Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
BMC Cancer ; 23(1): 949, 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803411

RESUMO

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.


Assuntos
Neoplasias do Colo , Neoplasias Retais , Humanos , Prognóstico , Colágenos não Fibrilares/metabolismo , Colágeno/química , Autoantígenos/metabolismo , Biomarcadores , Colágeno Tipo XVII
2.
Nature ; 608(7924): 784-794, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35798028

RESUMO

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.


Assuntos
Neoplasias do Colo , Células-Tronco Neoplásicas , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula , Proliferação de Células , Rastreamento de Células , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Quinase 1 de Adesão Focal/metabolismo , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Colágenos não Fibrilares/metabolismo , Organoides/metabolismo , Organoides/patologia , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição/metabolismo , Colágeno Tipo XVII
3.
Int J Mol Sci ; 23(8)2022 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-35456962

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/patologia , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Feminino , Humanos , Masculino , Colágenos não Fibrilares/metabolismo , Neoplasias Pancreáticas/patologia , Microambiente Tumoral , Neoplasias Pancreáticas
4.
J Cell Biol ; 220(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34550317

RESUMO

Skin regenerative capacity declines with age, but the underlying mechanisms are largely unknown. Here we demonstrate a functional link between epidermal growth factor receptor (EGFR) signaling and type XVII collagen (COL17A1) proteolysis on age-associated alteration of keratinocyte stem cell dynamics in skin regeneration. Live-imaging and computer simulation experiments predicted that human keratinocyte stem cell motility is coupled with self-renewal and epidermal regeneration. Receptor tyrosine kinase array identified the age-associated decline of EGFR signaling in mouse skin wound healing. Culture experiments proved that EGFR activation drives human keratinocyte stem cell motility with increase of COL17A1 by inhibiting its proteolysis through the secretion of tissue inhibitor of metalloproteinases 1 (TIMP1). Intriguingly, COL17A1 directly regulated keratinocyte stem cell motility and collective cell migration by coordinating actin and keratin filament networks. We conclude that EGFR-COL17A1 axis-mediated keratinocyte stem cell motility drives epidermal regeneration, which provides a novel therapeutic approach for age-associated impaired skin regeneration.


Assuntos
Autoantígenos/metabolismo , Movimento Celular/fisiologia , Colágenos não Fibrilares/metabolismo , Regeneração/fisiologia , Pele/metabolismo , Células 3T3 , Animais , Linhagem Celular , Células Epidérmicas/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Cicatrização/fisiologia , Colágeno Tipo XVII
5.
PLoS One ; 16(7): e0255179, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34293053

RESUMO

Collagen XVII (COL17), a cell-matrix adhesion protein, has been found to be suppressed in breast cancer. Our previous data demonstrated a preventive role of COL17 in breast cancer invasiveness. The present study used the stable COL17-overexpressing MCF7 and MDA-MB-231 cells to reveal an anti-proliferative effect of COL17 on breast cancer cell through mTOR deactivation. Cell proliferation was negatively correlated with the expression level of COL17 in a concentration-dependent manner in both conventional and three-dimensional (3D) culture systems. The correlation was confirmed by decreased expression of the proliferative marker Ki67 in COL17-expressing cells. In addition, overexpression of COL17 reduced the clonogenicity and growth of the cells. We demonstrated that COL17 affects the AKT/mTOR signaling pathway by deactivation of AKT, mTOR and downstream effectors, particularly 4EBP1. Moreover, mice xenografted with high COL17-expressing cells exhibited delayed tumor progression and prolonged survival time. The high expression of COL17A1 gene encoding COL17 is associated with low-proliferation tumors, extended tumor-free period, and overall survival of breast cancer patients. In conclusion, our results revealed the novel function of COL17 using in vitro and in vivo models and elucidated the related pathway in breast cancer cell growth and proliferation.


Assuntos
Autoantígenos/metabolismo , Neoplasias da Mama/patologia , Colágenos não Fibrilares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Autoantígenos/genética , Neoplasias da Mama/genética , Proliferação de Células/efeitos dos fármacos , Células Clonais , Progressão da Doença , Doxiciclina/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Antígeno Ki-67/metabolismo , Modelos Biológicos , Análise Multivariada , Colágenos não Fibrilares/genética , Modelos de Riscos Proporcionais , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Análise de Sobrevida , Colágeno Tipo XVII
6.
Curr Biol ; 31(14): 3086-3097.e7, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34087104

RESUMO

At the early stage of cancer development, oncogenic mutations often cause multilayered epithelial structures. However, the underlying molecular mechanism still remains enigmatic. By performing a series of screenings targeting plasma membrane proteins, we have found that collagen XVII (COL17A1) and CD44 accumulate in RasV12-, Src-, or ErbB2-transformed epithelial cells. In addition, the expression of COL17A1 and CD44 is also regulated by cell density and upon apical cell extrusion. We further demonstrate that the expression of COL17A1 and CD44 is profoundly upregulated at the upper layers of multilayered, transformed epithelia in vitro and in vivo. The accumulated COL17A1 and CD44 suppress mitochondrial membrane potential and reactive oxygen species (ROS) production. The diminished intracellular ROS level then promotes resistance against ferroptosis-mediated cell death upon cell extrusion, thereby positively regulating the formation of multilayered structures. To further understand the functional role of COL17A1, we performed comprehensive metabolome analysis and compared intracellular metabolites between RasV12 and COL17A1-knockout RasV12 cells. The data imply that COL17A1 regulates the metabolic pathway from the GABA shunt to mitochondrial complex I through succinate, thereby suppressing the ROS production. Moreover, we demonstrate that CD44 regulates membrane accumulation of COL17A1 in multilayered structures. These results suggest that CD44 and COL17A1 are crucial regulators for the clonal expansion of transformed cells within multilayered epithelia, thus being potential targets for early diagnosis and preventive treatment for precancerous lesions.


Assuntos
Transformação Celular Neoplásica , Epitélio/crescimento & desenvolvimento , Receptores de Hialuronatos/metabolismo , Colágenos não Fibrilares/metabolismo , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Cães , Ferroptose , Humanos , Células Madin Darby de Rim Canino , Potencial da Membrana Mitocondrial , Camundongos , Espécies Reativas de Oxigênio
7.
Biofactors ; 47(4): 627-644, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33864298

RESUMO

Bleomycin (BLM) injury is associated with the severity of acute lung injury (ALI) leading to fibrosis, a high-morbidity, and high-mortality respiratory disease of unknown etiology. BLM-induced ALI is marked by the activation of a potent fibrogenic cytokine transcription growth factor beta-1 (TGFß-1), which is considered a critical cytokine in the progression of alveolar injury. Previously, our work demonstrated that a diet-derived compound curcumin (diferuloylmethane), represents its antioxidative and antifibrotic application in TGF-ß1-mediated BLM-induced alveolar basal epithelial cells. However, curcumin-specific protein targets, as well as its mechanism using mass spectrometry-based proteomic approach, remain elusive. To elucidate the underlying mechanism, a quantitative proteomics approach and bioinformatics analysis were employed to identify the protein targets of curcumin in BLM or TGF-ß1-treated cells. With subsequent in vitro experiments, curcumin-related pathways and cellular processes were predicted and validated. The current study discusses two separate proteomics experiments using BLM and TGF-ß1-treated cells with the proteomics approach, various unique target proteins were identified, and proteomic analysis revealed that curcumin reversed the expressions of unique proteins like DNA topoisomerase 2-alpha (TOP2A), kinesin-like protein (KIF11), centromere protein F (CENPF), and so on BLM or TGF-ß1 injury. For the first time, the current study reveals that curcumin restores TGF-ß1 induced peroxisomes like PEX-13, PEX-14, PEX-19, and ACOX1. This was verified by subsequent in vitro assays. This study generated molecular evidence to deepen our understanding of the therapeutic role of curcumin at the proteomic level and may be useful to identify molecular targets for future drug discovery.


Assuntos
Antioxidantes/farmacologia , Bleomicina/antagonistas & inibidores , Curcumina/farmacologia , Proteômica/métodos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Células A549 , Acetil-CoA C-Acetiltransferase/genética , Acetil-CoA C-Acetiltransferase/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/prevenção & controle , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Antibióticos Antineoplásicos/farmacologia , Antioxidantes/química , Antioxidantes/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Sítios de Ligação , Bleomicina/farmacologia , Calreticulina/genética , Calreticulina/metabolismo , Curcumina/química , Curcumina/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Modelos Biológicos , Simulação de Acoplamento Molecular , Colágenos não Fibrilares/genética , Colágenos não Fibrilares/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia , Colágeno Tipo XVII
8.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33805154

RESUMO

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.


Assuntos
Autoantígenos/metabolismo , Epidermólise Bolhosa Juncional/genética , Colágenos não Fibrilares/metabolismo , Oligonucleotídeos Antissenso/genética , Splicing de RNA , Processamento Alternativo , Biópsia , Linhagem Celular , Sobrevivência Celular , Epidermólise Bolhosa Juncional/metabolismo , Epidermólise Bolhosa Juncional/terapia , Éxons , Genótipo , Homozigoto , Humanos , Queratinócitos/citologia , Lipossomos/química , Mutação , Técnicas de Cultura de Órgãos , RNA Mensageiro/metabolismo , Colágeno Tipo XVII
9.
Cell Rep ; 34(10): 108819, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33691112

RESUMO

The upper gastrointestinal tract, consisting of the esophagus, stomach, and duodenum, controls food transport, digestion, nutrient uptake, and hormone production. By single-cell analysis of healthy epithelia of these human organs, we molecularly define their distinct cell types. We identify a quiescent COL17A1high KRT15high stem/progenitor cell population in the most basal cell layer of the esophagus and detect substantial gene expression differences between identical cell types of the human and mouse stomach. Selective expression of BEST4, CFTR, guanylin, and uroguanylin identifies a rare duodenal cell type, referred to as BCHE cell, which likely mediates high-volume fluid secretion because of continual activation of the CFTR channel by guanylin/uroguanylin-mediated autocrine signaling. Serotonin-producing enterochromaffin cells in the antral stomach significantly differ in gene expression from duodenal enterochromaffin cells. We, furthermore, discover that the histamine-producing enterochromaffin-like cells in the oxyntic stomach express the luteinizing hormone, yet another member of the enteroendocrine hormone family.


Assuntos
Duodeno/citologia , Esôfago/citologia , Estômago/citologia , Trato Gastrointestinal Superior/citologia , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Bestrofinas/genética , Bestrofinas/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Duodeno/metabolismo , Duodeno/patologia , Esôfago/metabolismo , Esôfago/patologia , Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Queratina-15/genética , Queratina-15/metabolismo , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Colágenos não Fibrilares/genética , Colágenos não Fibrilares/metabolismo , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/metabolismo , Estômago/metabolismo , Estômago/patologia , Trato Gastrointestinal Superior/metabolismo , Trato Gastrointestinal Superior/patologia , Colágeno Tipo XVII
10.
Anticancer Res ; 41(2): 1089-1099, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33517320

RESUMO

BACKGROUND/AIM: Prognosis plays a vital role in head and neck squamous cell carcinoma (HNSCC) patient management and decision-making. This study aimed to identify the role of BP180 as a prognostic factor in HNSCC. PATIENTS AND METHODS: Protein expression of bullous pemphigoid antigen II (BP180) was verified by immunohistochemistry (IHC) in a tissue microarray study of 202 cases. RESULTS: IHC analysis revealed that protein expression of BP180 among HNSCC patients differed significantly in the presence and absence of neural invasion, and according to T status in laryngeal and pharyngeal cancer subgroups. Overall survival and multivariate analysis showed that positive BP180-IHC and advanced clinical stage were significant independent positive predictors of mortality in HNSCC patients. In addition, in the oral cancer subgroup, independent positive predictors were positive BP180-IHC, advanced N status and neural invasion. In laryngeal and pharyngeal cancer subgroups, predictors were positive BP180-IHC and advanced clinical stage. CONCLUSION: BP180 is a prognostic factor in head and neck squamous cell carcinoma.


Assuntos
Autoantígenos/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Colágenos não Fibrilares/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Tomada de Decisão Clínica , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Análise de Sobrevida , Análise Serial de Tecidos , Colágeno Tipo XVII
11.
Cells ; 9(12)2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33266100

RESUMO

Filamins are large dimeric F-actin cross-linking proteins, crucial for the mechanosensitive properties of a number of cell types. Due to their interaction with a variety of different proteins, they exert important regulatory functions. However, in the human testis the role of filamins has been insufficiently explored. Immunohistochemical staining of human testis samples identified filamin A (FLNA) in spermatogonia and peritubular myoid cells. Investigation of different testicular tumor samples indicated that seminoma also express FLNA. Moreover, mass spectrometric analyses identified FLNA as one of the most abundant proteins in human seminoma TCam-2 cells. We therefore focused on FLNA in TCam-2 cells, and identified by co-immunoprecipitation LAD1, RUVBL1 and DAZAP1, in addition to several cytoskeletal proteins, as interactors of FLNA. To study the role of FLNA in TCam-2 cells, we generated FLNA-deficient cells using the CRISPR/Cas9 system. Loss of FLNA causes an irregular arrangement of the actin cytoskeleton and mechanical instability, impaired adhesive properties and disturbed migratory behavior. Furthermore, transcriptional activity of typical stem cell factors is increased in the absence of FLNA. In summary, our data suggest that FLNA is crucially involved in balancing stem cell characteristics and invasive properties in human seminoma cells and possibly human testicular germ cells.


Assuntos
Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Filaminas/metabolismo , Seminoma/metabolismo , Células-Tronco/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Adulto , Idoso , Autoantígenos/metabolismo , Sistemas CRISPR-Cas/fisiologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Colágenos não Fibrilares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Neoplasias Testiculares/metabolismo , Testículo/metabolismo , Transcrição Gênica/fisiologia , Colágeno Tipo XVII
12.
BMC Cancer ; 20(1): 1180, 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267790

RESUMO

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.


Assuntos
Autoantígenos/metabolismo , Neoplasias Colorretais/metabolismo , Colágenos não Fibrilares/metabolismo , Animais , Neoplasias Colorretais/mortalidade , Feminino , Camundongos , Metástase Neoplásica , Análise de Sobrevida , Transfecção , Colágeno Tipo XVII
13.
J Immunol ; 205(10): 2786-2794, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32998984

RESUMO

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.


Assuntos
Autoantígenos/metabolismo , Medula Óssea/patologia , Leucopoese/imunologia , Células-Tronco Mesenquimais/metabolismo , Neutrófilos/fisiologia , Colágenos não Fibrilares/metabolismo , Animais , Autoantígenos/genética , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos/sangue , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Hiperplasia/genética , Hiperplasia/imunologia , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Colágenos não Fibrilares/genética , Domínios Proteicos/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Colágeno Tipo XVII
14.
Curr Med Sci ; 40(4): 795-800, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32862392

RESUMO

Non-Herlitz junctional epidermolysis bullosa (JEB-nH), an autosomal recessive bullous genodermatosis, is characterized by generalized skin blistering from birth onward, dental anomalies, universal alopecia and nail dystrophy. The underlying defect is mutation of the COL17A1 gene encoding the type XVII collagen, resulting in losing structure for attachment of basal epithelial cells to the matrix. In present study, we described one case of congenitally affected female child aged 10 years, with skin blistering. Dermatologic examination revealed sparse, mild blisters on the face and hand, with profound enamel pitting of the teeth. Skin biopsy from proband's bullous skin displayed subepidermal bulla formation without acantholysis. The immunofluorescence of anti-type XVII collagen antibody staining showed loss of type XVII collagen staining at the basement membrane zone. A combination of whole exome sequencing (WES) and Sanger sequencing revealed the novel heterozygous mutations (c.4324C>T;p.Q1442* and c.1834G>C;p.G612R) in COL17A1 gene, which could be associated with the observed JEB-nH. One allele had a novel nonsense mutation (c.4324C>T;p.Q1442*), resulting in nonsense-mediated mRNA decay and truncated collagen XVII; the other allele had a novel missense mutation of c.1834G>C;p.G612R in exon 22, causing a glycine-to-arginine substitution in the Gly-X-Y triple helical repeating motifs and decreasing the thermal stability of collagen XVII. Our findings indicate that the genetic test based on WES can be useful in diagnosing JEB-nH patients. The novel pathogenic mutations identified would further expand our understanding of the mutation spectrum of COL17A1 gene in association with the inherited blistering diseases.


Assuntos
Autoantígenos/genética , Epidermólise Bolhosa Juncional/diagnóstico , Mutação de Sentido Incorreto , Colágenos não Fibrilares/genética , Degradação do RNAm Mediada por Códon sem Sentido , Autoantígenos/química , Autoantígenos/metabolismo , Biópsia , Criança , China , Epidermólise Bolhosa Juncional/genética , Epidermólise Bolhosa Juncional/patologia , Feminino , Heterozigoto , Humanos , Colágenos não Fibrilares/química , Colágenos não Fibrilares/metabolismo , Linhagem , Estabilidade Proteica , Sequenciamento do Exoma , Colágeno Tipo XVII
15.
Prostate ; 80(9): 715-726, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32364250

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Proteínas de Membrana/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Autoantígenos/metabolismo , Membrana Basal/metabolismo , Catepsina L/metabolismo , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Masculino , Espectrometria de Massas , Invasividade Neoplásica , Colágenos não Fibrilares/metabolismo , Células PC-3 , Proteômica/métodos , Esferoides Celulares , Colágeno Tipo XVII
16.
Semin Cancer Biol ; 62: 134-148, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31479735

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Neoplasias/metabolismo , Colágenos não Fibrilares/metabolismo , Células-Tronco/metabolismo , Animais , Suscetibilidade a Doenças , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Neoplasias/etiologia , Neoplasias/patologia , Proteólise , Nicho de Células-Tronco/genética , Relação Estrutura-Atividade , Microambiente Tumoral
17.
Biochem Biophys Res Commun ; 514(4): 1115-1121, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31101337

RESUMO

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.


Assuntos
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adesão Celular/metabolismo , Integrina beta1/metabolismo , Colágenos não Fibrilares/metabolismo , Neoplasias Cutâneas/metabolismo , Autoantígenos/biossíntese , Carcinoma de Células Escamosas/patologia , Adesão Celular , Moléculas de Adesão Celular/biossíntese , Humanos , Integrina beta1/biossíntese , Colágenos não Fibrilares/biossíntese , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas , Calinina , Colágeno Tipo XVII
18.
J Invest Dermatol ; 139(6): 1221-1226, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31068252

RESUMO

Epidermolysis bullosa is a family of diseases characterized by blistering and fragility of the skin in response to mechanical trauma. Advances in our understanding of epidermolysis bullosa pathophysiology have provided the necessary foundation for the first clinical trials of gene therapy for junctional and dystrophic epidermolysis bullosa. These therapies show that gene therapy is both safe and effective, with the potential to correct the molecular and clinical phenotype of patients with epidermolysis bullosa. Improvements in gene delivery and in preventing immune reactions will be among the challenges that lie ahead during further therapeutic development.


Assuntos
Células Epidérmicas/transplante , Epidermólise Bolhosa/terapia , Terapia Genética/métodos , Vetores Genéticos/genética , Rejeição de Enxerto/prevenção & controle , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Técnicas de Cultura de Células/métodos , Ensaios Clínicos como Assunto , Clonagem Molecular , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Modelos Animais de Doenças , Células Epidérmicas/metabolismo , Epidermólise Bolhosa/genética , Técnicas de Transferência de Genes , Engenharia Genética/métodos , Terapia Genética/efeitos adversos , Rejeição de Enxerto/imunologia , Humanos , Mutação , Colágenos não Fibrilares/genética , Colágenos não Fibrilares/metabolismo , Retroviridae/genética , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos , Resultado do Tratamento , Calinina , Colágeno Tipo XVII
19.
J Biochem ; 166(2): 163-173, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30918974

RESUMO

Collagen type XVII (COL17) is expressed in various tissues and its aberrant expression is associated with tumour progression. In this study, we investigated the regulation of COL17 expression in oral squamous cell carcinoma (OSCC) using the cell lines NA, SAS, Ca9-22, and Sa3. COL17 was induced upon p53 activation by cisplatin in SAS; however, this effect was more limited in NA and hardly in Ca9-22 and Sa3, with mutated p53. Moreover, COL17 was found to be regulated by miR203a-3p in all cell lines. Our data suggest that COL17 expression in OSCC cell lines is regulated by p53 and miR203a-3p.


Assuntos
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , MicroRNAs/metabolismo , Neoplasias Bucais/metabolismo , Colágenos não Fibrilares/metabolismo , Autoantígenos/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Bucais/patologia , Colágenos não Fibrilares/genética , Reação em Cadeia da Polimerase , Proteína Supressora de Tumor p53/metabolismo , Colágeno Tipo XVII
20.
Sci Rep ; 9(1): 3525, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30837635

RESUMO

Bullous Pemphigoid is the most common auto-immune bullous skin disease. It is characterized by the production of auto-antibodies directed against 2 proteins of the hemi-desmosome (BP180 and BP230). We assessed the efficacy and mechanisms of action of rituximab, an anti-CD20 monoclonal antibody, in 17 patients with severe and relapsing type of bullous pemphigoid. The phenotype, cytokine gene expression, and rearrangement of BP180-specific B-cell receptor genes were performed over 2 years following treatment. At the end of the study, 5 patients had died, 3 had withdrawn from the study, and 9 patients were in complete remission. The one- and two-year relapse rates were 44.1% (95% Confidence Interval (CI): 21.0-76.0%) and 66.5%, (95% CI: 38.4-91.4%), respectively. Phenotypic analyses confirmed dramatic B-cell depletion, which lasted for 9 to 12 months. The ELISA values of serum anti-BP180 antibodies and the frequency of BP180-specific circulating B cells decreased dramatically following treatment, which paralleled the improvement of skin lesions. During B-cell reconstitution, a polyclonal IgM repertoire appeared and a shift in the rearrangement of the B-cell receptor genes of BP180-specific circulating B cells was observed. Concurrently, we observed a decrease of IL-15, IL-6 and TNFα expressing BP180-specific B cells, and the emergence of IL-10 and IL-1RA-expressing BP180-specific IgM+ B cells in patients in complete remission off therapy, suggesting the functional plasticity of BP180-specific auto-immune B cells after rituximab treatment.


Assuntos
Autoantígenos/metabolismo , Linfócitos B/metabolismo , Citocinas/metabolismo , Colágenos não Fibrilares/metabolismo , Penfigoide Bolhoso/patologia , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/sangue , Autoantígenos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Interleucina-10/metabolismo , Interleucina-15/metabolismo , Interleucina-6/metabolismo , Masculino , Colágenos não Fibrilares/imunologia , Penfigoide Bolhoso/tratamento farmacológico , Penfigoide Bolhoso/imunologia , Fenótipo , Recidiva , Rituximab/uso terapêutico , Colágeno Tipo XVII
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA