Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.759
Filtrar
1.
Mol Med Rep ; 30(6)2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-39370807

RESUMEN

Although both mucin1 (MUC1) and transient receptor potential cation channel subfamily V member 1 (TRPV1) have been reported to be associated with dry eye (DE) disease, whether they interact and their regulatory roles in diabetic DE disease are unknown. Diabetic DE model mice were generated by streptozotocin induction and assessed by corneal fluorescein staining, tear ferning (TF) tests, phenol red thread tests, hematoxylin and eosin staining of corneal sections and periodic acid Schiff staining of conjunctival sections. Cell proliferation was measured by CCK8 assay. Western blotting was performed to measure protein expression. Primary mouse corneal epithelial cells (MCECs) were cultured after enzymatic digestion. Immunofluorescence staining of MCECs and frozen corneal sections was conducted to assess protein expression and colocalization. Coimmunoprecipitation was performed to detect protein­protein interactions. It was found that, compared with control mice, diabetic DE mice exhibited increased corneal epithelial defects, reduced tear production, poorer TF pattern grades and impaired corneal and conjunctival tissues. In vivo and in vitro experiments showed that hyperglycemia impaired cell proliferation, accompanied by decreased levels of the MUC1 extracellular domain (MUC1­ND) and TRPV1. Additionally, it was found that capsazepine (a TRPV1 antagonist) inhibited the proliferation of MCECs. Notably, MUC1­ND was shown to interact with the TRPV1 protein in the control group but not in the diabetic DE group. It was also found that the AKT signaling pathway was attenuated in the diabetic DE mice and downstream of TRPV1. MUC1­ND interacted with TRPV1, partly activating the AKT signaling pathway to promote MCEC proliferation. The present study found that the interaction of MUC1­ND with TRPV1 promotes MCEC proliferation by partly activating the AKT signaling pathway, providing new insight into the pathogenesis of corneal epithelial dysfunction in diabetic DE disease.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Experimental , Síndromes de Ojo Seco , Mucina-1 , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Canales Catiónicos TRPV , Animales , Canales Catiónicos TRPV/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones , Mucina-1/metabolismo , Síndromes de Ojo Seco/metabolismo , Síndromes de Ojo Seco/patología , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
3.
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
4.
Chem Commun (Camb) ; 60(78): 10938-10941, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39258452

RESUMEN

Here, we present a biosynthesized material M1 for immune checkpoint blocking therapy. M1 could realize a morphological transformation from globular to fibrous in situ in the presence of cathepsin B (CtsB) after entering tumor cells. The GO203 peptides of M1 are exposed, which could bind to mucin 1 (MUC1) to suppress the homodimerization process of MUC1, thereby downregulating PD-L1 expression.


Asunto(s)
Antígeno B7-H1 , Catepsina B , Regulación hacia Abajo , Péptidos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/química , Humanos , Péptidos/química , Péptidos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Catepsina B/metabolismo , Mucina-1/metabolismo , Mucina-1/química , Línea Celular Tumoral
5.
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
6.
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
7.
Cancer Immunol Immunother ; 73(10): 203, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39105847

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR)-T cells have been used to treat blood cancers by producing a wide variety of cytokines. However, they are not effective in treating solid cancers and can cause severe side-effects, including cytokine release syndrome. TNFα is a tumoricidal cytokine, but it markedly increases the protein levels of cIAP1 and cIAP2, the members of inhibitor of apoptosis protein (IAP) family of E3 ubiquitin ligase that limits caspase-induced apoptosis. Degradation of IAP proteins by an IAP antagonist does not effectively kill cancer cells but enables TNFα to strongly induce cancer cell apoptosis. It would be a promising approach to treat cancers by targeted delivery of TNFα through an inactive adoptive cell in combination with an IAP antagonist. METHODS: Human dendritic cells (DCs) were engineered to express a single tumoricidal factor, TNFα, and a membrane-anchored Mucin1 antibody scFv, named Mucin 1 directed DCs expressing TNFα (M-DCsTNF). The efficacy of M-DCsTNF in recognizing and treating breast cancer was tested in vitro and in vivo. RESULTS: Mucin1 was highly expressed on the surface of a wide range of human breast cancer cell lines. M-DCsTNF directly associated with MDA-MB-231 cells in the bone of NSG mice. M-DCsTNF plus an IAP antagonist, SM-164, but neither alone, markedly induce MDA-MB-231 breast cancer cell apoptosis, which was blocked by TNF antibody. Importantly, M-DCsTNF combined with SM-164, but not SM-164 alone, inhibited the growth of patient-derived breast cancer in NSG mice. CONCLUSION: An adoptive cell targeting delivery of TNFα combined with an IAP antagonist is a novel effective approach to treat breast cancer and could be expanded to treat other solid cancers. Unlike CAR-T cell, this novel adoptive cell is not activated to produce a wide variety of cytokines, except for additional overexpressed TNF, and thus could avoid the severe side effects such as cytokine release syndrome.


Asunto(s)
Células Dendríticas , Receptores Quiméricos de Antígenos , Factor de Necrosis Tumoral alfa , Humanos , Animales , Ratones , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Receptores Quiméricos de Antígenos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Mucina-1/inmunología , Mucina-1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inmunoterapia Adoptiva/métodos , Apoptosis , Neoplasias de la Mama/terapia , Neoplasias de la Mama/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Neoplasias/inmunología , Ratones SCID
8.
Nanomedicine (Lond) ; 19(24): 1957-1975, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39136402

RESUMEN

Aim: To ascertain the impact of shape and surface modification of anisotropic nanoparticles on the toxicity and photothermal efficiency toward cancerous cell lines.Methods: Gold nanobipyramids and nanostars surface modified with MUC1 aptamer were used in the current study to explore the toxicity and photothermal efficiency on MCF7 breast cancer cell lines via MTT assay.Results: Surface functionalization with MUC1 aptamer showed significant reduction in % cytotoxicity and increase in % specific internalization of nanostructures into MCF7 cell lines. Further, the photothermal studies accomplished at IC50 concentration for 6 h of treatment and laser exposure for 15 min reported that aptamer-conjugated nanobipyramids were more effective and specific toward MCF7 cell lines than aptamer-conjugated nanostars.Conclusion: This work establishes a platform for the development of tailored photoablation based gold nanostructures for in vivo studies.


[Box: see text].


Asunto(s)
Aptámeros de Nucleótidos , Oro , Nanopartículas del Metal , Mucina-1 , Humanos , Oro/química , Células MCF-7 , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Mucina-1/metabolismo , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Terapia Fototérmica/métodos , Neoplasias de la Mama/terapia , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Supervivencia Celular/efectos de los fármacos
9.
Cancer Med ; 13(15): e70079, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39118454

RESUMEN

BACKGROUND: Cancer remains a formidable global health challenge, currently affecting nearly 20 million individuals worldwide. Due to the absence of universally effective treatments, ongoing research explores diverse strategies to combat this disease. Recent efforts have concentrated on developing combined drug regimens and targeted therapeutic approaches. OBJECTIVE: This study aimed to investigate the anticancer efficacy of a conjugated drug system, consisting of doxorubicin and cisplatin (Dox-Cis), encapsulated within niosomes and modified with MUC-1 aptamers to enhance biocompatibility and target specific cancer cells. METHODS: The chemical structure of the Dox-Cis conjugate was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-TOF/MS). The zeta potential and morphological parameters of the niosomal vesicles were determined through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). In vitro assessments of cell viability and apoptosis were conducted on MUC-1 positive HeLa cells and MUC-1 negative U87 cells. RESULTS: The findings confirmed the successful conjugation of Dox and Cis within the niosomes. The Nio/Dox-Cis/MUC-1 formulation demonstrated enhanced efficacy compared to the individual drugs and their unencapsulated combination in both cell lines. Notably, the Nio/Dox-Cis/MUC-1 formulation exhibited greater effectiveness on HeLa cells (38.503 ± 1.407) than on U87 cells (46.653 ± 1.297). CONCLUSION: The study underscores the potential of the Dox-Cis conjugate as a promising strategy for cancer treatment, particularly through platforms that facilitate targeted drug delivery to cancer cells. This targeted approach could lead to more effective and personalized cancer therapies.


Asunto(s)
Aptámeros de Nucleótidos , Supervivencia Celular , Cisplatino , Doxorrubicina , Liposomas , Mucina-1 , Humanos , Doxorrubicina/farmacología , Doxorrubicina/química , Mucina-1/metabolismo , Mucina-1/química , Liposomas/química , Cisplatino/farmacología , Cisplatino/química , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Supervivencia Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células HeLa , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Composición de Medicamentos/métodos
10.
J Virol ; 98(7): e0049924, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38953631

RESUMEN

Tibroviruses are novel rhabdoviruses detected in humans, cattle, and arthropods. Four tibroviruses are known to infect humans: Bas-Congo virus (BASV), Ekpoma virus 1 (EKV-1), Ekpoma virus 2, and Mundri virus. However, since none of them has been isolated, their biological properties are largely unknown. We aimed to characterize the human tibrovirus glycoprotein (G), which likely plays a pivotal role in viral tropism and pathogenicity. Human tibrovirus Gs were found to share some primary structures and display 14 conserved cysteine residues, although their overall amino acid homology was low (29%-48%). Multiple potential glycosylation sites were found on the G molecules, and endoglycosidase H- and peptide-N-glycosidase F-sensitive glycosylation was confirmed. AlphaFold-predicted three-dimensional (3D) structures of human tibrovirus Gs were overall similar. Membrane fusion mediated by these tibrovirus Gs was induced by acidic pH. The low pH-induced conformational change that triggers fusion was reversible. Virus-like particles (VLPs) were produced by transient expression of Gs in cultured cells and used to produce mouse antisera. Using vesicular stomatitis Indiana virus pseudotyped with Gs, we found that the antisera to the respective tibrovirus VLPs showed limited cross-neutralizing activity. It was also found that human C-type lectins and T-cell immunoglobulin mucin 1 acted as attachment factors for G-mediated entry into cells. Interestingly, BASV-G showed the highest ability to utilize these molecules. The viruses infected a wide range of cell lines with preferential tropism for human-derived cells whereas the preference of EKV-1 was unique compared with the other human tibroviruses. These findings provide fundamental information to understand the biological properties of the human tibroviruses. IMPORTANCE: Human tibroviruses are poorly characterized emerging rhabdoviruses associated with either asymptomatic infection or severe disease with a case fatality rate as high as 60% in humans. However, the extent and burden of human infection as well as factors behind differences in infection outcomes are largely unknown. In this study, we characterized human tibrovirus glycoproteins, which play a key role in virus-host interactions, mainly focusing on their structural and antigenic differences and cellular tropism. Our results provide critical information for understanding the biological properties of these novel viruses and for developing appropriate preparedness interventions such as diagnostic tools, vaccines, and effective therapies.


Asunto(s)
Proteínas del Envoltorio Viral , Humanos , Animales , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Ratones , Glicosilación , Internalización del Virus , Tropismo Viral , Línea Celular , Mucina-1/metabolismo , Células HEK293 , Anticuerpos Antivirales/inmunología , Secuencia de Aminoácidos
11.
Anal Chem ; 96(31): 12767-12775, 2024 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-39044392

RESUMEN

Bacteria inherently possess the capability of quorum sensing in response to the environment. In this work, we have proposed a strategy to confer bacteria with the ability to recognize targets with quorum-sensing behavior. Meanwhile, we have successfully achieved artificial control over the target-triggered aggregation of Escherichia coli (E. coli) by modifying the bacteria surface in a new way. Furthermore, by making use of green fluorescent protein (GFP) expressed by E. coli as the output signal, the aggregation of modified E. coli can be observed with the naked eye. Therefore, via the detection of the target, MUC1, an ovarian cancer biomarker, a simple and conveniently operated method to diagnose ovarian cancer is developed in this work. Experimental results show that the developed low-background and enzyme-free amplification method enables the highly sensitive detection of MUC1, achieving a remarkable limit of detection (LOD) of 5.47 fM and a linear detection range spanning from 1 pM to 50 nM and 50 nM to 100 nM, respectively. Clinical samples from healthy donors and patients can give distant assay results, showing great potential for clinical applications of this method.


Asunto(s)
Escherichia coli , Mucina-1 , Neoplasias Ováricas , Neoplasias Ováricas/diagnóstico , Escherichia coli/aislamiento & purificación , Humanos , Femenino , Mucina-1/análisis , Mucina-1/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/genética , Límite de Detección , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Percepción de Quorum , Técnicas Biosensibles/métodos
12.
Dermatol Online J ; 30(2)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38959917

RESUMEN

Squamoid eccrine ductal carcinoma (SEDC) is a cutaneous adnexal malignancy that is histologically challenging to distinguish from squamous cell carcinoma. We report three cases of this rare entity and review the present literature regarding clinical, histological, and immunohistochemical features. Patients presented with a single nodule or plaque lesion on their back and temple. The shave biopsies for Patient A and C were interpreted as SEDC. Patient B's initial shave biopsy was interpreted as probable surface of squamous cell carcinoma, and subsequent excision revealed SEDC. Ductal differentiation was confirmed by positive expression of epithelial membrane antigen and carcinoembryonic antigen immunostains in all three patients. Review of the 67 previously reported cases emphasizes the importance of diagnosing SEDC accurately and promptly given its potential for distant metastasis and mortality. Perineural or lymphatic invasion is associated with higher rate of recurrence or metastasis. There should be high pathologic suspicion for SEDC in an elderly patient presenting with a palpable lesion, even if located outside of the head and neck area, particularly when there is suggestion of ductal differentiation in a sample of a squamous neoplasm.


Asunto(s)
Carcinoma de Células Escamosas , Glándulas Ecrinas , Neoplasias de las Glándulas Sudoríparas , Humanos , Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/metabolismo , Carcinoma Ductal/patología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/diagnóstico , Diagnóstico Diferencial , Glándulas Ecrinas/patología , Inmunohistoquímica , Mucina-1/análisis , Mucina-1/metabolismo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/diagnóstico , Neoplasias de las Glándulas Sudoríparas/patología , Anciano
13.
Biochem Biophys Res Commun ; 731: 150402, 2024 Oct 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
14.
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
15.
J Control Release ; 372: 417-432, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908758

RESUMEN

This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.


Asunto(s)
Pulmón , Nanopartículas , SARS-CoV-2 , Nanopartículas/administración & dosificación , Humanos , Pulmón/metabolismo , SARS-CoV-2/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Inmunomodulación , Línea Celular , Mucina-1/metabolismo , COVID-19 , Lípidos/química , Lípidos/administración & dosificación , Moco/metabolismo , Polietilenglicoles/química , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Mucina 5AC/metabolismo , Liposomas
16.
Analyst ; 149(15): 3920-3927, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38912896

RESUMEN

Anti-cancer therapy is crucial in cancer prevention and anti-cancer, and thus, highly sensitive methods for detecting cancer biomarkers are essential for cancer early diagnosis. Herein, an electrochemical aptamer biosensor based on the CRISPR-Cas12a system was constructed for the detection of cancer tumor biomarker MUC1 mucin. The sensitivity was significantly prompted by enzyme-catalyzed signal amplification, and the selectivity was improved by the dual recognition of the aptamer to MUC1 and crRNA-Cas12a system to the aptamer. Glucose oxidase (GOD) was loaded on the surface of magnetic Fe3O4@Au (MGNP) via probe single-stranded DNA (pDNA) with the terminal modification of mercapto (-SH) to form GOD-pDNA/MGNP. The corresponding aptamer of MUC1 (MUC1 Apt) binds to its complementary ssDNA (cDNA) to form the activator Apt/cDNA, which is specifically recognized by crRNA-Cas12a and excites the trans-cleavage function of Cas12a, thus in turn trans-cleaves pDNA and detaches GOD from the magnetic particles. The magnetic beads were separated and transferred into a glucose solution, and the oxidation current of H2O2 produced by the catalytic reaction of GOD was measured on a Pt-modified magnetically-controlled glassy carbon electrode, resulting in an indirect determination of MUC1. The current change was linear with the logarithm of MUC1 concentration in the range from 1.0 × 10-17 g mL-1 to 1.0 × 10-10 g mL-1. The detection limit was as low as 7.01 × 10-18 g mL-1. The method was applied for the detection of MUC1 in medical samples.


Asunto(s)
Aptámeros de Nucleótidos , Biomarcadores de Tumor , Técnicas Biosensibles , Técnicas Electroquímicas , Glucosa Oxidasa , Mucina-1 , Humanos , Aptámeros de Nucleótidos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biomarcadores de Tumor/genética , Técnicas Biosensibles/métodos , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , ADN de Cadena Simple/química , Técnicas Electroquímicas/métodos , Endodesoxirribonucleasas , Enzimas Inmovilizadas/química , Glucosa Oxidasa/química , Glucosa Oxidasa/metabolismo , Oro/química , Peróxido de Hidrógeno/química , Límite de Detección , Nanopartículas de Magnetita/química , Mucina-1/metabolismo
17.
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
18.
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
19.
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
20.
J Mater Chem B ; 12(22): 5551-5560, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38747235

RESUMEN

Timely identification of cancers is pivotal in optimizing treatment efficacy and reducing their widespread impact. This study introduces a novel biosensor for the sensitive electrochemical detection of cancer cells overexpressing mucin 1 (MUC1), a well-established model for breast cancer. The sensor substrate comprises gold columnar nanostructures obtained through glancing angle deposition (GLAD) of copper nanostructures, subsequently replaced by gold via a facile galvanic replacement process. Functionalizing these gold nanostructures with aptamers targeting the MUC1 glycoproteins, a prominent cancer biomarker, enables specific recognition of MCF-7 breast cancer cells. The proposed electrochemical sensing platform offers several advantages, including high selectivity, a wide linear range of detection, a low detection limit of 30 cells per mL, and long-term stability, rendering this sensor highly desirable for definitive breast cancer diagnosis.


Asunto(s)
Técnicas Biosensibles , Neoplasias de la Mama , Técnicas Electroquímicas , Oro , Mucina-1 , Humanos , Técnicas Biosensibles/métodos , Oro/química , Células MCF-7 , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Mucina-1/análisis , Mucina-1/metabolismo , Femenino , Nanoestructuras/química , Nanopartículas del Metal/química , Aptámeros de Nucleótidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA