Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.362
Filtrar
Más filtros











Intervalo de año de publicación
1.
Methods Mol Biol ; 2823: 27-46, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39052212

RESUMEN

With the advent of promising lung cancer immunotherapies targeting proteins at the cell surface of RAS-driven human cancers, the mass spectrometry (MS)-based surfaceomics remains a feasible strategy for therapeutic target discovery. This chapter describes a protocol for discovery of druggable protein targets at the surface of RAS-driven human cancer cells. This method relies on bottom-up MS-based quantitative surfaceomics that employs in parallel, targeted hydrazide-based cell-surface glycoproteomics and global shotgun membrane proteomics to enable unbiased quantitative profiling of thousands of cell surface membrane proteins. A large-scale molecular map of the KRASG12V surface was attained, resulting in confident detection and quantitation of more than 500 cell surface membrane proteins that were found to be unique or upregulated at the surface of cells harboring the KRASG12V mutant. A multistep bioinformatic progression revealed a subset of unique and/or significantly upregulated proteins as priority drug targets selected for orthogonal cross-validation using immunofluorescence, structured illumination microscopy, and western blotting. Among cross-validated targets, CUB domain containing protein 1 (CDCP1) and basigin (BSG-CD147) were selected as leading targets due to their involvement in cell adhesion and migration, consistent with the KRASG12V malignant phenotype as revealed by scanning electron microscopy and phenotypic cancer cell assays. Follow-up studies confirmed CDCP1 as an actionable therapeutic target, resulting in development of recombinant antibodies capable of killing KRAS-transformed cancer cells in preclinical setting. The present MS-based surfaceomics workflow represents a powerful drug target discovery platform that enables development of innovative immunotherapeutics (e.g., antibody drug conjugate against CDCP1) for attacking oncogenic RAS-driven cancers at the cell surface.


Asunto(s)
Proteómica , Humanos , Proteómica/métodos , Línea Celular Tumoral , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Basigina/metabolismo , Basigina/genética , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/genética , Antígenos CD/metabolismo , Antígenos CD/genética , Membrana Celular/metabolismo , Descubrimiento de Drogas/métodos , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/genética , Espectrometría de Masas/métodos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas ras/metabolismo , Proteínas ras/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Antineoplásicos/farmacología
2.
J Immunother Cancer ; 12(7)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043605

RESUMEN

BACKGROUND: Anti-programmed death-1 (PD-1)/anti-PD-ligand-1 (PD-L1) pathway inhibition is a standard regimen for advanced urothelial carcinoma (UC); however, its limited efficacy has been reflected in reported medium response rates. This study explored the role of next-generation coinhibitory receptors (IRs; lymphocyte activation gene 3 (LAG-3), T-cell immunoglobulin and mucin domain 3 (TIM-3), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT)) and their ligands (LGs) in the response to PD-(L)1 blockade therapy and the oncological outcomes in patients with UC. METHODS: We investigated metastatic UC cases who underwent PD-(L)1 therapy (cohort 1: n=348, cohort 2: n=89, and cohort 4: n=29) or advanced UC cases involving surgery (cohort 3: n=293 and cohort 5: n=90). We assessed the mRNA expression profiles and corresponding clinical information regarding IRs and LGs using cohorts 1, 2, and 3. Additionally, we elucidated the spatial features of these targeted markers using multiplex immunohistochemistry (mIHC) on formalin-fixed paraffin-embedded samples from cohorts 4 and 5. Survival, differential expressed gene, and Gene Set Enrichment analyses were performed. For mIHC, quantitative analyses were also performed to correlate immune and tumor cell densities with patient survival. RESULTS: LAG-3 expression was strongly associated with the responsiveness of PD-(L)1 blockade compared with the expression of TIM-3 and TIGIT. In tumors with high LAG-3 levels, the increased expression of fibrinogen-like protein 1 (FGL1) had a significantly negative effect on the response to PD-(L)1 blockade and overall survival. Moreover, high FGL1 levels were associated with elevated CD4+ regulatory T-cell gene signatures and the upregulation of CD39 and neuropilin-1, with both indicating CD8+ T-cell exhaustion. mIHC analyses revealed that patients with stromal CD8+LAG-3+cellshigh-tumor FGL1+cellshigh exhibited a significant negative correlation with survival rates compared with those with stromal CD8+LAG-3+cellshigh-tumor FGL1+cellslow. CONCLUSIONS: LAG-3 expression and high FGL1 coexpression are important predictive factors of adverse oncological outcomes related to the presence of immunosuppressive contextures. These findings are hypothesis-generating, warranting further mechanistic and clinical studies aimed to evaluate LAG-3/FGL1 blockade in UC.


Asunto(s)
Antígenos CD , Proteína del Gen 3 de Activación de Linfocitos , Humanos , Masculino , Femenino , Antígenos CD/metabolismo , Antígenos CD/genética , Anciano , Persona de Mediana Edad , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/inmunología , Neoplasias Urológicas/genética , Neoplasias Urológicas/patología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
3.
Mol Genet Genomic Med ; 12(7): e2496, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39056403

RESUMEN

BACKGROUND: Hereditary diffuse gastric cancer (HDGC) (OMIM# 137215) is an autosomal dominant cancer syndrome associated with CDH1 (OMIM# 192090) mutations. Prophylactic total gastrectomy (PTG) is the most recommended preventive treatment when a pathogenic mutation is found. However, the increasing use of genetic testing has led to the identification of incidental CDH1 mutations in individuals without a family history of gastric cancer. It remains unclear whether these patients should undergo prophylactic total gastrectomy. METHODS: Germline DNA, obtained from peripheral blood, was analysed by NGS. RESULTS: A 47-year-old woman was diagnosed with high-grade serous ovarian carcinoma, FIGO stage IIIC, with a Homologous Recombination Deficiency (HRD) GIS status of 78 (positive, cut-off: 43). She received chemotherapy and niraparib treatment. A multigene panel test revealed no pathogenic mutations in BRCA1 (OMIM# 113705)/BRCA2 (OMIM# 600185) genes, but a de novo deletion of exon 16 in CDH1 was found incidentally. She had no previous family history of gastric or breast cancer. The patient was enrolled in a surveillance program involving periodic endoscopy and was diagnosed with diffuse gastric cancer through biopsies of a pale area in the antrum after 1 year of close endoscopic follow-up. CONCLUSION: This case presents supportive evidence for the pathogenic classification of the loss of the last exon of CDH1.


Asunto(s)
Antígenos CD , Cadherinas , Neoplasias Ováricas , Neoplasias Gástricas , Humanos , Femenino , Persona de Mediana Edad , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Antígenos CD/genética , Cadherinas/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Gastrectomía , Mutación de Línea Germinal
4.
BMC Geriatr ; 24(1): 603, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009979

RESUMEN

BACKGROUND: High-grade endometrial stromal sarcoma (HG-ESS) is a rare malignant tumor with poor prognosis. To overcome the limitations of current treatment for advanced patients, the intervention of targeted drug therapy is urgently needed. CASE PRESENTATION: A 74-year-old married woman who presented with abdominal distension and lower abdominal pain was admitted to Hebei General Hospital. After surgery, immunohistochemical staining revealed a malignant tumor which was consistent with HG-ESS. Tumor recurrence occurred 2 months after surgery. Then the patient underwent chemotherapy with two courses but responded poorly. Subsequently we observed ATM, BLM, and CDH1 co-mutations by Next Generation Sequencing (NGS). Then the patient received pamiparib, which resulted in a 10-month progression-free survival (PFS) and is now stable with the administration of sintilimab in combination with pamiparib and anlotinib. CONCLUSIONS: Due to the successful use of poly ADP-ribose polymerase inhibitor (PARPi) on HG-ESS, we suggest that the selection of effective targeted drugs combined with anti- programmed death-1 (PD-1) drug therapy based on genetic testing may become a new option for the treatment of homologous repair deficient (HR-deficient) HG-ESS.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Cadherinas , Neoplasias Endometriales , RecQ Helicasas , Sarcoma Estromático Endometrial , Humanos , Femenino , Anciano , Neoplasias Endometriales/genética , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/patología , Sarcoma Estromático Endometrial/genética , Sarcoma Estromático Endometrial/tratamiento farmacológico , Sarcoma Estromático Endometrial/diagnóstico , Proteínas de la Ataxia Telangiectasia Mutada/genética , RecQ Helicasas/genética , Cadherinas/genética , Antígenos CD/genética , Mutación
5.
Int J Mol Sci ; 25(13)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-39000377

RESUMEN

A macrophage shift from the M1 to the M2 phenotype is relevant for promoting tissue repair and regeneration. In a previous in vivo study, we found that direct current (DC) electrical stimulation (EStim) increased the proportion of M2 macrophages in healing tissues and directed the balance of the injury response away from healing/scarring towards regeneration. These observations led us to hypothesize that DC EStim regulates macrophage polarization towards an M2 phenotype. THP-1-derived M0, M1 (IFN-γ and LPS), and M2 (IL-4 and IL-13) macrophages were exposed (or not: control group) to 100 mV/mm of DC EStim, 1 h/day for three days. Macrophage polarization was assessed through gene and surface marker expressions and cytokine secretion profiles. Following DC EStim treatment, M0 cells exhibited an upregulation of M2 marker genes IL10, CD163, and PPARG. In M1 cells, DC EStim upregulated the gene expressions of M2 markers IL10, TGM2, and CD206 and downregulated M1 marker gene CD86. EStim treatment also reduced the surface expression of CD86 and secretion of pro-inflammatory cytokines IL-1ß and IL-6. Our results suggest that DC EStim differentially exerts pro-M2 effects depending on the macrophage phenotype: it upregulates typical M2 genes in M0 and M1 cells while inhibiting M1 marker CD86 at the nuclear and protein levels and the secretion of pro-inflammatory interleukins in M1 cells. Conversely, M2 cells appear to be less responsive to the EStim treatment employed in this study.


Asunto(s)
Estimulación Eléctrica , Macrófagos , Fenotipo , Humanos , Macrófagos/metabolismo , Estimulación Eléctrica/métodos , Células THP-1 , Activación de Macrófagos , Citocinas/metabolismo , Polaridad Celular , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/genética , Interleucina-4/metabolismo , Antígenos CD/metabolismo , Antígenos CD/genética
6.
Int J Mol Sci ; 25(13)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38999968

RESUMEN

Renal cell carcinoma (RCC) accounts for approximately 90-95% of all kidney cancers in adults, with clear cell RCC (ccRCC) being the most frequently identified subtype. RCC is known for its responsiveness to immunotherapy, making it an area of significant research interest. Immune checkpoint (IC) molecules, which regulate immune surveillance, are established therapeutic targets in RCC. The aim of this study was to analyze the influence of HVEM and CD160 gene polymorphisms on ccRCC susceptibility and patient overall survival (OS) over a ten-year period of observation. We genotyped three HVEM single nucleotide polymorphisms (SNPs): rs1886730, rs2234167, and rs8725, as well as two CD160 SNPs: rs744877 and rs2231375, in 238 ccRCC patients and 521 controls. Our findings indicated that heterozygosity within rs2231375 and/or rs2234167 increases ccRCC risk. Furthermore, in women, heterozygosity within HVEM SNPs rs8725 and rs1886730 is also associated with an increased ccRCC risk. The presence of a minor allele for rs1886730, rs2234167, rs8725, and rs2231375 was also correlated with certain clinical features of ccRCC. Moreover, rs1886730 was found to be associated with OS. In conclusion, our study highlights an association between HVEM and CD160 polymorphisms and the risk of developing ccRCC as well as OS.


Asunto(s)
Antígenos CD , Carcinoma de Células Renales , Proteínas Ligadas a GPI , Predisposición Genética a la Enfermedad , Neoplasias Renales , Polimorfismo de Nucleótido Simple , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Humanos , Femenino , Masculino , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Persona de Mediana Edad , Antígenos CD/genética , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Anciano , Proteínas Ligadas a GPI/genética , Receptores Inmunológicos/genética , Adulto , Estudios de Casos y Controles , Genotipo
7.
Nat Commun ; 15(1): 5748, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982041

RESUMEN

Autoimmune thyroid disease (AITD) is a common autoimmune disease. In a GWAS meta-analysis of 110,945 cases and 1,084,290 controls, 290 sequence variants at 225 loci are associated with AITD. Of these variants, 115 are previously unreported. Multiomics analysis yields 235 candidate genes outside the MHC-region and the findings highlight the importance of genes involved in T-cell regulation. A rare 5'-UTR variant (rs781745126-T, MAF = 0.13% in Iceland) in LAG3 has the largest effect (OR = 3.42, P = 2.2 × 10-16) and generates a novel start codon for an open reading frame upstream of the canonical protein translation initiation site. rs781745126-T reduces mRNA and surface expression of the inhibitory immune checkpoint LAG-3 co-receptor on activated lymphocyte subsets and halves LAG-3 levels in plasma among heterozygotes. All three homozygous carriers of rs781745126-T have AITD, of whom one also has two other T-cell mediated diseases, that is vitiligo and type 1 diabetes. rs781745126-T associates nominally with vitiligo (OR = 5.1, P = 6.5 × 10-3) but not with type 1 diabetes. Thus, the effect of rs781745126-T is akin to drugs that inhibit LAG-3, which unleash immune responses and can have thyroid dysfunction and vitiligo as adverse events. This illustrates how a multiomics approach can reveal potential drug targets and safety concerns.


Asunto(s)
Antígenos CD , Codón Iniciador , Predisposición Genética a la Enfermedad , Proteína del Gen 3 de Activación de Linfocitos , Humanos , Codón Iniciador/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Polimorfismo de Nucleótido Simple , Vitíligo/genética , Masculino , Estudio de Asociación del Genoma Completo , Tiroiditis Autoinmune/genética , Regiones no Traducidas 5'/genética , Estudios de Casos y Controles , Islandia , Adulto
8.
Eur J Med Res ; 29(1): 357, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970071

RESUMEN

BACKGROUND AND PURPOSE: PD-1/PD-L1 inhibitors have become a promising therapy. However, the response rate is lower than 30% in patients with cervical cancer (CC), which is related to immunosuppressive components in tumor microenvironment (TME). Tumor-associated macrophages (TAMs), as one of the most important immune cells, are involved in the formation of tumor suppressive microenvironment. Therefore, it will provide a theoretical basis for curative effect improvement about the regulatory mechanism of TAMs on PD-L1 expression. METHODS: The clinical data and pathological tissues of CC patients were collected, and the expressions of PD-L1, CD68 and CD163 were detected by immunohistochemistry. Bioinformatics was used to analyze the macrophage subtypes involved in PD-L1 regulation. A co-culture model was established to observe the effects of TAMs on the morphology, migration and invasion function of CC cells, and the regulatory mechanism of TAMs on PD-L1. RESULTS: PD-L1 expression on tumor cells could predict the poor prognosis of patients. And there was a strong correlation between PD-L1 expression with CD163+TAMs infiltration. Similarly, PD-L1 expression was associated with M1/M2-type TAMs infiltration in bioinformatics analysis. The results of cell co-culture showed that M1/M2-type TAMs could upregulate PD-L1 expression, especially M2-type TAMs may elevate the PD-L1 expression via PI3K/AKT pathway. Meanwhile, M1/M2-type TAMs can affect the morphological changes, and enhance migration and invasion abilities of CC cells. CONCLUSIONS: PD-L1 expression in tumor cells can be used as a prognostic factor and is closely related to CD163+TAMs infiltration. In addition, M2-type TAMs can upregulate PD-L1 expression in CC cells through PI3K/AKT pathway, enhance the migration and invasion capabilities, and affect the tumor progression.


Asunto(s)
Antígeno B7-H1 , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Macrófagos Asociados a Tumores , Neoplasias del Cuello Uterino , Humanos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/inmunología , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/inmunología , Femenino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Microambiente Tumoral/inmunología , Regulación hacia Arriba , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Persona de Mediana Edad , Antígenos CD/metabolismo , Antígenos CD/genética , Pronóstico , Regulación Neoplásica de la Expresión Génica , Movimiento Celular , Receptores de Superficie Celular
9.
Front Cell Infect Microbiol ; 14: 1410015, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957797

RESUMEN

Background: Tuberculosis (TB) persists as a global health challenge, with its treatment hampered by the side effects of long-term combination drug therapies and the growing issue of drug resistance. Therefore, the development of novel therapeutic strategies is critical. This study focuses on the role of immune checkpoint molecules (ICs) and functions of CD8+ T cells in the search for new potential targets against TB. Methods: We conducted differential expression genes analysis and CD8+ T cell functional gene analysis on 92 TB samples and 61 healthy individual (HI) samples from TB database GSE83456, which contains data on 34,603 genes. The GSE54992 dataset was used to validated the findings. Additionally, a cluster analysis on single-cell data from primates infected with mycobacterium tuberculosis and those vaccinated with BCG was performed. Results: The overexpression of LAG-3 gene was found as a potentially important characteristic of both pulmonary TB (PTB) and extrapulmonary TB (EPTB). Further correlation analysis showed that LAG-3 gene was correlated with GZMB, perforin, IL-2 and IL-12. A significant temporal and spatial variation in LAG-3 expression was observed in T cells and macrophages during TB infection and after BCG vaccination. Conclusion: LAG-3 was overexpressed in TB samples. Targeting LAG-3 may represent a potential therapeutic target for tuberculosis.


Asunto(s)
Antígenos CD , Linfocitos T CD8-positivos , Proteína del Gen 3 de Activación de Linfocitos , Mycobacterium tuberculosis , Tuberculosis , Humanos , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/genética , Linfocitos T CD8-positivos/inmunología , Tuberculosis/inmunología , Tuberculosis/microbiología , Animales , Antígenos CD/genética , Vacuna BCG/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Interleucina-2/metabolismo , Interleucina-2/genética , Perfilación de la Expresión Génica , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Interleucina-12/genética , Interleucina-12/metabolismo , Perforina/genética , Perforina/metabolismo , Masculino
10.
Nat Commun ; 15(1): 5910, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003273

RESUMEN

Lymphoid specification in human hematopoietic progenitors is not fully understood. To better associate lymphoid identity with protein-level cell features, we conduct a highly multiplexed single-cell proteomic screen on human bone marrow progenitors. This screen identifies terminal deoxynucleotidyl transferase (TdT), a specialized DNA polymerase intrinsic to VDJ recombination, broadly expressed within CD34+ progenitors prior to B/T cell emergence. While these TdT+ cells coincide with granulocyte-monocyte progenitor (GMP) immunophenotype, their accessible chromatin regions show enrichment for lymphoid-associated transcription factor (TF) motifs. TdT expression on GMPs is inversely related to the SLAM family member CD84. Prospective isolation of CD84lo GMPs demonstrates robust lymphoid potentials ex vivo, while still retaining significant myeloid differentiation capacity, akin to LMPPs. This multi-omic study identifies human bone marrow lymphoid-primed progenitors, further defining the lympho-myeloid axis in human hematopoiesis.


Asunto(s)
ADN Nucleotidilexotransferasa , Células Progenitoras Linfoides , Humanos , Antígenos CD/metabolismo , Antígenos CD/genética , Antígenos CD34/metabolismo , Diferenciación Celular , ADN Nucleotidilexotransferasa/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citología , Proteómica/métodos , Análisis de la Célula Individual
11.
Front Immunol ; 15: 1422342, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39076998

RESUMEN

Invasive lobular breast cancer (ILC) is characterized by a relatively high risk for late recurrence and a unique metastatic pattern with an increased risk for metastasis to gynecologic organs and peritoneum. We present a unique case of recurrent ILC with metastasis to the abdominal peritoneum as well as the uterine myometrium and cervix. Treatment was complicated by the discovery of concomitant uterine carcinosarcoma. This patient was effectively treated with a combination of hormonal therapy for her metastatic ILC and a combination of chemotherapy and immunotherapy for uterine carcinosarcoma. Molecular evaluation revealed a characteristic CDH1 mutation within the ILC and a PI3KCA mutation within the uterine carcinosarcoma, both of which have been linked to epithelial-to-mesenchymal transitions. Examination of the tumor immune microenvironment revealed proportionally more cytotoxic NK cells. This robust immune infiltration may be an indicator of the response to immunotherapy observed in this tumor or a result of the metastatic breast cancer within the uterus. This report provides a characterization of the molecular and immunologic landscape in this case with metastatic ILC and uterine carcinosarcoma.


Asunto(s)
Neoplasias de la Mama , Carcinoma Lobular , Carcinosarcoma , Inmunoterapia , Neoplasias Uterinas , Humanos , Femenino , Carcinosarcoma/terapia , Carcinosarcoma/inmunología , Carcinosarcoma/genética , Neoplasias Uterinas/terapia , Neoplasias Uterinas/genética , Neoplasias Uterinas/inmunología , Neoplasias Uterinas/patología , Carcinoma Lobular/inmunología , Carcinoma Lobular/terapia , Carcinoma Lobular/secundario , Carcinoma Lobular/genética , Inmunoterapia/métodos , Neoplasias de la Mama/terapia , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Persona de Mediana Edad , Antígenos CD/genética , Antígenos CD/inmunología , Mutación , Microambiente Tumoral/inmunología , Fosfatidilinositol 3-Quinasa Clase I/genética , Cadherinas/genética , Células Asesinas Naturales/inmunología
12.
Oncoimmunology ; 13(1): 2367843, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38887373

RESUMEN

Conventional type 1 dendritic cells (cDC1) are critical regulators of anti-tumoral T-cell responses. The structure and abundance of intercellular contacts between cDC1 and CD8 T cells in cancer tissues is important to determine the outcome of the T-cell response. However, the molecular determinants controlling the stability of cDC1-CD8 interactions during cancer progression remain poorly investigated. Here, we generated a genetic model of non-small cell lung cancer crossed to a fluorescent cDC1 reporter (KP-XCR1venus) to allow the detection of cDC1-CD8T cell clusters in tumor tissues across tumor stages. We found that cDC1-CD8 clusters are abundant and productive at the early stages of tumor development but progressively diminish in advanced tumors. Transcriptional profiling and flow cytometry identified the adhesion molecule ALCAM/CD166 (Activated Leukocyte Cell Adhesion Molecule, ligand of CD6) as highly expressed by lung cDC1 and significantly downregulated in advanced tumors. Analysis of human datasets indicated that ALCAM is downregulated in non-small cell lung cancer and its expression correlates to better prognosis. Mechanistically, triggering ALCAM on lung cDC1 induces cytoskeletal remodeling and contact formation whereas its blockade prevents T-cell activation. Together, our results indicate that ALCAM is important to stabilize cDC1-CD8 interactions at early tumor stages, while its loss in advanced tumors contributes to immune evasion.


Asunto(s)
Antígenos CD , Linfocitos T CD8-positivos , Carcinoma de Pulmón de Células no Pequeñas , Células Dendríticas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Ratones , Antígenos CD/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Proteínas Fetales/metabolismo , Proteínas Fetales/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Comunicación Celular/inmunología , Molécula de Adhesión Celular del Leucocito Activado
13.
Emerg Microbes Infect ; 13(1): 2366359, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38855910

RESUMEN

Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.


Asunto(s)
Coinfección , Granuloma , Infecciones por VIH , Pulmón , Macrófagos , Receptores de Interleucina-6 , Factor de Transcripción STAT3 , Humanos , Coinfección/virología , Coinfección/inmunología , Coinfección/microbiología , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Macrófagos/inmunología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Granuloma/inmunología , Pulmón/patología , Pulmón/inmunología , Receptores de Interleucina-6/metabolismo , Receptores de Interleucina-6/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos CD/metabolismo , Antígenos CD/genética , Transducción de Señal , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/complicaciones , Masculino , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/complicaciones , Femenino , Adulto , Interleucina-6/metabolismo , Interleucina-6/genética , Molécula CD68
14.
Iran J Allergy Asthma Immunol ; 23(2): 220-230, 2024 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-38822516

RESUMEN

During epithelial to mesenchymal transition, the ability of cancer cells to transform and metastasize is primarily determined by N-cadherin-mediated migration and invasion. This study aimed to evaluate whether the N-cadherin promoter can induce diphtheria toxin expression as a suicide gene in epithelial to mesenchymal transition (EMT)-induced cancer cells and whether this can be used as potential gene therapy. To investigate the expression of diphtheria toxin under the N-cadherin promoter, the promoter was synthesized, and was cloned upstream of diphtheria toxin in a pGL3-Basic vector. The A-549 cells was transfected by electroporation. After induction of EMT by TGF-ß and hypoxia treatment, the relative expression of diphtheria toxin, mesenchymal genes such as N-cadherin and Vimentin, and epithelial genes such as E-cadherin and ß-catenin were measured by real-time PCR. MTT assay was also performed to measure cytotoxicity. Finally, cell motility was assessed by the Scratch test. After induction of EMT in transfected cells, the expression of mesenchymal markers such as Vimentin and N-cadherin significantly decreased, and the expression of ß-catenin increased. In addition, the MTT assay showed promising toxicity results after induction of EMT with TGF-ß in transfected cells, but toxicity was less effective in hypoxia. The scratch test results also showed that cell movement was successfully prevented in EMT-transfected cells and thus confirmed EMT occlusion. Our findings indicate that by using structures containing diphtheria toxin downstream of a specific EMT promoter such as the N-cadherin promoter, the introduced toxin can kill specifically and block EMT in cancer cells.


Asunto(s)
Cadherinas , Toxina Diftérica , Transición Epitelial-Mesenquimal , Regiones Promotoras Genéticas , Humanos , Células A549 , Antígenos CD/genética , Antígenos CD/metabolismo , beta Catenina/metabolismo , beta Catenina/genética , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular/genética , Movimiento Celular/efectos de los fármacos , Toxina Diftérica/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Genes Transgénicos Suicidas , Regiones Promotoras Genéticas/genética , Vimentina/genética , Vimentina/metabolismo
15.
Nat Commun ; 15(1): 5180, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890323

RESUMEN

Siglec-6 is a lectin receptor with restricted expression in the placenta, mast cells and memory B-cells. Although Siglec-6 is expressed in patients with chronic lymphocytic leukemia (CLL), its pathophysiological role has not been elucidated. We describe here a role for Siglec-6 in migration and adhesion of CLL B cells to CLL- bone marrow stromal cells (BMSCs) in vitro and compromised migration to bone marrow and spleen in vivo. Mass spectrometry analysis revealed interaction of Siglec-6 with DOCK8, a guanine nucleotide exchange factor. Stimulation of MEC1-002 CLL cells with a Siglec-6 ligand, sTn, results in Cdc42 activation, WASP protein recruitment and F-actin polymerization, which are all associated with cell migration. Therapeutically, a Siglec-6/CD3-bispecific T-cell-recruiting antibody (T-biAb) improves overall survival in an immunocompetent mouse model and eliminates CLL cells in a patient derived xenograft model. Our findings thus reveal a migratory role for Siglec-6 in CLL, which can be therapeutically targeted using a Siglec-6 specific T-biAb.


Asunto(s)
Adhesión Celular , Movimiento Celular , Lectinas , Leucemia Linfocítica Crónica de Células B , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Humanos , Animales , Lectinas/metabolismo , Ratones , Antígenos CD/metabolismo , Antígenos CD/genética , Femenino , Linfocitos B/metabolismo , Linfocitos B/inmunología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Línea Celular Tumoral , Células Madre Mesenquimatosas/metabolismo , Masculino , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Sci Rep ; 14(1): 14251, 2024 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902412

RESUMEN

Acute myeloid leukaemia (AML) is an aggressive leukaemia characterised by uncontrolled blast cell proliferation. miRNAs and Clusters of Differentiation (CD) molecules play essential roles in AML progression. This study aims to investigate the effect of COVID-19 on the expression of circulating miRNA and CD molecules in AML. This cross-sectional study recruited 32 AML patients and 20 controls. Blood samples were collected and analysed using molecular cytogenetic, miRNA/mRNA expression, and flow cytometry techniques. The expression of miRNAs varied significantly between patients with AML and control individuals. The co-expression of these miRNAs was higher (P < 0.05), indicating that the presence of one miRNA led to increased expression of other miRNAs. A differential correlation was observed between miRNAs and CD markers. Additionally, miRNA 16, miRNA 21, and miRNA 221 showed significant downregulation (P < 0.05 and P < 0.01, respectively) in AML patients with COVID-19 infection compared to those without a disease. Interestingly, this study identified a higher expression level (P < 0.01) of miRNA 137 as a novel biomarker for AML patients. Moreover, the expression of miRNA 137 showed a high correlation (P < 0.05) with most of the CD markers examined in this study and FISH features data. Furthermore, a strong correlation (P < 0.01) was observed between CD markers and miRNA among AML patients with positive and negative COVID-19 infection. These data demonstrated that COVID-19 contributed to increased expression of microRNAs in AML patients. MicroRNA 137 was identified as a novel microRNA that exhibited significant differences between patients and healthy individuals, highlighting its role in AML pathogenesis.


Asunto(s)
COVID-19 , Leucemia Mieloide Aguda , MicroARNs , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/sangre , COVID-19/genética , COVID-19/sangre , COVID-19/virología , Masculino , Femenino , MicroARNs/sangre , MicroARNs/genética , Persona de Mediana Edad , Adulto , Estudios Transversales , Anciano , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , SARS-CoV-2/aislamiento & purificación , Antígenos CD/genética , Antígenos CD/sangre , Antígenos CD/metabolismo
17.
Medicine (Baltimore) ; 103(23): e38244, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38847676

RESUMEN

BACKGROUND: Numerous studies have investigated the association between CDH1 polymorphisms and gastric cancer (GC) risk. However, the results have been inconsistent and controversial. To further determine whether CDH1 polymorphisms increase the risk of GC, we conducted a meta-analysis by pooling the data. METHODS: Relevant case-control studies were collected from PubMed, Embase, Web of Science and Cochrane databases up to January 7, 2024. Subsequently, odds ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate the strength of correlations. A sensitivity analysis was performed to evaluate the robustness and reliability of these included studies. RESULTS: A total of 25 articles including 44 studies, were included in this meta-analysis, including 26 studies on rs16260, 6 studies on rs3743674, 7 studies on rs5030625, and 5 studies on rs1801552. The pooled results showed that rs16260 was remarkably associated with an increased GC risk of GC among Caucasians. Moreover, the rs5030625 variation dramatically enhanced GC predisposition in the Asian population. However, no evident correlations between CDH1 rs3743674 and rs1801552 polymorphisms and GC risk were observed. CONCLUSIONS: Our findings suggested that CDH1 gene polymorphisms were significantly correlated with GC risk, especially in rs16260 and rs5030625 polymorphisms.


Asunto(s)
Antígenos CD , Cadherinas , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Neoplasias Gástricas , Neoplasias Gástricas/genética , Humanos , Cadherinas/genética , Antígenos CD/genética , Pueblo Asiatico/genética , Estudios de Casos y Controles , Población Blanca/genética , Factores de Riesgo
18.
Sci Rep ; 14(1): 14343, 2024 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-38906929

RESUMEN

Non-small cell lung cancer (NSCLC)-originating cancer-associated fibroblasts (CAFs) expressing CD248 regulate interaction with immune cells to accelerate cancer progression. Epithelial-mesenchymal transition (EMT) is a key feature of metastatic cells. In our pervious study, we found that CD248+CAFs activated M2-polarized macrophages, enhancing the progression of NSCLC. However, it is yet unclear how CD248+CAFs inducing M2-polarized macrophages induce EMT program in NSCLC cells. Herein, we examined CD248 expression from CAFs derived from NSCLC patient tumour tissues. Furthermore, we determined the influence of CD248 knock down CAFs on macrophages polarization. Next, we explored the influences of CD248-harboring CAFs-mediated M2 macrophage polarization to promote NSCLC cells EMT in vitro. We constructed fibroblasts specific CD248 gene knock out mice to examine the significance of CD248-harboring CAFs-induced M2-polarized macrophages to promote NSCLC cells EMT in vivo. Based on our analysis, CD248 is ubiquitously expressed within NSCLC-originating CAFs. CD248+CAFs mediated macrophages polarized to M2 type macrophages. CD248+CAFs induced M2 macrophage polarization to enhance NSCLC cells EMT both in vivo and in vitro. Our findings indicate that CD248-harboring CAFs promote NSCLC cells EMT by regulating M2-polarized macrophages.


Asunto(s)
Antígenos CD , Fibroblastos Asociados al Cáncer , Carcinoma de Pulmón de Células no Pequeñas , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares , Macrófagos , Transición Epitelial-Mesenquimal/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Humanos , Animales , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Macrófagos/metabolismo , Ratones , Antígenos CD/metabolismo , Antígenos CD/genética , Ratones Noqueados , Línea Celular Tumoral , Antígenos de Neoplasias
19.
Cancer Genomics Proteomics ; 21(4): 405-413, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38944419

RESUMEN

BACKGROUND/AIM: Metastatic prostate cancer (mPCa) results in high morbidity and mortality. Visceral metastases in particular are associated with a shortened survival. Our aim was to unravel the molecular mechanisms that underly pulmonary spread in mPCa. MATERIALS AND METHODS: We performed a comprehensive transcriptomic analysis of PCa lung metastases, followed by functional validation of candidate genes. Digital gene expression analysis utilizing the NanoString technology was performed on mRNA extracted from formalin-fixed, paraffin-embedded (FFPE) tissue from PCa lung metastases. The gene expression data from primary PCa and PCa lung metastases were compared, and several publicly available bioinformatic analysis tools were used to annotate and validate the data. RESULTS: In PCa lung metastases, 234 genes were considerably up-regulated, and 78 genes were significantly down-regulated when compared to primary PCa. Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) was identified as suitable candidate gene for further functional validation. CEACAM6 as a cell adhesion molecule has been implicated in promoting metastatic disease in several solid tumors, such as colorectal or gastric cancer. We showed that siRNA knockdown of CEACAM6 in PC-3 and LNCaP cells resulted in decreased cell viability and migration as well as enhanced apoptosis. Comprehensive transcriptomic analyses identified several genes of interest that might promote metastatic spread to the lung. CONCLUSION: Functional validation revealed that CEACAM6 might play an important role in fostering metastatic spread to the lung of PCa patients via enhancing proliferation, migration and suppressing apoptosis in PC-3 and LNCaP cells. CEACAM6 might pose an attractive therapeutic target to prevent metastatic disease.


Asunto(s)
Antígenos CD , Apoptosis , Moléculas de Adhesión Celular , Movimiento Celular , Proliferación Celular , Proteínas Ligadas a GPI , Neoplasias Pulmonares , Neoplasias de la Próstata , Humanos , Masculino , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/metabolismo , Antígenos CD/metabolismo , Antígenos CD/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
20.
Oncol Rep ; 52(2)2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38940341

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a type of liver cancer associated with poor prognosis and increased mortality; the limited treatment strategy highlights the urgent need for investigation. Traditional Chinese Medicine (TCM), used alone or in combination with other treatments, can enhance therapeutic efficacy, improve life quality of patients and extend overall survival. In total, two rounds of screening of a TCM library of 2,538 active compounds were conducted using a Cell Counting Kit­8 assay and ICC cell lines. Cell proliferation and migration abilities were assessed through colony formation, 5­ethynyl­2'­deoxyuridine, would healing and Transwell assays. The impact of digitoxin (DT) on signaling pathways was initially investigated using RNA sequencing and further validated using reverse transcription­quantitative PCR, western blotting, lectin blotting and flow cytometry. ICC cells stably overexpressing ST6 ß­galactoside α­2,6­sialyltransferase 1 (ST6GAL1) were generated through lentiviral transfection. It was shown that DT emerged as a highly effective anti­ICC candidate from two rounds high­throughput library screening. DT could inhibit the proliferation and migration of ICC cells by suppressing NF­κB activation and reducing nuclear phosphorylated­NF­κB levels, along with diminishing ST6GAL1 mRNA and protein expression. The aforementioned biological effects and signal pathways of DT could be counteracted by overexpressing ST6GAL1 in ICC cells. In conclusion, DT suppressed ICC cell proliferation and migration by targeting the NF­κB/ST6GAL1 signaling axis. The findings of the present study indicated the promising therapeutic effects of DT in managing ICC, offering new avenues for treatment strategies.


Asunto(s)
Neoplasias de los Conductos Biliares , Proliferación Celular , Colangiocarcinoma , Digitoxina , Transducción de Señal , Humanos , Antígenos CD/metabolismo , Antígenos CD/genética , beta-D-Galactósido alfa 2-6-Sialiltransferasa/metabolismo , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/genética , Digitoxina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , FN-kappa B/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA