Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Nature ; 608(7924): 784-794, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35798028

RESUMEN

Cancer relapse after chemotherapy remains a main cause of cancer-related death. Although the relapse is thought to result from the propagation of resident cancer stem cells1, a lack of experimental platforms that enable the prospective analysis of cancer stem cell dynamics with sufficient spatiotemporal resolution has hindered the testing of this hypothesis. Here we develop a live genetic lineage-tracing system that allows the longitudinal tracking of individual cells in xenotransplanted human colorectal cancer organoids, and identify LGR5+ cancer stem cells that exhibit a dormant behaviour in a chemo-naive state. Dormant LGR5+ cells are marked by the expression of p27, and intravital imaging provides direct evidence of the persistence of LGR5+p27+ cells during chemotherapy, followed by clonal expansion. Transcriptome analysis reveals that COL17A1-a cell-adhesion molecule that strengthens hemidesmosomes-is upregulated in dormant LGR5+p27+ cells. Organoids in which COL17A1 is knocked out lose the dormant LGR5+p27+ subpopulation and become sensitive to chemotherapy, which suggests that the cell-matrix interface has a role in the maintenance of dormancy. Chemotherapy disrupts COL17A1 and breaks the dormancy in LGR5+p27+ cells through FAK-YAP activation. Abrogation of YAP signalling prevents chemoresistant cells from exiting dormancy and delays the regrowth of tumours, highlighting the therapeutic potential of YAP inhibition in preventing cancer relapse. These results offer a viable therapeutic approach to overcome the refractoriness of human colorectal cancer to conventional chemotherapy.


Asunto(s)
Neoplasias del Colon , Células Madre Neoplásicas , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linaje de la Célula , Proliferación Celular , Rastreo Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Quinasa 1 de Adhesión Focal/metabolismo , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Colágenos no Fibrilares/metabolismo , Organoides/metabolismo , Organoides/patología , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/metabolismo , Colágeno Tipo XVII
2.
Nature ; 568(7752): 344-350, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944469

RESUMEN

Stem cells underlie tissue homeostasis, but their dynamics during ageing-and the relevance of these dynamics to organ ageing-remain unknown. Here we report that the expression of the hemidesmosome component collagen XVII (COL17A1) by epidermal stem cells fluctuates physiologically through genomic/oxidative stress-induced proteolysis, and that the resulting differential expression of COL17A1 in individual stem cells generates a driving force for cell competition. In vivo clonal analysis in mice and in vitro 3D modelling show that clones that express high levels of COL17A1, which divide symmetrically, outcompete and eliminate adjacent stressed clones that express low levels of COL17A1, which divide asymmetrically. Stem cells with higher potential or quality are thus selected for homeostasis, but their eventual loss of COL17A1 limits their competition, thereby causing ageing. The resultant hemidesmosome fragility and stem cell delamination deplete adjacent melanocytes and fibroblasts to promote skin ageing. Conversely, the forced maintenance of COL17A1 rescues skin organ ageing, thereby indicating potential angles for anti-ageing therapeutic intervention.


Asunto(s)
Homeostasis , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/fisiología , Piel/citología , Piel/patología , Células Madre/citología , Células Madre/patología , Animales , Atrofia , Autoantígenos/química , Autoantígenos/metabolismo , División Celular , Proliferación Celular , Células Clonales/citología , Células Epidérmicas/citología , Células Epidérmicas/patología , Femenino , Genoma , Hemidesmosomas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Colágenos no Fibrilares/química , Colágenos no Fibrilares/metabolismo , Estrés Oxidativo , Proteolisis , Colágeno Tipo XVII
3.
J Med Genet ; 61(4): 347-355, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-37979963

RESUMEN

BACKGROUND: Collagen XVII is most typically associated with human disease when biallelic COL17A1 variants (>230) cause junctional epidermolysis bullosa (JEB), a rare, genetically heterogeneous, mucocutaneous blistering disease with amelogenesis imperfecta (AI), a developmental enamel defect. Despite recognition that heterozygous carriers in JEB families can have AI, and that heterozygous COL17A1 variants also cause dominant corneal epithelial recurrent erosion dystrophy (ERED), the importance of heterozygous COL17A1 variants causing dominant non-syndromic AI is not widely recognised. METHODS: Probands from an AI cohort were screened by single molecule molecular inversion probes or targeted hybridisation capture (both a custom panel and whole exome sequencing) for COL17A1 variants. Patient phenotypes were assessed by clinical examination and analyses of affected teeth. RESULTS: Nineteen unrelated probands with isolated AI (no co-segregating features) had 17 heterozygous, potentially pathogenic COL17A1 variants, including missense, premature termination codons, frameshift and splice site variants in both the endo-domains and the ecto-domains of the protein. The AI phenotype was consistent with enamel of near normal thickness and variable focal hypoplasia with surface irregularities including pitting. CONCLUSION: These results indicate that COL17A1 variants are a frequent cause of dominantly inherited non-syndromic AI. Comparison of variants implicated in AI and JEB identifies similarities in type and distribution, with five identified in both conditions, one of which may also cause ERED. Increased availability of genetic testing means that more individuals will receive reports of heterozygous COL17A1 variants. We propose that patients with isolated AI or ERED, due to COL17A1 variants, should be considered as potential carriers for JEB and counselled accordingly, reflecting the importance of multidisciplinary care.


Asunto(s)
Amelogénesis Imperfecta , Colágenos no Fibrilares , Humanos , Colágenos no Fibrilares/genética , Colágenos no Fibrilares/metabolismo , Autoantígenos/genética , Amelogénesis Imperfecta/genética , Heterocigoto , Fenotipo , Mutación/genética
4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33859045

RESUMEN

The extracellular matrix (ECM) provides a precise physical and molecular environment for cell maintenance, self-renewal, and differentiation in the stem cell niche. However, the nature and organization of the ECM niche is not well understood. The adult freshwater planarian Schmidtea mediterranea maintains a large population of multipotent stem cells (neoblasts), presenting an ideal model to study the role of the ECM niche in stem cell regulation. Here we tested the function of 165 planarian homologs of ECM and ECM-related genes in neoblast regulation. We identified the collagen gene family as one with differential effects in promoting or suppressing proliferation of neoblasts. col4-1, encoding a type IV collagen α-chain, had the strongest effect. RNA interference (RNAi) of col4-1 impaired tissue maintenance and regeneration, causing tissue regression. Finally, we provide evidence for an interaction between type IV collagen, the discoidin domain receptor, and neuregulin-7 (NRG-7), which constitutes a mechanism to regulate the balance of symmetric and asymmetric division of neoblasts via the NRG-7/EGFR pathway.


Asunto(s)
Colágeno Tipo IV/genética , Planarias/genética , Planarias/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Colágeno Tipo IV/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Homeostasis , Colágenos no Fibrilares/metabolismo , Regeneración , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
5.
BMC Cancer ; 23(1): 949, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803411

RESUMEN

BACKGROUND: Collagens are the major components of the extracellular matrix (ECM) and are known to contribute to tumor progression and metastasis. There are 28 different types of collagens each with unique functions in maintaining tissue structure and function. Type XVII collagen (BP180) is a type II transmembrane protein that provides stable adhesion between epithelial cells and the underlying basement membrane. Aberrant expression and ectodomain shedding of type XVII collagen have been associated with epithelial damage, tumor invasiveness, and metastasis in multiple tumor types and may consequently be used as a potential (non-invasive) biomarker in cancer and treatment target. METHOD: An ELISA targeting the type XVII collagen ectodomain (PRO-C17) was developed for use in serum. PRO-C17 was measured in a cohort of patients with 11 different cancer types (n = 214) and compared to healthy controls (n = 23) (cohort 1). Based on the findings from cohort 1, PRO-C17 and its association with survival was explored in patients with metastatic colorectal cancer (mCRC) treated with bevacizumab in combination with chemotherapy (n = 212) (cohort 2). RESULTS: PRO-C17 was robust and specific towards the ectodomain of type XVII collagen. In cohort 1, PRO-C17 levels were elevated (p < 0.05) in serum from patients with CRC, kidney, ovarian, bladder, breast, and head and neck cancer compared to healthy controls. PRO-C17 was especially good at discriminating between CRC patients and healthy controls with an AUROC of 0.904. In cohort 2, patients with mCRC and high levels (tertile 3) of PRO-C17 had shorter overall survival (OS) with a median OS of 390 days compared to 539 days for patients with low levels of PRO-C17. When evaluated by multivariate Cox regression analysis, high PRO-C17 was predictive for poor OS independent of risk factors and the tumor fibrosis biomarker PRO-C3. CONCLUSION: PRO-C17 measures the ectodomain of type XVII collagen in serum and is a promising non-invasive biomarker that can aid in understanding tumor heterogeneity as well as elaborate on the role of collagen XVII in tumor progression. Moreover, the findings in the study proposes PRO-C17 as novel biomarker of epithelial damage in specific cancer types including CRC.


Asunto(s)
Neoplasias del Colon , Neoplasias del Recto , Humanos , Pronóstico , Colágenos no Fibrilares/metabolismo , Colágeno/química , Autoantígenos/metabolismo , Biomarcadores , Colágeno Tipo XVII
6.
J Immunol ; 205(10): 2786-2794, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32998984

RESUMEN

BP180 (also termed type XVII collagen) is a hemidesmosomal protein and plays a critical role in cell-cell matrix adhesion in the skin; however, its other biological functions are largely unclear. In this study, we generated a BP180 functional-deficient mouse strain by deleting its extracellular domain of humanized NC16A (termed ΔNC16A mice). We found that BP180 is expressed by bone marrow mesenchymal stem cells (BM-MSC), and its functional deficiency leads to myeloid hyperplasia. Altered granulopoiesis in ΔNC16A mice is through bone marrow stromal cells evidenced by bone marrow transplantation. Furthermore, the level of G-CSF in bone marrow and circulation were significantly increased in ΔNC16A mice as compared with wild-type mice. The increased G-CSF was accompanied by an increased activation of the NF-κB signaling pathway in bone marrow and BM-MSC of ΔNC16A mice. Blockade of G-CSF restored normal granulopoiesis in ΔNC16A mice. Inhibition of NF-κB signaling pathway significantly reduces the release of G-CSF from ΔNC16A BM-MSC in vitro and the level of serum G-CSF in ΔNC16A mice. To our knowledge, these findings provide the first direct evidence that BP180 plays an important role in granulopoiesis through regulating NF-κB signaling pathway in BM-MSC.


Asunto(s)
Autoantígenos/metabolismo , Médula Ósea/patología , Leucopoyesis/inmunología , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/fisiología , Colágenos no Fibrilares/metabolismo , Animales , Autoantígenos/genética , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Hiperplasia/genética , Hiperplasia/inmunología , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Colágenos no Fibrilares/genética , Dominios Proteicos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Colágeno Tipo XVII
7.
Int J Mol Sci ; 23(8)2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35456962

RESUMEN

In the tumor microenvironment, the extracellular matrix (ECM) has been recognized as an important part of cancer development. The dominant ECM proteins are the 28 types of collagens, each with a unique function in tissue architecture. Type XX collagen, however, is poorly characterized, and little is known about its involvement in cancer. We developed an ELISA quantifying type XX collagen, named PRO-C20, using a monoclonal antibody raised against the C-terminus. PRO-C20 and PRO-C1, an ELISA targeting the N-terminal pro-peptide of type I collagen, was measured in sera of 219 patients with various solid cancer types and compared to sera levels of 33 healthy controls. PRO-C20 was subsequently measured in a separate cohort comprising 36 patients with pancreatic ductal adenocarcinoma (PDAC) and compared to 20 healthy controls and 11 patients with chronic pancreatitis. PRO-C20 was significantly elevated in all cancers tested: bladder, breast, colorectal, head and neck, kidney, lung, melanoma, ovarian, pancreatic, prostate, and stomach cancer (p < 0.01−p < 0.0001). PRO-C1 was only elevated in patients with ovarian cancer. PRO-C20 could discriminate between patients and healthy controls with AUROC values ranging from 0.76 to 0.92. Elevated levels were confirmed in a separate cohort of patients with PDAC (p < 0.0001). High PRO-C20 levels (above 2.57 nM) were predictive of poor survival after adjusting for the presence of metastasis, age, and sex (HR: 4.25, 95% CI: 1.52−11.9, p-value: 0.006). Circulating type XX collagen is elevated in sera of patients with various types of cancer and has prognostic value in PDAC. If validated, PRO-C20 may be a novel biomarker for patients with solid tumors and can help understand the ECM biology of cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/patología , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Masculino , Colágenos no Fibrilares/metabolismo , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
8.
Semin Cancer Biol ; 62: 134-148, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31479735

RESUMEN

The extracellular matrix (ECM) is ubiquitously involved in neoplastic transformation, tumour growth and metastatic dissemination, and the interplay between tumour and stromal cells and the ECM is now considered crucial for the formation of a tumour-supporting microenvironment. The 28 different collagens (Col) form a major ECM protein family and display extraordinary functional diversity in tissue homeostasis as well as in pathological conditions, with functions ranging from structural support for tissues to regulatory binding activities and storage of biologically active cryptic domains releasable through ECM proteolysis. Two subfamilies of collagens, namely the plasma membrane-associated collagens with interrupted triple-helices (MACITs, including ColXIII, ColXXIII and ColXXV) and the basement membrane-associated collagens with multiple triple-helix domains with interruptions (multiplexins, including ColXV and ColXVIII), have highly interesting regulatory functions in tissue and organ development, as well as in various diseases, including cancer. An increasing, albeit yet sparse, data suggest that these collagens play crucial roles in conveying regulatory signals from the extracellular space to cells. We summarize here the current knowledge about MACITs and multiplexins as regulators of stemness and oncogenic processes, as well as their roles in influencing cell fate decisions in healthy and cancerous tissues. In addition, we present a bioinformatic analysis of the impacts of MACITs and multiplexins transcript levels on the prognosis of patients representing a wide array of malignant diseases, to aid future diagnostic and therapeutic efforts.


Asunto(s)
Membrana Celular/metabolismo , Neoplasias/metabolismo , Colágenos no Fibrilares/metabolismo , Células Madre/metabolismo , Animales , Susceptibilidad a Enfermedades , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/patología , Proteolisis , Nicho de Células Madre/genética , Relación Estructura-Actividad , Microambiente Tumoral
9.
Exp Dermatol ; 30(1): 62-67, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32970880

RESUMEN

Type XVII collagen (COL17) is a transmembrane protein expressed in the basal epidermis. COL17 serves as a niche for epidermal stem cells, and although its reduction has been implicated in altering cell polarity and ageing of the epidermis, it is unknown how COL17 affects epidermal cell polarity. Here, we uncovered COL17 as a binding partner of the aPKC-PAR complex, which is a key regulating factor of cell polarity. Immunoprecipitation-immunoblot assay and protein-protein binding assay revealed that COL17 interacts with aPKC and PAR3. COL17 deficiency or epidermis-specific aPKCλ deletion destabilized PAR3 distribution in the epidermis, while aPKCζ knockout did not. Asymmetrical cell division was pronounced in COL17-null neonatal paw epidermis. These results show that COL17 is pivotal for maintaining epidermal cell polarity. Our study highlights the previously unrecognized role of COL17 in the basal keratinocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Polaridad Celular , Epidermis/metabolismo , Colágenos no Fibrilares/metabolismo , Proteína Quinasa C/metabolismo , Animales , Autoantígenos/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Colágenos no Fibrilares/genética , Isoformas de Proteínas/metabolismo , Colágeno Tipo XVII
10.
Cell Mol Biol Lett ; 26(1): 42, 2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-34602061

RESUMEN

BACKGROUND: Keratinocytes and fibroblasts represent the major cell types in the epidermis and dermis of the skin and play a significant role in maintenance of skin homeostasis. However, the biological characteristics of keratinocytes and fibroblasts remain to be elucidated. The purpose of this study was to compare the gene expression pattern between keratinocytes and fibroblasts and to explore novel biomarker genes so as to provide potential therapeutic targets for skin-related diseases such as burns, wounds, and aging. METHODS: Skin keratinocytes and fibroblasts were isolated from newborn mice. To fully understand the heterogeneity of gene expression between keratinocytes and fibroblasts, differentially expressed genes (DEGs) between the two cell types were detected by RNA-seq technology. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the known genes of keratinocytes and fibroblasts and verify the RNA-seq results. RESULTS: Transcriptomic data showed a total of 4309 DEGs (fold-change > 1.5 and q-value < 0.05). Among them, 2197 genes were highly expressed in fibroblasts and included 10 genes encoding collagen, 16 genes encoding transcription factors, and 14 genes encoding growth factors. Simultaneously, 2112 genes were highly expressed in keratinocytes and included 7 genes encoding collagen, 14 genes encoding transcription factors, and 8 genes encoding growth factors. Furthermore, we summarized 279 genes specifically expressed in keratinocytes and 33 genes specifically expressed in fibroblasts, which may represent distinct molecular signatures of each cell type. Additionally, we observed some novel specific biomarkers for fibroblasts such as Plac8 (placenta-specific 8), Agtr2 (angiotensin II receptor, type 2), Serping1 (serpin peptidase inhibitor, clade G, member 1), Ly6c1 (lymphocyte antigen 6 complex, locus C1), Dpt (dermatopontin), and some novel specific biomarkers for keratinocytes such as Ly6a (lymphocyte antigen 6 complex, locus A) and Lce3c (late cornified envelope 3C), Ccer2 (coiled-coil glutamate-rich protein 2), Col18a1 (collagen, type XVIII, alpha 1) and Col17a1 (collagen type XVII, alpha 1). In summary, these data provided novel identifying biomarkers for two cell types, which can provide a resource of DEGs for further investigations.


Asunto(s)
Biomarcadores/metabolismo , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Enfermedades de la Piel/metabolismo , Piel/metabolismo , Animales , Autoantígenos/metabolismo , Células Cultivadas , Masculino , Ratones , Colágenos no Fibrilares/metabolismo , Análisis de Secuencia de ARN/métodos , Colágeno Tipo XVII
11.
Proc Natl Acad Sci U S A ; 115(25): 6434-6439, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29866844

RESUMEN

BP180, also known as collagen XVII, is a hemidesmosomal component and plays a key role in maintaining skin dermal/epidermal adhesion. Dysfunction of BP180, either through genetic mutations in junctional epidermolysis bullosa (JEB) or autoantibody insult in bullous pemphigoid (BP), leads to subepidermal blistering accompanied by skin inflammation. However, whether BP180 is involved in skin inflammation remains unknown. To address this question, we generated a BP180-dysfunctional mouse strain and found that mice lacking functional BP180 (termed ΔNC16A) developed spontaneous skin inflammatory disease, characterized by severe itch, defective skin barrier, infiltrating immune cells, elevated serum IgE levels, and increased expression of thymic stromal lymphopoietin (TSLP). Severe itch is independent of adaptive immunity and histamine, but dependent on increased expression of TSLP by keratinocytes. In addition, a high TSLP expression is detected in BP patients. Our data provide direct evidence showing that BP180 regulates skin inflammation independently of adaptive immunity, and BP180 dysfunction leads to a TSLP-mediated itch. The newly developed mouse strain could be a model for elucidation of disease mechanisms and development of novel therapeutic strategies for skin inflammation and BP180-related skin conditions.


Asunto(s)
Autoantígenos/metabolismo , Inflamación/metabolismo , Colágenos no Fibrilares/metabolismo , Piel/metabolismo , Inmunidad Adaptativa/inmunología , Animales , Autoantígenos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Histamina/inmunología , Histamina/metabolismo , Humanos , Inmunoglobulina E/sangre , Inflamación/sangre , Inflamación/inmunología , Queratinocitos/inmunología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Colágenos no Fibrilares/inmunología , Penfigoide Ampolloso/inmunología , Penfigoide Ampolloso/metabolismo , Prurito/sangre , Prurito/inmunología , Prurito/metabolismo , Piel/inmunología , Linfopoyetina del Estroma Tímico , Colágeno Tipo XVII
12.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830116

RESUMEN

BP180 is a type II collagenous transmembrane protein and is best known as the major autoantigen in the blistering skin disease bullous pemphigoid (BP). The BP180 trimer is a central component in type I hemidesmosomes (HD), which cause the adhesion between epidermal keratinocytes and the basal lamina, but BP180 is also expressed in several non-HD locations, where its functions are poorly characterized. The immunological roles of intact and proteolytically processed BP180, relevant in BP, have been subject to intensive research, but novel functions in cell proliferation, differentiation, and aging have also recently been described. To better understand the multiple physiological functions of BP180, the focus should return to the protein itself. Here, we comprehensively review the properties of the BP180 molecule, present new data on the biochemical features of its intracellular domain, and discuss their significance with regard to BP180 folding and protein-protein interactions.


Asunto(s)
Autoantígenos , Hemidesmosomas , Queratinocitos , Colágenos no Fibrilares , Penfigoide Ampolloso , Pliegue de Proteína , Autoantígenos/inmunología , Autoantígenos/metabolismo , Hemidesmosomas/inmunología , Hemidesmosomas/metabolismo , Humanos , Queratinocitos/inmunología , Queratinocitos/metabolismo , Colágenos no Fibrilares/inmunología , Colágenos no Fibrilares/metabolismo , Penfigoide Ampolloso/inmunología , Penfigoide Ampolloso/metabolismo , Colágeno Tipo XVII
13.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33805154

RESUMEN

Intermediate junctional epidermolysis bullosa caused by mutations in the COL17A1 gene is characterized by the frequent development of blisters and erosions on the skin and mucous membranes. The rarity of the disease and the heterogeneity of the underlying mutations renders therapy developments challenging. However, the high number of short in-frame exons facilitates the use of antisense oligonucleotides (AON) to restore collagen 17 (C17) expression by inducing exon skipping. In a personalized approach, we designed and tested three AONs in combination with a cationic liposomal carrier for their ability to induce skipping of COL17A1 exon 7 in 2D culture and in 3D skin equivalents. We show that AON-induced exon skipping excludes the targeted exon from pre-mRNA processing, which restores the reading frame, leading to the expression of a slightly truncated protein. Furthermore, the expression and correct deposition of C17 at the dermal-epidermal junction indicates its functionality. Thus, we assume AON-mediated exon skipping to be a promising tool for the treatment of junctional epidermolysis bullosa, particularly applicable in a personalized manner for rare genotypes.


Asunto(s)
Autoantígenos/metabolismo , Epidermólisis Ampollosa de la Unión/genética , Colágenos no Fibrilares/metabolismo , Oligonucleótidos Antisentido/genética , Empalme del ARN , Empalme Alternativo , Biopsia , Línea Celular , Supervivencia Celular , Epidermólisis Ampollosa de la Unión/metabolismo , Epidermólisis Ampollosa de la Unión/terapia , Exones , Genotipo , Homocigoto , Humanos , Queratinocitos/citología , Liposomas/química , Mutación , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Colágeno Tipo XVII
14.
Prostate ; 80(9): 715-726, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32364250

RESUMEN

BACKGROUND: Tumor microenvironment or stroma has the potency to regulate the behavior of malignant cells. Fibroblast-like cells are abundant in tumor stroma and they are also responsible for the synthesis of many extracellular matrix components. Fibroblast-cancer cell interplay can modify the functions of both cell types. METHODS: We applied mass spectrometry and proteomics to unveil the matrisome in 3D spheroids formed by DU145 prostate cancer cells, PC3 prostate cancer cells, or prostate-derived fibroblasts. Similarly, DU145/fibroblast and PC3/fibroblast coculture spheroids were also analyzed. Western blot analysis and immunofluorescence were used to confirm the presence of specific proteins in spheroids. Cancer dissemination was studied by utilizing "out of spheroids" migration and invasion assays. RESULTS: In the spheroid model cancer cell-fibroblast interplay caused remarkable changes in the extracellular matrix and accelerated the invasion of DU145 cells. Fibroblasts produced structural matrix proteins, growth factors, and matrix metalloproteinases. In cancer cell/fibroblast cocultures basement membrane components, including laminins (α3, α5, ß2, and ß3), heparan sulfate proteoglycan (HSPG2 gene product), and collagen XVIII accumulated in a prominent manner when compared with spheroids that contained fibroblasts or cancer cells only. Furthermore, collagen XVIII was intensively processed to different endostatin-containing isoforms by cancer cell-derived cathepsin L. CONCLUSIONS: Fibroblasts can promote carcinoma cell dissemination by several different mechanisms. Extracellular matrix and basement membrane proteins provide attachment sites for cell locomotion promoting adhesion receptors. Growth factors and metalloproteinases are known to accelerate cell invasion. In addition, cancer cell-fibroblast interplay generates biologically active fragments of basement membrane proteins, such as endostatin.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Autoantígenos/metabolismo , Membrana Basal/metabolismo , Catepsina L/metabolismo , Comunicación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Masculino , Espectrometría de Masas , Invasividad Neoplásica , Colágenos no Fibrilares/metabolismo , Células PC-3 , Proteómica/métodos , Esferoides Celulares , Colágeno Tipo XVII
15.
BMC Cancer ; 20(1): 1180, 2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33267790

RESUMEN

BACKGROUND: Anchoring filament protein ladinin-1 (LAD1) was related to the aggressive progression of breast, lung, laryngeal and thyroid cancers. However, the association of LAD1 with colorectal cancer remained unknown. Here, to determine the relationship of LAD1 with colorectal cancer progression, we explored the effect of LAD1 loss on the malignant features of colorectal cancer cells. METHODS: We constructed LAD1-depleted cell lines and examined the effect of LAD1 deficiency on the phenotypic and molecular features of colorectal cancer cells in vitro. The function of LAD1 in metastasis in vivo was examined by establishing a spleen-to-liver metastasis mouse model. LAD1 protein expression in colorectal cancer patient specimens was assessed by immunohistochemistry of tumor microarrays. RESULTS: We found that LAD1 was abundant in most colorectal cancer cells. In addition, high expression of LAD1 significantly correlated with poor patient outcome. LAD1 depletion inhibited the migration and invasion of two different colorectal cancer cell lines, SW620 and Caco-2, without affecting their proliferation. In addition, LAD1 loss led to defects in liver metastasis of SW620 cells in the mouse model. Immunohistochemistry of colorectal cancer tissues revealed LAD1 enrichment in metastatic tissues compared to that in primary tumor and normal tissues. CONCLUSION: These results suggest that LAD1 expression is associated with the metastatic progression of colorectal cancer by promoting the migration and invasion of cancer cells.


Asunto(s)
Autoantígenos/metabolismo , Neoplasias Colorrectales/metabolismo , Colágenos no Fibrilares/metabolismo , Animales , Neoplasias Colorrectales/mortalidad , Femenino , Ratones , Metástasis de la Neoplasia , Análisis de Supervivencia , Transfección , Colágeno Tipo XVII
16.
J Pathol ; 247(3): 371-380, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30426510

RESUMEN

Immunoglobulins (Igs) consist of two antigen-binding regions (Fab) and one constant region (Fc). Protein A and protein G are bacterial proteins used for the purification of IgG by virtue of their high affinities for the Fc fragment. Rheumatoid factors are autoantibodies against IgG Fc fragments, which are present in the body under physiological conditions. Little is known about the influence of Fc-binding proteins on the pathogenicity of antibody-induced autoimmune diseases. Pemphigoid diseases are a group of autoimmune subepidermal blistering disorders that includes bullous pemphigoid and mucous membrane pemphigoid. IgGs targeting the non-collagenous NC16A domain of the 180-kDa bullous pemphigoid antigen (BP180) are known to induce skin fragility in mice and the depletion of BP180 in keratinocytes. In this study, mAb against NC16A in combination with Fc-binding proteins was found to enhance BP180 depletion. Although mAb against the C-terminus of BP180 does not show pathogenicity in vivo or in vitro, mAb treatment with Fc-binding proteins clearly induced skin fragility in mice and BP180 depletion in keratinocytes. Anti-BP180 mAbs and Fc-binding proteins were colocalized in the cytoplasm and at the basement membrane zone. Cell adhesion strengths were decreased in parallel with BP180 amounts. Clinically, bullous pemphigoid patients had higher rheumatoid factor titers than controls. Anti-BP180 mAb in combination with high-titer rheumatoid factor serum was found to enhance BP180 depletion. Furthermore, saliva from mucous membrane pemphigoid patients contained larger quantities of bacteria and Fc-binding proteins than controls. Our results suggest that Fc-binding proteins (rheumatoid factor or protein G) may enhance the pathogenicity of autoantibodies in pemphigoid diseases. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Autoantígenos/metabolismo , Enfermedades Autoinmunes/inmunología , Colágenos no Fibrilares/metabolismo , Penfigoide Ampolloso/inmunología , Receptores Fc/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/patología , Proteínas Portadoras/inmunología , Células Cultivadas , Femenino , Humanos , Inmunoglobulina G/inmunología , Queratinocitos/metabolismo , Masculino , Ratones Transgénicos , Penfigoide Benigno de la Membrana Mucosa/inmunología , Penfigoide Benigno de la Membrana Mucosa/patología , Penfigoide Ampolloso/patología , Factor Reumatoide/sangre , Saliva/inmunología , Colágeno Tipo XVII
17.
Lab Invest ; 99(1): 48-57, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30089857

RESUMEN

The basement membrane zone (BMZ) is framed by hemidesmosomes and extracellular matrix (ECM) including collagen IV (COL4). Hemidesmosomes are multiprotein complexes that include collagen XVII (COL17). BMZ proteins can be targeted in autoimmune subepidermal blistering diseases, e.g., pemphigoid targeting COL17. The blistering mechanisms in pemphigoid have not been fully elucidated, especially in mucous membrane pemphigoid (MMP), which mainly affects the mucosa. In this study, we showed that oral lesions in pemphigoid may be attributed to the inhibition of protein-protein interactions by autoantibodies. Using immunoprecipitation, we revealed that COL17 directly binds to COL4 in normal human keratinocytes and normal human oral keratinocytes. In particular, the C-terminus of COL17 is binding site to COL4 in oral keratinocytes. The precise COL4-binding region on COL17 was determined by protein-protein binding assay to be from amino acid Gly1175 to Asp1340 on the C-terminus. MMP-IgG or mAb recognizing the C-terminus hindered the interaction of COL17 with COL4 in oral keratinocytes. Furthermore, keratinocyte adhesion strength to COL4-coated plates was significantly reduced by the treatment of mAb against the C-terminus. In addition, the inflammatory infiltrates around perilesions were significantly less in MMP compared to BP. These results indicate that pemphigoid IgG targeting the C-terminus plays a pathogenic role in blister formation in the oral mucosa to inhibit protein interactions with less inflammation.


Asunto(s)
Autoantígenos/metabolismo , Colágeno Tipo IV/metabolismo , Colágenos no Fibrilares/metabolismo , Penfigoide Benigno de la Membrana Mucosa/inmunología , Penfigoide Ampolloso/inmunología , Autoanticuerpos/metabolismo , Células Cultivadas , Humanos , Queratinocitos/metabolismo , Mucosa Bucal/patología , Penfigoide Benigno de la Membrana Mucosa/metabolismo , Penfigoide Benigno de la Membrana Mucosa/patología , Penfigoide Ampolloso/metabolismo , Penfigoide Ampolloso/patología , Colágeno Tipo XVII
18.
Hum Mol Genet ; 26(3): 479-488, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28365758

RESUMEN

The function and stability of collagens depend on the accurate triple helix formation of three distinct polypeptide chains. Disruption of this triple-helical structure can result in connective-tissue disorders. Triple helix formation is thought to depend on three-stranded coiled-coil oligomerization sites within non-collagenous domains. However, only little is known about the physiological relevance of these coiled-coil structures. Transmembrane collagen XVII, also known as 180 kDa bullous pemphigoid antigen provides mechanical stability through the anchorage of epithelial cells to the basement membrane. Mutations in the collagen XVII gene, COL17A1, cause junctional epidermolysis bullosa (JEB), characterized by chronic trauma-induced skin blistering. Here we exploited a novel naturally occurring COL17A1 mutation, leading to an in-frame lysine duplication within the coiled-coil structure of the juxtamembranous NC16A domain of collagen XVII, which resulted in a mild phenotype of JEB due to reduced membrane-anchored collagen XVII molecules. This mutation causes structural changes in the mutant molecule and interferes with its maturation. The destabilized coiled-coil structure of the mutant collagen XVII unmasks a furin cleavage site that results in excessive and non-physiological ectodomain shedding during its maturation. Furthermore, it decreases its triple-helical stability due to defective coiled-coil oligomerization, which makes it highly susceptible to proteolytic degradation. As a consequence of altered maturation and decreased stability of collagen XVII trimers, reduced collagen XVII is incorporated into the cell membrane, resulting in compromised dermal-epidermal adhesion. Taken together, using this genetic model, we provide the first proof that alteration of the coiled-coil structure destabilizes oligomerization and impairs physiological shedding of collagen XVII in vivo.


Asunto(s)
Autoantígenos/genética , Epidermólisis Ampollosa de la Unión/genética , Proteínas Mutantes/genética , Colágenos no Fibrilares/genética , Relación Estructura-Actividad , Adolescente , Aminoácidos/genética , Autoantígenos/química , Autoantígenos/metabolismo , Vesícula/fisiopatología , Epidermólisis Ampollosa de la Unión/metabolismo , Epidermólisis Ampollosa de la Unión/patología , Femenino , Furina/genética , Humanos , Masculino , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Colágenos no Fibrilares/química , Colágenos no Fibrilares/metabolismo , Conformación Proteica en Hélice alfa/genética , Multimerización de Proteína , Estabilidad Proteica , Colágeno Tipo XVII
19.
Biochem Biophys Res Commun ; 514(4): 1115-1121, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31101337

RESUMEN

Cancer cells can invade as a population in various cancer tissues. This phenomenon is called collective invasion, which is associated with the metastatic potential and prognosis of cancer patients. The collectiveness of cancer cells is necessary for collective invasion. However, the mechanism underlying the generation of collectiveness by cancer cells is not well known. In this study, the phenomenon of contact following, where neighboring cells move in the same direction via intercellular adhesion, was investigated. An experimental system was created to observe the two-dimensional invasion using a collagen gel overlay to study contact following in collective invasion. The role of integrin-ß1, one of the major extracellular matrix (ECM) receptors, in contact following was examined through the experimental system. Integrin-ß1 was localized to the intercellular site in squamous carcinoma cells. Moreover, the intercellular adhesion and contact following were suppressed by treatment of an integrin-ß1 inhibitory antibody. ECM proteins such as laminin-332 and type-XVII collagen were also localized to the intercellular site and critical for contact following. Collectively, it was demonstrated that the activity of integrin-ß1 and expression of ECM proteins in the intercellular site promote contact following in the collective invasion of a cancer cell population.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Integrina beta1/metabolismo , Colágenos no Fibrilares/metabolismo , Neoplasias Cutáneas/metabolismo , Autoantígenos/biosíntesis , Carcinoma de Células Escamosas/patología , Adhesión Celular , Moléculas de Adhesión Celular/biosíntesis , Humanos , Integrina beta1/biosíntesis , Colágenos no Fibrilares/biosíntesis , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas , Kalinina , Colágeno Tipo XVII
20.
Mol Ther ; 26(1): 17-30, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29055623

RESUMEN

Squamous cell carcinoma (SCC) is one of the most common skin cancers and causes significant morbidity. Although the expression of the epithelial adhesion molecule collagen XVII (ColXVII) has been linked to SCC invasion, only little is known about its mechanistic contribution. Here, we demonstrate that ColXVII expression is essential for SCC cell proliferation and motility. Moreover, it revealed that particularly the post-translational modification of ColXVII by ectodomain shedding is the major driver of SCC progression, because ectodomain-selective immunostaining was mainly localized at the invasive front of human cutaneous SCCs, and exclusive expression of a non-sheddable ColXVII mutant in SCC-25 cells inhibits their matrix-independent growth and invasiveness. This cell surface proteolysis, which is strongly elevated during SCC invasion and metastasis, releases soluble ectodomains and membrane-anchored endodomains. Both released ColXVII domains play distinct roles in tumor progression: the endodomain induces proliferation and survival, whereas the ectodomain accelerates invasiveness. Furthermore, specific blockage of shedding by monoclonal ColXVII antibodies repressed matrix-independent growth and invasion of SCC cells in organotypic co-cultures. Thus, selective inhibition of ColXVII shedding may offer a promising therapeutic strategy to prevent SCC progression.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Membrana Celular/metabolismo , Colágenos no Fibrilares/metabolismo , Animales , Autoantígenos/química , Autoantígenos/genética , Biomarcadores , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ectodermo/metabolismo , Expresión Génica , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Colágenos no Fibrilares/química , Colágenos no Fibrilares/genética , Unión Proteica , Proteolisis , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Colágeno Tipo XVII
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda