Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.160
Filtrar
1.
Sci Rep ; 14(1): 24104, 2024 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-39406784

RESUMEN

Breast cancer is one of the most common cancers in women. One of the best therapeutic methods against breast cancer is gene therapy, while having an appropriate gene carrier is the biggest challenge of gene therapy. Hence, developing carriers with low cytotoxicity and high gene transfection efficiency, and preferentially with the selective function of gene delivery is a critical demand for this method. In the present study, we introduce a novel targeted carrier to deliver the inducible caspase-9 suicide gene (pLVSIN-iC9) into breast cancer cells. The carrier is composed of graphene oxide quantum dots decorated with polyethyleneimine, and S2.2; an aptamer with high affinity to MUC1 (GOQD-PEI/S2.2). Due to the overexpression of MUC1 in breast cancer cells, the designed GOQD-PEI/S2.2/pLVSIN-iC9 can selectively target cancer cells. Moreover, to better mimic solid tumor conditions, and to evaluate the selective effect of the GOQD-PEI/S2.2/pLVSIN-iC9, an organoid model derived from human dermal fibroblasts (HDF) and MCF-7 cells (coculture organoid) was generated and characterized. The results demonstrate that the coculture organoid model adapts the tissue structure of luminal breast cancer, as well. Therefore, the organoids were subjected to treatment with targeted gene therapy using GOQD-PEI/S2.2/pLVSIN-iC9. Our evidence supports the targeted killing effect of iC9 on the breast cancer cells of the organoids and suggests the good potential of the newly introduced carriers in targeted gene delivery.


Asunto(s)
Aptámeros de Nucleótidos , Neoplasias de la Mama , Genes Transgénicos Suicidas , Terapia Genética , Grafito , Organoides , Puntos Cuánticos , Humanos , Grafito/química , Puntos Cuánticos/química , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Neoplasias de la Mama/patología , Terapia Genética/métodos , Aptámeros de Nucleótidos/genética , Organoides/metabolismo , Células MCF-7 , Caspasa 9/metabolismo , Caspasa 9/genética , Polietileneimina/química , Técnicas de Transferencia de Gen , Mucina-1/genética , Mucina-1/metabolismo
2.
Cell Death Dis ; 15(9): 709, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39349458

RESUMEN

Breast cancer is the most frequent type of tumor in women and is characterized by variable outcomes due to its heterogeneity and the presence of many cancer cell-autonomous and -non-autonomous factors. A major determinant of breast cancer aggressiveness is represented by immune infiltration, which can support tumor development. In our work, we studied the role of mast cells in breast cancer and identified a novel activity in promoting the tumor-initiating properties of cancer cells. Mast cells are known to affect breast cancer prognosis, but show different effects according to the diverse subtypes. Starting from the observation that co-injection of mast cells with limiting concentrations of cancer cells increased their in vivo engraftment rate, we characterized the molecular mechanisms by which mast cells promote the tumor stem-like features. We provide evidence that mast cell heparanase plays a pivotal role since both its activity and the stimulation of mast cells with heparan sulfate, the product of heparanase activity, are crucial for this process. Moreover, the pharmacological inhibition of heparanase prevents the function of mast cells. Our data show that soluble factors released by mast cells favor the expression of estrogen receptor in a MUC1-dependent manner. The MUC1/estrogen receptor axis is eventually essential for cancer stem-like features, specifically in HER2-negative cells, and promotes the capability of cancer cells to form mammospheres and express stem-related genes, also reducing their sensitivity to tamoxifen administration. Altogether our findings describe a novel mechanism by which mast cells could increase the aggressiveness of breast cancer uncovering a molecular mechanism displaying differences based on the specific breast cancer subtype.


Asunto(s)
Neoplasias de la Mama , Glucuronidasa , Mastocitos , Mucina-1 , Células Madre Neoplásicas , Receptores de Estrógenos , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Mastocitos/metabolismo , Femenino , Receptores de Estrógenos/metabolismo , Mucina-1/metabolismo , Mucina-1/genética , Glucuronidasa/metabolismo , Glucuronidasa/genética , Animales , Ratones , Línea Celular Tumoral , Transducción de Señal
3.
Nat Commun ; 15(1): 8416, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341835

RESUMEN

Hypoxia occurs in 90% of solid tumors and is associated with metastasis and mortality. Breast cancer cells that experience intratumoral hypoxia are 5x more likely to develop lung metastasis in animal models. Using spatial transcriptomics, we determine that hypoxic cells localized in more oxygenated tumor regions (termed 'post-hypoxic') retain expression of hypoxia-inducible and NF-kB-regulated genes, even in the oxygen-rich bloodstream. This cellular response is reproduced in vitro under chronic hypoxic conditions followed by reoxygenation. A subset of genes remains increased in reoxygenated cells. MUC1/MUC1-C is upregulated by both HIF-1α and NF-kB-p65 during chronic hypoxia. Abrogating MUC1 decreases the expression of superoxide dismutase enzymes, causing reactive oxygen species (ROS) production and cell death. A hypoxia-dependent genetic deletion of MUC1, or MUC1-C inhibition by GO-203, increases ROS levels in circulating tumor cells (CTCs), reducing the extent of metastasis. High MUC1 expression in tumor biopsies is associated with recurrence, and MUC1+ CTCs have lower ROS levels than MUC1- CTCs in patient-derived xenograft models. This study demonstrates that therapeutically targeting MUC1-C reduces hypoxia-driven metastasis.


Asunto(s)
Neoplasias de la Mama , Mucina-1 , Especies Reactivas de Oxígeno , Mucina-1/metabolismo , Mucina-1/genética , Humanos , Animales , Especies Reactivas de Oxígeno/metabolismo , Femenino , Línea Celular Tumoral , Ratones , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Oxígeno/metabolismo , Factor de Transcripción ReIA/metabolismo , Metástasis de la Neoplasia , Hipoxia/metabolismo , Hipoxia de la Célula
4.
Int Immunopharmacol ; 142(Pt B): 113204, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39317052

RESUMEN

Mucin 1 plays an important role in tumor signaling and is overexpressed in adenocarcinoma and the digestive system. Many antibodies have been developed against MUC1 targets. Previously developed antibodies were mainly directed against distal membrane-terminal MUC1-N, but distal membrane-terminal MUC1-N is shed during cell growth and therefore binds to antibodies developed against tandem repeat sequences and becomes ineffective. Here, we provide a simple and rapid method for preparing antibodies targeting the proximal membrane end of MUC1. Immunological target antigens were designed based on Biocytogen Renlite KO mice. With the help of B-cell high-throughput screening technology, we rapidly screened and prepared fully human antibodies with human-macaque cross-reactivity, high affinity, high specificity, and endocytosis. Using this method, we screened 40 antibodies with human-monkey cross-reactivity, which specifically recognized breast cancer cell lines with human and monkey affinities ranging from (1.04E-07-2.91E-09). Of these, the antibodies with germline genes IGHV4-59*01 and IGHV3-30*03 had nanomolar affinities, with high endocytosis effects in breast cancer cells. Ab.07 (IGHV3-30*03) coupled with monomethyl auristatin E (MMAE) showed good anti-tumor activity in different tumor cells. In summary, we describe a method for designing and producing excellent antibodies that can be assembled into antibody-drug conjugates and bispecific antibodies by proximal-membrane-end immunization and B-cell high-throughput screening that can rapidly generate high-quality antibodies.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Mucina-1 , Humanos , Animales , Ensayos Analíticos de Alto Rendimiento/métodos , Mucina-1/inmunología , Mucina-1/genética , Ratones , Línea Celular Tumoral , Linfocitos B/inmunología , Ratones Noqueados , Reacciones Cruzadas , Femenino , Endocitosis , Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/inmunología , Oligopéptidos
5.
Sci Adv ; 10(35): eadn9857, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39213364

RESUMEN

Solid tumors, such as triple-negative breast cancer (TNBC), are biologically complex due to cellular heterogeneity, lack of tumor-specific antigens, and an immunosuppressive tumor microenvironment (TME). These challenges restrain chimeric antigen receptor (CAR) T cell efficacy, underlining the importance of armoring. In solid cancers, a localized tumor mass allows alternative administration routes, such as intratumoral delivery with the potential to improve efficacy and safety but may compromise metastatic-site treatment. Using a multi-layered CAR T cell engineering strategy that allowed a synergy between attributes, we show enhanced cytotoxic activity of MUC1 CAR T cells armored with PD1KO, tumor-specific interleukin-12 release, and TGFBR2KO attributes catered towards the TNBC TME. Intratumoral treatment effectively reduced distant tumors, suggesting retention of antigen-recognition benefits at metastatic sites. Overall, we provide preclinical evidence of armored non-alloreactive MUC1 CAR T cells greatly reducing high TNBC tumor burden in a TGFB1- and PD-L1-rich TME both at local and distant sites while preserving safety.


Asunto(s)
Inmunoterapia Adoptiva , Mucina-1 , Receptores Quiméricos de Antígenos , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Mucina-1/metabolismo , Mucina-1/genética , Mucina-1/inmunología , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Humanos , Femenino , Animales , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Ratones , Inmunoterapia Adoptiva/métodos , Línea Celular Tumoral , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Talanta ; 279: 126665, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39116728

RESUMEN

Mucin 1 (MUC1) is frequently overexpressed in various cancers and is essential for early cancer detection. Current methods to detect MUC1 are expensive, time-consuming, and require skilled personnel. Therefore, developing a simple, sensitive, highly selective MUC1 detection sensor is necessary. In this study, we proposed a novel "signal-on-off" strategy that, in the presence of MUC1, synergistically integrates catalytic hairpin assembly (CHA) with DNA tetrahedron (Td)-based nonlinear hybridization chain reaction (HCR) to enhance the immobilization of electrochemically active methylene blue (MB) on magnetic nanoparticles (MNP), marking the MB signal "on". Concurrently, the activation of CRISPR-Cas12a by isothermal amplification products triggers the cleavage of single-stranded DNA (ssDNA) at the electrode surface, resulting in a reduction of MgAl-LDH@Fc-AuFe-MIL-101 (containing ferrocene, Fc) on the electrode, presenting the "signal-off" state. Both MB and MgAl-LDH@Fc-AuFe-MIL-101 electrochemical signals were measured and analyzed. Assay parameters were optimized, and sensitivity, stability, and linear range were assessed. Across a concentration spectrum of MUC1 spanning from 10 fg/mL to 100 ng/mL, the MB and MgAl-LDH@Fc-AuFe-MIL-101 signals were calibrated with each other, demonstrating a "signal-on-off" dual electrochemical signaling pattern. This allows for the precise and quantitative detection of MUC1 in clinical samples, offering significant potential for medical diagnosis.


Asunto(s)
Técnicas Biosensibles , Sistemas CRISPR-Cas , Técnicas Electroquímicas , Mucina-1 , Hibridación de Ácido Nucleico , Mucina-1/análisis , Mucina-1/genética , Técnicas Electroquímicas/métodos , Humanos , Técnicas Biosensibles/métodos , Sistemas CRISPR-Cas/genética , Azul de Metileno/química , Nanopartículas de Magnetita/química , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Electrodos , Límite de Detección , Oro/química
7.
Biochem Biophys Res Commun ; 731: 150402, 2024 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-39024979

RESUMEN

Given that the corneal epithelium is situated on the outermost part of the eye, its functions can be influenced by external temperatures and chemical substances. This study aimed to elucidate the expression profile of chemosensory receptors in corneal epithelial cells and analyze their role in eye function regulation. A comprehensive analysis of 425 chemosensory receptors in human corneal epithelial cells-transformed (HCE-T) revealed the functional expression of TRPV4. The activation of TRPV4 in HCE-T cells significantly increased the expression of membrane-associated mucins MUC1, MUC4, and MUC16, which are crucial for stabilizing tear films, with efficacy comparable to the active components of dry eye medications. The present study suggests that TRPV4, which is activated by body temperature, regulates mucin expression and proposes it as a novel target for dry eye treatment.


Asunto(s)
Epitelio Corneal , Mucina-1 , Mucina 4 , Mucinas , Canales Catiónicos TRPV , Humanos , Antígeno Ca-125/metabolismo , Antígeno Ca-125/genética , Células Epiteliales/metabolismo , Células Epiteliales/citología , Epitelio Corneal/metabolismo , Epitelio Corneal/citología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Mucina-1/metabolismo , Mucina-1/genética , Mucina 4/metabolismo , Mucina 4/genética , Mucinas/metabolismo , Mucinas/biosíntesis , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/genética
8.
Mol Ther ; 32(9): 3114-3127, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-38910324

RESUMEN

T cell-focused cancer immunotherapy including checkpoint inhibitors and cell therapies has been rapidly evolving over the past decade. Nevertheless, there remains a major unmet medical need in oncology generally and immuno-oncology specifically. We have constructed an oncolytic adenovirus, Ad5/3-E2F-d24-aMUC1aCD3-IL-2 (TILT-322), which is armed with a human aMUC1aCD3 T cell engager and IL-2. TILT-322 treatment stimulated T cell cytotoxicity through the increased presence of granzyme B, perforin, and interferon-gamma. Additional immune profiling indicated TILT-322 increased gamma delta T cell activation and impacted other cell types such as natural killer cells and natural killer-like T cells that are traditionally involved in cancer immunotherapy. TILT-322 treatment also decreased the proportion of exhausted CD8+ T cells as demarked by immune checkpoint expression in ovarian ascites samples. Overall, our data showed that TILT-322 treatment led to an enhanced T cell activation and reversed T cell exhaustion translating into high antitumor efficacy when given locally or intravenously. The analysis of blood and tumors isolated from an in vivo patient-derived ovarian cancer xenograft model suggested TILT-322 mediated tumor control through improved T cell functions. Therefore, TILT-322 is a promising novel anti-tumor agent for clinical translation.


Asunto(s)
Adenoviridae , Anticuerpos Biespecíficos , Ascitis , Interleucina-2 , Mucina-1 , Neoplasias Ováricas , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Neoplasias Ováricas/terapia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/genética , Femenino , Animales , Adenoviridae/genética , Ratones , Línea Celular Tumoral , Ascitis/terapia , Ascitis/inmunología , Interleucina-2/metabolismo , Mucina-1/genética , Mucina-1/inmunología , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Modelos Animales de Enfermedad , Inmunoterapia/métodos , Agotamiento de Células T
9.
Genes (Basel) ; 15(6)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38927743

RESUMEN

Ionizing radiation (IR) and chemotherapy with DNA-damaging drugs such as cisplatin are vital cancer treatment options. These treatments induce double-strand breaks (DSBs) as cytotoxic DNA damage; thus, the DSB repair activity in each cancer cell significantly influences the efficacy of the treatments. Pancreatic cancers are known to be resistant to these treatments, and the overexpression of MUC1, a member of the glycoprotein mucins, is associated with IR- and chemo-resistance. Therefore, we investigated the impact of MUC1 on DSB repair. This report examined the effect of the overexpression of MUC1 on homologous recombination (HR) and non-homologous end-joining (NHEJ) using cell-based DSB repair assays. In addition, the therapeutic potential of NHEJ inhibitors including HDAC inhibitors was also studied using pancreatic cancer cell lines. The MUC1-overexpression enhances NHEJ, while partially suppressing HR. Also, MUC1-overexpressed cancer cell lines are preferentially killed by a DNA-PK inhibitor and HDAC1/2 inhibitors. Altogether, MUC1 induces metabolic changes that create an imbalance between NHEJ and HR activities, and this imbalance can be a target for selective killing by HDAC inhibitors. This is a novel mechanism of MUC1-mediated IR-resistance and will form the basis for targeting MUC1-overexpressed pancreatic cancer.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Mucina-1 , Neoplasias Pancreáticas , Regulación hacia Arriba , Humanos , Mucina-1/genética , Mucina-1/metabolismo , Reparación del ADN por Unión de Extremidades/genética , Línea Celular Tumoral , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Recombinación Homóloga , Inhibidores de Histona Desacetilasas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
10.
Cell Death Dis ; 15(5): 330, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740827

RESUMEN

The long non-coding RNA X-inactive specific transcript (lncRNA XIST) and MUC1 gene are dysregulated in chronic inflammation and cancer; however, there is no known interaction of their functions. The present studies demonstrate that MUC1-C regulates XIST lncRNA levels by suppressing the RBM15/B, WTAP and METTL3/14 components of the m6A methylation complex that associate with XIST A repeats. MUC1-C also suppresses the YTHDF2-CNOT1 deadenylase complex that recognizes m6A sites and contributes to XIST decay with increases in XIST stability and expression. In support of an auto-regulatory pathway, we show that XIST regulates MUC1-C expression by promoting NF-κB-mediated activation of the MUC1 gene. Of significance, MUC1-C and XIST regulate common genes associated with inflammation and stemness, including (i) miR-21 which is upregulated across pan-cancers, and (ii) TDP-43 which associates with the XIST E repeats. Our results further demonstrate that the MUC1-C/XIST pathway (i) is regulated by TDP-43, (ii) drives stemness-associated genes, and (iii) is necessary for self-renewal capacity. These findings indicate that the MUC1-C/XIST auto-regulatory axis is of importance in cancer progression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mucina-1 , ARN Largo no Codificante , Animales , Humanos , Ratones , Línea Celular Tumoral , Progresión de la Enfermedad , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , MicroARNs/metabolismo , MicroARNs/genética , Mucina-1/metabolismo , Mucina-1/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , FN-kappa B/metabolismo , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/genética
11.
Int J Mol Med ; 54(1)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38785138

RESUMEN

Ovarian cancer (OC) is one of the most common gynecological malignancies. Currently, chemoradiotherapy is the primary clinical treatment approach for OC; however, it has severe side effects and a high rate of recurrence. Thus, there is an urgent need to develop innovative therapeutic options. Paeoniflorigenone (PFG) is a monoterpene compound isolated from the traditional Chinese medicine Paeoniae Radix Rubra. PFG can inhibit the proliferation of tumor cells; however, its anticancer activity against OC has yet to be elucidated. Mucin 1 (MUC1) is highly expressed in various malignant tumors, and is associated with tumor proliferation, metastasis and epithelial­mesenchymal transition (EMT). In addition, MUC1 affects numerous signaling pathways in tumor cells. In order to develop a possible treatment approach for metastatic OC, the antitumor activity of PFG in OC cells was investigated using Cell Counting Kit­8 assay, Edu assay, flow cytometry, Transwell assay and western blot analysis. In addition, it was assessed how PFG affects MUC1 expression and function. The experiments revealed that PFG significantly inhibited OC cell proliferation, migration, invasion and EMT. PFG also induced S­phase cell cycle arrest in OC cells. Furthermore, PFG inhibited MUC1 promoter activity, which led to a decrease in MUC1 protein expression. By contrast, MUC1 promoted OC progression, including cell proliferation, cell cycle progression and cell migration. Stable knockdown of MUC1 in OC cells improved the ability of PFG to block the Wnt/ß­catenin pathway, and to limit tumor cell invasion and migration, whereas MUC1 overexpression partially counteracted the antitumor effects of PFG. In conclusion, the present study demonstrated that PFG may inhibit the MUC1/Wnt/ß­catenin pathway to induce anti­metastatic, anti­invasive and anti­EMT effects on OC. Notably, MUC1 may be a direct target of PFG. Thus, PFG holds promise as a specific antitumor agent for the treatment of OC.


Asunto(s)
Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Mucina-1 , Neoplasias Ováricas , Vía de Señalización Wnt , Femenino , Humanos , Vía de Señalización Wnt/efectos de los fármacos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/tratamiento farmacológico , Mucina-1/metabolismo , Mucina-1/genética , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Monoterpenos/farmacología , Metástasis de la Neoplasia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
12.
J Transl Med ; 22(1): 476, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764010

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) is the leading cause of blinding eye disease among working adults and is primarily attributed to the excessive proliferation of microvessels, which leads to vitreous hemorrhage and retinal traction, thereby significantly impairing patient vision. NSUN2-mediated RNA m5C methylation is implicated in various diseases, and in this investigation, we focused on elucidating the impact of NSUN2 on the regulation of the expression of the downstream gene MUC1, specifically through RNA m5C methylation, on the progression of DR. METHOD: Utilizing Microarray analysis, we examined patient vitreous fluid to pinpoint potential therapeutic targets for DR. Differential expression of NSUN2 was validated through qRT-PCR, Western blot, and immunofluorescence in human tissue, animal tissue, and cell model of DR. The relationship between NSUN2 and DR was explored in vitro and in vivo through gene knockdown and overexpression. Various techniques, such as MeRIP-qPCR and dot blot, were applied to reveal the downstream targets and mechanism of action of NSUN2. RESULTS: The levels of both NSUN2 and RNA m5C methylation were significantly elevated in the DR model. Knockdown of NSUN2 mitigated DR lesion formation both in vitro and in vivo. Mechanistically, NSUN2 promoted MUC1 expression by binding to the RNA m5C reader ALYREF. Knockdown of ALYREF resulted in DR lesion alterations similar to those observed with NSUN2 knockdown. Moreover, MUC1 overexpression successfully reversed a series of DR alterations induced by NSUN2 silencing. CONCLUSIONS: NSUN2 regulates the expression of MUC1 through ALYREF-mediated RNA m5C methylation, thereby regulating the progression of DR and providing a new option for the treatment of DR in the future.


Asunto(s)
Retinopatía Diabética , Progresión de la Enfermedad , Metiltransferasas , Mucina-1 , Metilación de ARN , Animales , Humanos , Masculino , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Metilación , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones Endogámicos C57BL , Mucina-1/metabolismo , Mucina-1/genética
13.
Proc Natl Acad Sci U S A ; 121(20): e2322688121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38709925

RESUMEN

Brain metastatic breast cancer is particularly lethal largely due to therapeutic resistance. Almost half of the patients with metastatic HER2-positive breast cancer develop brain metastases, representing a major clinical challenge. We previously described that cancer-associated fibroblasts are an important source of resistance in primary tumors. Here, we report that breast cancer brain metastasis stromal cell interactions in 3D cocultures induce therapeutic resistance to HER2-targeting agents, particularly to the small molecule inhibitor of HER2/EGFR neratinib. We investigated the underlying mechanisms using a synthetic Notch reporter system enabling the sorting of cancer cells that directly interact with stromal cells. We identified mucins and bulky glycoprotein synthesis as top-up-regulated genes and pathways by comparing the gene expression and chromatin profiles of stroma-contact and no-contact cancer cells before and after neratinib treatment. Glycoprotein gene signatures were also enriched in human brain metastases compared to primary tumors. We confirmed increased glycocalyx surrounding cocultures by immunofluorescence and showed that mucinase treatment increased sensitivity to neratinib by enabling a more efficient inhibition of EGFR/HER2 signaling in cancer cells. Overexpression of truncated MUC1 lacking the intracellular domain as a model of increased glycocalyx-induced resistance to neratinib both in cell culture and in experimental brain metastases in immunodeficient mice. Our results highlight the importance of glycoproteins as a resistance mechanism to HER2-targeting therapies in breast cancer brain metastases.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Resistencia a Antineoplásicos , Glicocálix , Quinolinas , Receptor ErbB-2 , Células del Estroma , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Femenino , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Glicocálix/metabolismo , Animales , Línea Celular Tumoral , Células del Estroma/metabolismo , Células del Estroma/patología , Quinolinas/farmacología , Ratones , Comunicación Celular , Técnicas de Cocultivo , Mucina-1/metabolismo , Mucina-1/genética , Transducción de Señal , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inhibidores
14.
Oncogene ; 43(28): 2199-2214, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38802648

RESUMEN

The MUC1 gene evolved in mammals for adaptation of barrier tissues in response to infections and damage. Paraspeckles are nuclear bodies formed on the NEAT1 lncRNA in response to loss of homeostasis. There is no known intersection of MUC1 with NEAT1 or paraspeckles. Here, we demonstrate that the MUC1-C subunit plays an essential role in regulating NEAT1 expression. MUC1-C activates the NEAT1 gene with induction of the NEAT1_1 and NEAT1_2 isoforms by NF-κB- and MYC-mediated mechanisms. MUC1-C/MYC signaling also induces expression of the SFPQ, NONO and FUS RNA binding proteins (RBPs) that associate with NEAT1_2 and are necessary for paraspeckle formation. MUC1-C integrates activation of NEAT1 and RBP-encoding genes by recruiting the PBAF chromatin remodeling complex and increasing chromatin accessibility of their respective regulatory regions. We further demonstrate that MUC1-C and NEAT1 form an auto-inductive pathway that drives common sets of genes conferring responses to inflammation and loss of homeostasis. Of functional significance, we find that the MUC1-C/NEAT1 pathway is of importance for the cancer stem cell (CSC) state and anti-cancer drug resistance. These findings identify a previously unrecognized role for MUC1-C in the regulation of NEAT1, RBPs, and paraspeckles that has been co-opted in promoting cancer progression.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Mucina-1 , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Mucina-1/genética , Mucina-1/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo , FN-kappa B/metabolismo , FN-kappa B/genética , Factor de Empalme Asociado a PTB/genética , Factor de Empalme Asociado a PTB/metabolismo , Transducción de Señal/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ADN
15.
BMC Cancer ; 24(1): 559, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702644

RESUMEN

In contrast to the decreasing trends in developed countries, the incidence and mortality rates of cervical squamous cell carcinoma in China have increased significantly. The screening and identification of reliable biomarkers and candidate drug targets for cervical squamous cell carcinoma are urgently needed to improve the survival rate and quality of life of patients. In this study, we demonstrated that the expression of MUC1 was greater in neoplastic tissues than in non-neoplastic tissues of the cervix, and cervical squamous cell carcinoma patients with high MUC1 expression had significantly worse overall survival than did those with low MUC1 expression, indicating its potential for early diagnosis of cervical squamous cell carcinoma. Next, we explored the regulatory mechanism of MUC1 in cervical squamous cell carcinoma. MUC1 could upregulate ITGA2 and ITGA3 expression via ERK phosphorylation, promoting the proliferation and metastasis of cervical cancer cells. Further knockdown of ITGA2 and ITGA3 significantly inhibited the tumorigenesis of cervical cancer cells. Moreover, we designed a combination drug regimen comprising MUC1-siRNA and a novel ERK inhibitor in vivo and found that the combination of these drugs achieved better results in animals with xenografts than did MUC1 alone. Overall, we discovered a novel regulatory pathway, MUC1/ERK/ITGA2/3, in cervical squamous cell carcinoma that may serve as a potential biomarker and therapeutic target in the future.


MUC1 is overexpressed in cervical squamous cell carcinoma. MUC1 regulates ERK phosphorylation, and subsequently upregulates ITGA2 and ITGA3 expression to promote tumorigenesis in cervical squamous cell carcinoma. A combination drug regimen targeting MUC1 and ERK achieved better results compared than MUC1 alone.


Asunto(s)
Carcinoma de Células Escamosas , Proliferación Celular , Integrina alfa2 , Integrina alfa3 , Mucina-1 , Neoplasias del Cuello Uterino , Humanos , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Femenino , Integrina alfa2/metabolismo , Integrina alfa2/genética , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/tratamiento farmacológico , Mucina-1/metabolismo , Mucina-1/genética , Ratones , Fosforilación , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Sistema de Señalización de MAP Quinasas , Ratones Desnudos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
16.
Clin Transl Med ; 14(5): e1701, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38778448

RESUMEN

BACKGROUND: Mucinous colorectal adenocarcinoma (MCA) is a distinct subtype of colorectal cancer (CRC) with the most aggressive pattern, but effective treatment of MCA remains a challenge due to its vague pathological characteristics. An in-depth understanding of transcriptional dynamics at the cellular level is critical for developing specialised MCA treatment strategies. METHODS: We integrated single-cell RNA sequencing and spatial transcriptomics data to systematically profile the MCA tumor microenvironment (TME), particularly the interactome of stromal and immune cells. In addition, a three-dimensional bioprinting technique, canonical ex vivo co-culture system, and immunofluorescence staining were further applied to validate the cellular communication networks within the TME. RESULTS: This study identified the crucial intercellular interactions that engaged in MCA pathogenesis. We found the increased infiltration of FGF7+/THBS1+ myofibroblasts in MCA tissues with decreased expression of genes associated with leukocyte-mediated immunity and T cell activation, suggesting a crucial role of these cells in regulating the immunosuppressive TME. In addition, MS4A4A+ macrophages that exhibit M2-phenotype were enriched in the tumoral niche and high expression of MS4A4A+ was associated with poor prognosis in the cohort data. The ligand-receptor-based intercellular communication analysis revealed the tight interaction of MUC1+ malignant cells and ZEB1+ endothelial cells, providing mechanistic information for MCA angiogenesis and molecular targets for subsequent translational applications. CONCLUSIONS: Our study provides novel insights into communications among tumour cells with stromal and immune cells that are significantly enriched in the TME during MCA progression, presenting potential prognostic biomarkers and therapeutic strategies for MCA. KEY POINTS: Tumour microenvironment profiling of MCA is developed. MUC1+ tumour cells interplay with FGF7+/THBS1+ myofibroblasts to promote MCA development. MS4A4A+ macrophages exhibit M2 phenotype in MCA. ZEB1+ endotheliocytes engage in EndMT process in MCA.


Asunto(s)
Adenocarcinoma Mucinoso , Neoplasias Colorrectales , Mucina-1 , Análisis de la Célula Individual , Microambiente Tumoral , Humanos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Microambiente Tumoral/genética , Análisis de la Célula Individual/métodos , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/patología , Mucina-1/genética , Mucina-1/metabolismo , Comunicación Celular/genética
17.
Biosens Bioelectron ; 259: 116380, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38754193

RESUMEN

Exosomes, as novel biomarker for liquid biopsy, exhibit huge important potential value for cancer diagnosis. However, various proteins show different expression levels on exosomal membrane, and the absolute concentration of exosomes in clinical samples is easily influenced by a number of factors. Here, we developed a CRISPR/Cas12a and aptamer-chemiluminescence based analysis (CACBA) for the relative abundance determination of tumor-related protein positive exosomes in plasma for breast cancer diagnosis. The total concentration of exosomes was determined through captured CD63 using a CRISPR/Cas12a-based method with the LoD of 8.97 × 103 particles/µl. Meanwhile, EpCAM and MUC1 positive exosomes were quantitatively detected by aptamer-chemiluminescence (ACL) based method with the LoD of 1.45 × 102 and 3.73 × 102 particles/µl, respectively. It showed that the percentages of EpCAM and MUC1 positive exosomes offered an excellent capability to differentiate breast cancer patients and healthy donors. The high sensitivity, strong specificity, outstanding anti-interference capability, and steady recovery rate of this approach offered higher accuracy and robustness than the commercialized method in clinical trial. In addition with good stability, easy preparation and low cost, this method not only provides a new approach to rapid analysis of exosome proteins, it may be quickly extended to the diagnoses of various cancers.


Asunto(s)
Aptámeros de Nucleótidos , Biomarcadores de Tumor , Técnicas Biosensibles , Neoplasias de la Mama , Sistemas CRISPR-Cas , Molécula de Adhesión Celular Epitelial , Exosomas , Mucina-1 , Humanos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Exosomas/química , Exosomas/genética , Femenino , Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Mucina-1/sangre , Mucina-1/genética , Mucina-1/análisis , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Molécula de Adhesión Celular Epitelial/genética , Mediciones Luminiscentes/métodos , Tetraspanina 30 , Límite de Detección
18.
Mol Carcinog ; 63(7): 1303-1318, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38634741

RESUMEN

The expression pattern of MUC1-C in tumors is closely linked to tumor progression; however, its specific mechanism remains unclear. The expression of MUC1-C in cancer and adjacent normal tissues was detected using immunohistochemistry and Western blot. The IC50 of cells to gemcitabine was determined using the CCK8 assay. The effects of hypoxia and MUC1-C on the behavioral and metabolic characteristics of bladder cancer cells were investigated. Gene expression was assessed through Western blot and polymerase chain reaction. The relationship between the genes was analyzed by co-immunoprecipitation, immunofluorescence and Western blot. Finally, the role of the EGLN2 and NF-κB signaling pathways in the interaction between MUC1-C and hypoxia-inducible factor-1α (HIF-1α) was investigated. MUC1-C expression is significantly higher in bladder cancer tissues than in adjacent normal tissues, particularly in large-volume tumors, and is closely correlated with clinical features such as tumor grade. Tumor volume-mediated hypoxia resulted in increased expression of MUC1-C and HIF-1α in bladder cancer cells. Under stimulation of hypoxia, the inhibitory effect of EGLN2 on the NF-κB signaling pathway was weakened, allowing NF-κB to promote the positive feedback formation of MUC1-C and HIF-1α. Simultaneously, EGLN2-mediated degradation of HIF-1α was reduced. This ultimately led to elevated HIF-1α-mediated downstream gene expression, promoting increased glucose uptake and glycolysis, and ultimately resulting in heightened chemotherapy resistance and malignancy.


Asunto(s)
Resistencia a Antineoplásicos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Mucina-1 , Transducción de Señal , Neoplasias de la Vejiga Urinaria , Femenino , Humanos , Masculino , Persona de Mediana Edad , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Gemcitabina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Mucina-1/metabolismo , Mucina-1/genética , FN-kappa B/metabolismo , FN-kappa B/genética , Transducción de Señal/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/genética
19.
Cancer Res Commun ; 4(5): 1268-1281, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38619287

RESUMEN

The MUC1-C protein is aberrantly expressed in adenocarcinomas of epithelial barrier tissues and contributes to their progression. Less is known about involvement of MUC1-C in the pathogenesis of squamous cell carcinomas (SCC). Here, we report that the MUC1 gene is upregulated in advanced head and neck SCCs (HNSCC). Studies of HNSCC cell lines demonstrate that the MUC1-C subunit regulates expression of (i) RIG-I and MDA5 pattern recognition receptors, (ii) STAT1 and IFN regulatory factors, and (iii) downstream IFN-stimulated genes. MUC1-C integrates chronic activation of the STAT1 inflammatory pathway with induction of the ∆Np63 and SOX2 genes that are aberrantly expressed in HNSCCs. In extending those dependencies, we demonstrate that MUC1-C is necessary for NOTCH3 expression, self-renewal capacity, and tumorigenicity. The findings that MUC1 associates with ∆Np63, SOX2 and NOTCH3 expression by single-cell RNA sequencing analysis further indicate that MUC1-C drives the HNSCC stem cell state and is a target for suppressing HNSCC progression. SIGNIFICANCE: This work reports a previously unrecognized role for MUC1-C in driving STAT1-mediated chronic inflammation with the progression of HNSCC and identifies MUC1-C as a druggable target for advanced HNSCC treatment.


Asunto(s)
Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello , Mucina-1 , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Mucina-1/genética , Mucina-1/metabolismo , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/metabolismo , Línea Celular Tumoral , Ratones , Animales , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Receptor Notch3/genética , Receptor Notch3/metabolismo
20.
Scand J Immunol ; 99(5): e13356, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605549

RESUMEN

In light of increasing resistance to PD1 antibody therapy among certain patient populations, there is a critical need for in-depth research. Our study assesses the synergistic effects of a MUC1 DNA vaccine and PD1 antibody for surmounting PD1 resistance, employing a murine CT26/MUC1 colon carcinoma model for this purpose. When given as a standalone treatment, PD1 antibodies showed no impact on tumour growth. Additionally, there was no change observed in the intra-tumoural T-cell ratios or in the functionality of T-cells. In contrast, the sole administration of a MUC1 DNA vaccine markedly boosted the cytotoxicity of CD8+ T cells by elevating IFN-γ and granzyme B production. Our compelling evidence highlights that combination therapy more effectively inhibited tumour growth and prolonged survival compared to either monotherapy, thus mitigating the limitations intrinsic to single-agent therapies. This enhanced efficacy was driven by a significant alteration in the tumour microenvironment, skewing it towards pro-immunogenic conditions. This assertion is backed by a raised CD8+/CD4+ T-cell ratio and a decrease in immunosuppressive MDSC and Treg cell populations. On the mechanistic front, the synergistic therapy amplified expression levels of CXCL13 in tumours, subsequently facilitating T-cell ingress into the tumour setting. In summary, our findings advocate for integrated therapy as a potent mechanism for surmounting PD1 antibody resistance, capitalizing on improved T-cell functionality and infiltration. This investigation affords critical perspectives on enhancing anti-tumour immunity through the application of innovative therapeutic strategies.


Asunto(s)
Anticuerpos , Mucina-1 , Neoplasias , Receptor de Muerte Celular Programada 1 , Vacunas de ADN , Animales , Ratones , Anticuerpos/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Mucina-1/genética , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...