CLL-involved areas of bone marrow biopsies (BMBs) without difference between CD49d negative and positive cases, displaying at least three different expression patterns: "fibrillar", "dot-like" and "mixed". The lack in CLL-BMB of neutrophil elastase, whose proteolytic activity degrades EMILIN-1 and impairs EMILIN-1 function, suggests full functional EMILIN-1 in CLL independently of its expression pattern. Functionally, EMILIN-1 gC1q domain promotes adhesion of CLL cells through specific interaction with VLA-4, and releases pro-survival signals for CLL cells, as demonstrated by enhanced ERK and AKT phosphorylation and impairment of in-vitro-induced apoptosis. EMILIN-1/VLA-4 interaction can efficiently contribute to the maintenance of the neoplastic clone in CLL.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Elastina , Humanos , Integrina alfa4beta1/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ligandos , Glicoproteínas de Membrana , Microfibrillas/metabolismo , Microfibrillas/patología , Microambiente Tumoral
17.
Invest Ophthalmol Vis Sci ; 62(10): 26, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34424262

RESUMEN

Purpose: Previously, we identified a G661R mutation of ADAMTS10 (a disintegrin-like and metalloprotease with thrombospondin type 1 motif 10) as being disease causative in a colony of Beagles with inherited primary open-angle glaucoma (POAG). Mutations in ADAMTS10 are known to cause Weill-Marchesani syndrome (WMS), which is also caused by mutations in the fibrillin-1 gene (FBN1), suggesting functional linkage between ADAMTS10 and fibrillin-1, the principal component of microfibrils. Here, we established a mouse line with the G661R mutation of Adamts10 (Adamts10G661R/G661R) to determine if they develop features of WMS and alterations of ocular fibrillin microfibrils. Methods: Intraocular pressure (IOP) was measured using a TonoLab rebound tonometer. Central cornea thickness (CCT), anterior chamber depth (ACD) and axial length (AL) of the eye were examined by spectral-domain optical coherence tomography. Sagittal eye sections from mice at postnatal day 10 (P10) and at 3 and 24 months of age were stained with antibodies against fibrillin-1, fibrillin-2, and ADAMTS10. Results: IOP was not elevated in Adamts10G661R/G661R mice. Adamts10G661R/G661R mice had smaller bodies, thicker CCT, and shallower ACD compared to wild-type mice but normal AL. Adamts10G661R/G661R mice displayed persistent fibrillin-2 and enhanced fibrillin-1 immunofluorescence in the lens zonules and in the hyaloid vasculature and its remnants in the vitreous. Conclusions: Adamts10G661R/G661R mice recapitulate the short stature and ocular phenotypes of WMS. The altered fibrillin-1 and fibrillin-2 immunoactivity in Adamts10G661R/G661R mice suggests that the G661R mutation of Adamts10 perturbs regulation of the fibrillin isotype composition of microfibrils in the mouse eye.


Asunto(s)
Proteínas ADAMTS/genética , Cámara Anterior/metabolismo , ADN/genética , Fibrilinas/metabolismo , Glaucoma de Ángulo Abierto/genética , Microfibrillas/metabolismo , Mutación , Proteínas ADAMTS/metabolismo , Animales , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Glaucoma de Ángulo Abierto/metabolismo , Glaucoma de Ángulo Abierto/fisiopatología , Masculino , Ratones , Transducción de Señal
18.
PLoS One ; 16(3): e0248532, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33735269

RESUMEN

The human FBN1 gene encodes fibrillin-1 (FBN1); the main component of the 10-12 nm diameter extracellular matrix microfibrils. Marfan syndrome (MFS) is a common inherited connective tissue disorder, caused by FBN1 mutations. It features a wide spectrum of disease severity, from mild cases to the lethal neonatal form (nMFS), that is yet to be explained at the molecular level. Mutations associated with nMFS generally affect a region of FBN1 between domains TB3-cbEGF18-the "neonatal region". To gain insight into the process of fibril assembly and increase our understanding of the mechanisms determining disease severity in MFS, we compared the secretion and assembly properties of FBN1 variants containing nMFS-associated substitutions with variants associated with milder, classical MFS (cMFS). In the majority of cases, both nMFS- and cMFS-associated neonatal region variants were secreted at levels comparable to wild type. Microfibril incorporation by the nMFS variants was greatly reduced or absent compared to the cMFS forms, however, suggesting that nMFS substitutions disrupt a previously undefined site of microfibril assembly. Additional analysis of a domain deletion variant caused by exon skipping also indicates that register in the neonatal region is likely to be critical for assembly. These data demonstrate for the first time new requirements for microfibril biogenesis and identify at least two distinct molecular mechanisms associated with disease substitutions in the TB3-cbEGF18 region; incorporation of mutant FBN1 into microfibrils changing their integral properties (cMFS) or the blocking of wild type FBN1 assembly by mutant molecules that prevents late-stage lateral assembly (nMFS).


Asunto(s)
Fibrilina-1/genética , Síndrome de Marfan/genética , Microfibrillas/metabolismo , Fibrilina-1/metabolismo , Células HEK293 , Humanos , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/patología , Microfibrillas/patología , Mutagénesis , Mutación , Multimerización de Proteína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Índice de Severidad de la Enfermedad
19.
Dev Cell ; 56(1): 67-80.e3, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33434527

RESUMEN

Tissue folding is a central building block of plant and animal morphogenesis. In dicotyledonous plants, hypocotyl folds to form hooks after seedling germination that protects their aerial stem cell niche during emergence from soil. Auxin response factors and auxin transport are reported to play a key role in this process. Here, we show that the microtubule-severing enzyme katanin contributes to hook formation. However, by exposing hypocotyls to external mechanical cues mimicking the natural soil environment, we reveal that auxin response factors ARF7/ARF19, auxin influx carriers, and katanin are dispensable for apical hook formation, indicating that these factors primarily play the role of catalyzers of tissue bending in the absence of external mechanical cues. Instead, our results reveal the key roles of the non-canonical TMK-mediated auxin pathway, PIN efflux carriers, and cellulose microfibrils as components of the core pathway behind hook formation in the presence or absence of external mechanical cues.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Hipocótilo/metabolismo , Ácidos Indolacéticos/metabolismo , Katanina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Morfogénesis/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/genética , Señales (Psicología) , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/genética , Katanina/genética , Proteínas de Transporte de Membrana/genética , Microfibrillas/metabolismo , Microscopía Confocal , Microtúbulos/enzimología , Microtúbulos/metabolismo , Morfogénesis/fisiología , Plantas Modificadas Genéticamente/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Plantones/genética , Plantones/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Microtomografía por Rayos X
20.
FASEB J ; 35(2): e21310, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33484187

RESUMEN

ADAMTS proteases mediate biosynthesis and breakdown of secreted extracellular matrix (ECM) molecules in numerous physiological and disease processes. In addition to their catalytic domains, ADAMTS proteases contain ancillary domains, which mediate substrate recognition and ECM binding and confer distinctive properties and roles to individual ADAMTS proteases. Although alternative splicing can greatly expand the structural and functional diversity of ADAMTS proteases, it has been infrequently reported and functional consequences have been rarely investigated. Here, we characterize the structural and functional impact of alternative splicing of ADAMTS17, mutations in which cause Weill-Marchesani syndrome 4. Two novel ADAMTS17 splice variants, ADAMTS17A and ADAMTS17B, were investigated by structural modeling, mass spectrometry, and biochemical approaches. Our results identify a novel disulfide-bridged insertion in the ADAMTS17A spacer that originates from inclusion of a novel exon. This insertion results in differential autoproteolysis of ADAMTS17, and thus, predicts altered proteolytic activity against other substrates. The second variant, ADAMTS17B, results from an in-frame exon deletion and prevents ADAMTS17B secretion. Thus, alternative splicing of the ADAMTS spacer significantly regulates the physiologically relevant proteolytic activity of ADAMTS17, either by altering proteolytic specificity (ADAMTS17A) or by altering cellular localization (ADAMTS17B).


Asunto(s)
Proteínas ADAMTS/metabolismo , Empalme Alternativo/fisiología , Proteínas ADAMTS/genética , Empalme Alternativo/genética , Western Blotting , Técnicas de Cocultivo , Matriz Extracelular/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Células HEK293 , Humanos , Espectrometría de Masas , Microfibrillas/metabolismo , Mutación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...