Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.697
Filtrar
1.
PeerJ ; 12: e17350, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827297

RESUMEN

Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related deaths, with very limited therapeutic options available. This study aims to comprehensively depict the heterogeneity and identify prognostic targets for PDAC with single-cell RNA sequencing (scRNA-seq) analysis. Methods: ScRNA-seq analysis was performed on 16 primary PDAC and three adjacent lesions. A series of analytical methods were applied for analysis in cell clustering, gene profiling, lineage trajectory analysis and cell-to-cell interactions. In vitro experiments including colony formation, wound healing and sphere formation assay were performed to assess the role of makers. Results: A total of 32,480 cells were clustered into six major populations, among which the ductal cell cluster expressing high copy number variants (CNVs) was defined as malignant cells. Malignant cells were further subtyped into five subgroups which exhibited specific features in immunologic and metabolic activities. Pseudotime trajectory analysis indicated that components of various oncogenic pathways were differentially expressed along tumor progression. Furthermore, intensive substantial crosstalk between ductal cells and stromal cells was identified. Finally, genes (REG4 and SPINK1) screened out of differentially expressed genes (DEGs) were upregulated in PDAC cell lines. Silencing either of them significantly impaired proliferation, invasion, migration and stemness of PDAC cells. Conclusions: Our findings offer a valuable resource for deciphering the heterogeneity of malignant ductal cells in PDAC. REG4 and SPINK1 are expected to be promising targets for PDAC therapy.


Asunto(s)
Carcinoma Ductal Pancreático , Lectinas Tipo C , Neoplasias Pancreáticas , Análisis de la Célula Individual , Transcriptoma , Inhibidor de Tripsina Pancreática de Kazal , Humanos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Inhibidor de Tripsina Pancreática de Kazal/genética , Inhibidor de Tripsina Pancreática de Kazal/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Pronóstico , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Femenino , Masculino , Proteínas Asociadas a Pancreatitis
2.
Eur J Dermatol ; 34(2): 119-130, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38907541

RESUMEN

Psoriasis is a common skin disease with a high recurrence rate. Aberrant keratinocyte proliferation is a significant pathogenic characteristic of psoriatic lesions, and studies have revealed that the development of psoriasis is significantly influenced by pro-inflammatory cytokines, such as IL-17A and TNF-α. Biologics targeting these cytokines have been widely used in psoriasis treatment and achieve remarkable effects, however, the underlying mechanism of how IL-17A and TNF-α specifically regulate keratinocyte proliferation has not been fully elucidated. Dectin-1 is an essential membrane protein that is directly related to the immune microenvironment and the proliferation of multiple cell types. To elucidate how IL-17A and TNF-α may promote keratinocyte proliferation in psoriatic lesions and whether Dectin-1 is involved. The expression of Dectin-1 in keratinocytes from psoriatic lesions was detected by real-time PCR, western blot and immunofluorescence. Correlation analysis and cytological experiments were then performed to determine the relationship between Dectin-1 and IL-17A/TNF-α in psoriatic lesions. Finally, we investigated the signalling pathway through which Dectin-1 may promote keratinocyte proliferation. Dectin-1 was significantly increased in keratinocytes from psoriatic lesions. Moreover, IL-17A and TNF-α effectively induced the expression of Dectin-1 in HaCaT cells, which was shown to activate the Syk/NF-κB signalling pathway and promote the proliferation of keratinocytes. IL-17A and TNF-α may promote the proliferation of keratinocytes in psoriatic lesions through induction of Dectin-1, indicating that Dectin-1 could be a potential therapeutic target for the treatment of psoriasis.


Asunto(s)
Proliferación Celular , Interleucina-17 , Queratinocitos , Lectinas Tipo C , Psoriasis , Transducción de Señal , Factor de Necrosis Tumoral alfa , Humanos , Psoriasis/metabolismo , Psoriasis/patología , Queratinocitos/metabolismo , Interleucina-17/metabolismo , Lectinas Tipo C/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , FN-kappa B/metabolismo , Quinasa Syk/metabolismo , Masculino , Femenino , Células Cultivadas , Adulto
3.
Cell Commun Signal ; 22(1): 337, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898461

RESUMEN

Killer cell lectin-like receptor G1 (KLRG1) is an immune checkpoint receptor expressed predominantly in NK and T-cell subsets that downregulates the activation and proliferation of immune cells and participates in cell-mediated immune responses. Accumulating evidence has demonstrated the importance of KLRG1 as a noteworthy disease marker and therapeutic target that can influence disease onset, progression, and prognosis. Blocking KLRG1 has been shown to effectively mitigate the effects of downregulation in various mouse tumor models, including solid tumors and hematologic malignancies. However, KLRG1 inhibitors have not yet been approved for human use, and the understanding of KLRG1 expression and its mechanism of action in various diseases remains incomplete. In this review, we explore alterations in the distribution, structure, and signaling pathways of KLRG1 in immune cells and summarize its expression patterns and roles in the development and progression of autoimmune diseases, infectious diseases, and cancers. Additionally, we discuss the potential applications of KLRG1 as a tool for tumor immunotherapy.


Asunto(s)
Lectinas Tipo C , Neoplasias , Receptores Inmunológicos , Humanos , Receptores Inmunológicos/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/antagonistas & inhibidores , Animales , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Biomarcadores/metabolismo , Transducción de Señal , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Inmunoterapia
4.
ACS Appl Bio Mater ; 7(6): 3877-3889, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38832760

RESUMEN

Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.


Asunto(s)
Antígenos Bacterianos , Lectinas Tipo C , Nanopartículas , Dióxido de Silicio , Células Th17 , Lectinas Tipo C/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/agonistas , Nanopartículas/química , Células Th17/inmunología , Animales , Dióxido de Silicio/química , Ratones , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/química , Mycobacterium tuberculosis/inmunología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Tamaño de la Partícula , Ensayo de Materiales , Humanos , Femenino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/agonistas
5.
Nat Commun ; 15(1): 4728, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830864

RESUMEN

Due to their exceptional solubility and stability, nanobodies have emerged as powerful building blocks for research tools and therapeutics. However, their generation in llamas is cumbersome and costly. Here, by inserting an engineered llama immunoglobulin heavy chain (IgH) locus into IgH-deficient mice, we generate a transgenic mouse line, which we refer to as 'LamaMouse'. We demonstrate that LamaMice solely express llama IgH molecules without association to Igκ or λ light chains. Immunization of LamaMice with AAV8, the receptor-binding domain of the SARS-CoV-2 spike protein, IgE, IgG2c, and CLEC9A enabled us to readily select respective target-specific nanobodies using classical hybridoma and phage display technologies, single B cell screening, and direct cloning of the nanobody-repertoire into a mammalian expression vector. Our work shows that the LamaMouse represents a flexible and broadly applicable platform for a facilitated selection of target-specific nanobodies.


Asunto(s)
Camélidos del Nuevo Mundo , Cadenas Pesadas de Inmunoglobulina , Ratones Transgénicos , Anticuerpos de Dominio Único , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Camélidos del Nuevo Mundo/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Ratones , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/química , Lectinas Tipo C/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/genética , SARS-CoV-2/inmunología , SARS-CoV-2/genética , Inmunoglobulina E/inmunología , Humanos , Dependovirus/genética , Dependovirus/inmunología , Inmunoglobulina G/inmunología , COVID-19/inmunología , Linfocitos B/inmunología
6.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38892398

RESUMEN

Myocardial infarction activates an intense fibro-inflammatory reaction that is essential for cardiac remodeling and heart failure (HF). Bioactive peptide galanin plays a critical role in regulating cardiovascular homeostasis; however, its specific functional relevance in post-infarction fibro-inflammatory reprogramming remains obscure. Here, we show that galanin coordinates the fibro-inflammatory trajectory and mitochondrial integrity in post-infarction reperfusion injury. Aberrant deposition of collagen was associated with a marked increase in CD68-positive macrophage infiltration in cardiac tissue in mice subjected to myocardial ischemia/reperfusion (I/R) for 14 days compared to sham controls. Furthermore, we found that the myocardial expression level of a specific marker of M2 macrophages, CD206, was significantly down-regulated in I/R-challenged mice. In contrast, galanin treatment started during the reperfusion phase blunted the fibro-inflammatory responses and promoted the expression of CD206 in I/R-remodeled hearts. In addition, we found that the anti-apoptotic and anti-hypertrophic effects of galanin were associated with the preservation of mitochondrial integrity and promotion of mitochondrial biogenesis. These findings depict galanin as a key arbitrator of fibro-inflammatory responses to cardiac I/R injury and offer a promising therapeutic trajectory for the treatment of post-infarct cardiovascular complications.


Asunto(s)
Galanina , Macrófagos , Daño por Reperfusión Miocárdica , Animales , Galanina/metabolismo , Galanina/farmacología , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Macrófagos/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Mitocondrias/metabolismo , Ratones Endogámicos C57BL , Receptores de Superficie Celular/metabolismo , Inflamación/metabolismo , Inflamación/patología , Receptor de Manosa , Lectinas Tipo C/metabolismo , Miocardio/metabolismo , Miocardio/patología , Lectinas de Unión a Manosa/metabolismo , Modelos Animales de Enfermedad , Apoptosis
7.
Front Immunol ; 15: 1361351, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38846954

RESUMEN

Background: Gliomas constitute a category of malignant tumors originating from brain tissue, representing the majority of intracranial malignancies. Previous research has demonstrated the pivotal role of CLEC7A in the progression of various cancers, yet its specific implications within gliomas remain elusive. The primary objective of this study was to investigate the prognostic significance and immune therapeutic potential of CLEC7A in gliomas through the integration of bioinformatics and clinical pathological analyses. Methods: This investigation involved examining and validating the relationship between CLEC7A and glioma using samples from Hospital, along with data from TCGA, GEO, GTEx, and CGGA datasets. Subsequently, we explored its prognostic value, biological functions, expression location, and impact on immune cells within gliomas. Finally, we investigated its potential impact on the chemotaxis and polarization of macrophages. Results: The expression of CLEC7A is upregulated in gliomas, and its levels escalate with the malignancy of tumors, establishing it as an independent prognostic factor. Functional enrichment analysis revealed a significant correlation between CLEC7A and immune function. Subsequent examination of immune cell differential expression demonstrated a robust association between CLEC7A and M2 macrophages. This conclusion was further substantiated through single-cell analysis, immunofluorescence, and correlation studies. Finally, the knockout of CLEC7A in M2 macrophages resulted in a noteworthy reduction in macrophage chemotaxis and polarization factors. Conclusion: CLEC7A expression is intricately linked to the pathology and molecular characteristics of gliomas, establishing its role as an independent prognostic factor for gliomas and influencing macrophage function. It could be a promising target for immunotherapy in gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Lectinas Tipo C , Macrófagos , Microambiente Tumoral , Humanos , Glioma/inmunología , Glioma/genética , Glioma/patología , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Pronóstico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
8.
Scand J Immunol ; 99(6): e13364, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38720521

RESUMEN

Mucosal-associated invariant T-cells (MAIT) are unconventional T-cells with cytotoxic and pro-inflammatory properties. Previous research has reported contradictory findings on their role in cancerogenesis with data being even scarcer in haematological malignancies. Here, we report the results of a systematic analysis of MAIT cells in treatment-naïve patients with a broad range of haematological malignancies. We analysed peripheral blood of 204 patients and 50 healthy subjects. The pool of haematological patients had a statistically significant lower both the absolute value (median values, 0.01 × 109/L vs. 0.05 × 109/L) of MAIT cells and their percentage (median values 0.94% vs. 2.56%) among T-cells compared to the control group. Separate analysis showed that the decrease in the absolute number of MAIT cells is significant in patients with acute myeloid leukaemia, myeloproliferative neoplasms, plasma cell myeloma, B-cell non-Hodgkin lymphomas, otherwise not specified, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone lymphoma compared to the control population. Furthermore, in haematological malignancies, MAIT cells overexpress PD-1 (average values, 51.7% vs. 6.7%), HLA-DR (average values, 40.2% vs. 7%), CD38 (average values, 25.9% vs. 4.9%) and CD69 (average values, 40.2% vs. 9.2%). Similar results were obtained when comparing patients with individual malignancies to the control population. Our data show that the depletion of circulating MAIT cells is a common observation in a broad spectrum of haematological malignancies. In addition to their reduced numbers, MAIT cells acquire an activated/exhausted phenotype.


Asunto(s)
Neoplasias Hematológicas , Células T Invariantes Asociadas a Mucosa , Receptor de Muerte Celular Programada 1 , Humanos , Células T Invariantes Asociadas a Mucosa/inmunología , Neoplasias Hematológicas/inmunología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Adulto , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Antígenos CD/metabolismo , Anciano de 80 o más Años , Antígenos de Diferenciación de Linfocitos T/metabolismo , Recuento de Linfocitos , ADP-Ribosil Ciclasa 1/metabolismo , ADP-Ribosil Ciclasa 1/inmunología , Inmunofenotipificación , Adulto Joven , Glicoproteínas de Membrana/inmunología , Lectinas Tipo C
9.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791534

RESUMEN

C-type lectins play a crucial role as pathogen-recognition receptors for the dengue virus, which is responsible for causing both dengue fever (DF) and dengue hemorrhagic fever (DHF). DHF is a serious illness caused by the dengue virus, which exists in four different serotypes: DEN-1, DEN-2, DEN-3, and DEN-4. We conducted a genetic association study, during a significant DEN-2 outbreak in southern Taiwan, to explore how variations in the neck-region length of L-SIGN (also known as CD209L, CD299, or CLEC4M) impact the severity of dengue infection. PCR genotyping was utilized to identify polymorphisms in variable-number tandem repeats. We constructed L-SIGN variants containing either 7- or 9-tandem repeats and transfected these constructs into K562 and U937 cells, and cytokine and chemokine levels were evaluated using enzyme-linked immunosorbent assays (ELISAs) following DEN-2 virus infection. The L-SIGN allele 9 was observed to correlate with a heightened risk of developing DHF. Subsequent results revealed that the 9-tandem repeat was linked to elevated viral load alongside predominant T-helper 2 (Th2) cell responses (IL-4 and IL-10) in K562 and U937 cells. Transfecting K562 cells in vitro with L-SIGN variants containing 7- and 9-tandem repeats confirmed that the 9-tandem repeat transfectants facilitated a higher dengue viral load accompanied by increased cytokine production (MCP-1, IL-6, and IL-8). Considering the higher prevalence of DHF and an increased frequency of the L-SIGN neck's 9-tandem repeat in the Taiwanese population, individuals with the 9-tandem repeat may necessitate more stringent protection against mosquito bites during dengue outbreaks in Taiwan.


Asunto(s)
Virus del Dengue , Lectinas Tipo C , Receptores de Superficie Celular , Dengue Grave , Replicación Viral , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Dengue Grave/inmunología , Dengue Grave/virología , Dengue Grave/genética , Virus del Dengue/genética , Virus del Dengue/inmunología , Replicación Viral/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Masculino , Células K562 , Femenino , Células U937 , Taiwán/epidemiología , Repeticiones de Minisatélite/genética , Adulto , Citocinas/metabolismo , Citocinas/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Persona de Mediana Edad , Carga Viral
10.
Fish Shellfish Immunol ; 149: 109594, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697376

RESUMEN

Non-specific cytotoxic cells (NCCs) are vital immune cells involved in teleost's non-specific immunity. As a receptor molecule on the NCCs' surface, the non-specific cytotoxic cell receptor protein 1 (NCCRP-1) is known to play a crucial role in mediating their activity. Nevertheless, there have been limited studies on the signal molecule that transmits signals via NCCRP-1. In this study, a yeast two-hybrid (Y2H) library of tilapia liver and head kidney was constructed and subsequently screened with the bait vector NCCRP-1 of Oreochromis niloticus (On-NCCRP-1) to obtain a C-type lectin (On-CTL) with an interacting protein sequence. Consequently, the full-length sequence of On-CTL was cloned and analyzed. The expression analysis revealed that On-CTL is highly expressed in the liver and is widely distributed in other tissues. Furthermore, On-CTL expression was significantly up-regulated in the brain, intestine, and head kidney following a challenge with Streptococcus agalactiae. A point-to-point Y2H method was also used to confirm the binding between On-NCCRP-1 and On-CTL. The recombinant On-CTL (rOn-CTL) protein was purified. In vitro experiments demonstrated that rOn-CTL can up-regulate the expression of killer effector molecules in NCCs via its interaction with On-NCCRP-1. Moreover, activation of NCCs by rOn-CTL resulted in a remarkable enhancement in their ability to eliminate fathead minnow cells, indicating that rOn-CTL effectively modulates the killing activity of NCCs through the NCC receptor molecule On-NCCRP-1. These findings significantly contribute to our comprehension of the regulatory mechanisms governing NCC activity, paving the way for future research in this field.


Asunto(s)
Cíclidos , Enfermedades de los Peces , Proteínas de Peces , Lectinas Tipo C , Streptococcus agalactiae , Animales , Cíclidos/inmunología , Cíclidos/genética , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Lectinas Tipo C/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Enfermedades de los Peces/inmunología , Streptococcus agalactiae/fisiología , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/veterinaria , Regulación de la Expresión Génica/inmunología , Secuencia de Aminoácidos , Inmunidad Innata/genética , Alineación de Secuencia/veterinaria , Filogenia , Perfilación de la Expresión Génica/veterinaria
11.
PLoS One ; 19(5): e0303945, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38776335

RESUMEN

Killer cell lectin-like receptor G1 (KLRG1) has traditionally been regarded as an inhibitory receptor of T cell exhaustion in chronic infection and inflammation. However, its exact role in hepatitis B virus (HBV) infection remains elusive. CD8+ T cells from 190 patients with chronic hepatitis B were analyzed ex vivo for checkpoint and apoptosis markers, transcription factors, cytokines and subtypes in 190 patients with chronic hepatitis B. KLRG1+ and KLRG1- CD8+ T cells were sorted for transcriptome analysis. The impact of the KLRG1-E-cadherin pathway on the suppression of HBV replication mediated by virus-specific T cells was validated in vitro. As expected, HBV-specific CD8+ T cells expressed higher levels of KLRG1 and showed an exhausted molecular phenotype and function. However, despite being enriched for the inhibitory molecules, thymocyte selection-associated high mobility group box protein (TOX), eomesodermin (EOMES), and Helios, CD8+ T cells expressing KLRG1 produced significant levels of tumour necrosis factor (TNF)-α, interferon (IFN)-γ, perforin, and granzyme B, demonstrating not exhausted but active function. Consistent with the in vitro phenotypic assay results, RNA sequencing (RNA-seq) data showed that signature effector T cell and exhausted T cell genes were enriched in KLRG1+ CD8+ T cells. Furthermore, in vitro testing confirmed that KLRG1-E-cadherin binding inhibits the antiviral efficacy of HBV-specific CD8+ T cells. Based on these findings, we concluded that KLRG1+ CD8+ T cells are not only a terminally exhausted subgroup but also exhibit functional diversity, despite inhibitory signs in HBV infection.


Asunto(s)
Linfocitos T CD8-positivos , Virus de la Hepatitis B , Hepatitis B Crónica , Lectinas Tipo C , Receptores Inmunológicos , Humanos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Receptores Inmunológicos/metabolismo , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Femenino , Masculino , Virus de la Hepatitis B/inmunología , Adulto , Persona de Mediana Edad , Replicación Viral , Cadherinas/metabolismo , Cadherinas/genética , Perforina/metabolismo , Perforina/genética
12.
Sci Rep ; 14(1): 11898, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789479

RESUMEN

We have previously reported the transcriptomic and lipidomic profile of the first-generation, hygromycin-resistant (HygR) version of the BCGΔBCG1419c vaccine candidate, under biofilm conditions. We recently constructed and characterized the efficacy, safety, whole genome sequence, and proteomic profile of a second-generation version of BCGΔBCG1419c, a strain lacking the BCG1419c gene and devoid of antibiotic markers. Here, we compared the antibiotic-less BCGΔBCG1419c with BCG. We assessed their colonial and ultrastructural morphology, biofilm, c-di-GMP production in vitro, as well as their transcriptomic and lipidomic profiles, including their capacity to activate macrophages via Mincle and Myd88. Our results show that BCGΔBCG1419c colonial and ultrastructural morphology, c-di-GMP, and biofilm production differed from parental BCG, whereas we found no significant changes in its lipidomic profile either in biofilm or planktonic growth conditions. Transcriptomic profiling suggests changes in BCGΔBCG1419c cell wall and showed reduced transcription of some members of the DosR, MtrA, and ArgR regulons. Finally, induction of TNF-α, IL-6 or G-CSF by bone-marrow derived macrophages infected with either BCGΔBCG1419c or BCG required Mincle and Myd88. Our results confirm that some differences already found to occur in HygR BCGΔBCG1419c compared with BCG are maintained in the antibiotic-less version of this vaccine candidate except changes in production of PDIM. Comparison with previous characterizations conducted by OMICs show that some differences observed in BCGΔBCG1419c compared with BCG are maintained whereas others are dependent on the growth condition employed to culture them.


Asunto(s)
Vacuna BCG , Biopelículas , GMP Cíclico , Lipidómica , Macrófagos , Mycobacterium bovis , Factor 88 de Diferenciación Mieloide , Transcriptoma , Animales , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Ratones , Macrófagos/metabolismo , Macrófagos/inmunología , Vacuna BCG/inmunología , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Mycobacterium bovis/genética , Mycobacterium bovis/inmunología , Biopelículas/crecimiento & desarrollo , Citocinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Perfilación de la Expresión Génica , Lectinas Tipo C
13.
Front Cell Infect Microbiol ; 14: 1334211, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817444

RESUMEN

Parasites possess remarkable abilities to evade and manipulate the immune response of their hosts. Echinococcus granulosus is a parasitic tapeworm that causes cystic echinococcosis in animals and humans. The hydatid fluid released by the parasite is known to contain various immunomodulatory components that manipulate host´s defense mechanism. In this study, we focused on understanding the effect of hydatid fluid on dendritic cells and its impact on autophagy induction and subsequent T cell responses. Initially, we observed a marked downregulation of two C-type lectin receptors in the cell membrane, CLEC9A and CD205 and an increase in lysosomal activity, suggesting an active cellular response to hydatid fluid. Subsequently, we visualized ultrastructural changes in stimulated dendritic cells, revealing the presence of macroautophagy, characterized by the formation of autophagosomes, phagophores, and phagolysosomes in the cell cytoplasm. To further elucidate the underlying molecular mechanisms involved in hydatid fluid-induced autophagy, we analyzed the expression of autophagy-related genes in stimulated dendritic cells. Our results demonstrated a significant upregulation of beclin-1, atg16l1 and atg12, indicating the induction of autophagy machinery in response to hydatid fluid exposure. Additionally, using confocal microscopy, we observed an accumulation of LC3 in dendritic cell autophagosomes, confirming the activation of this catabolic pathway associated with antigen presentation. Finally, to evaluate the functional consequences of hydatid fluid-induced autophagy in DCs, we evaluated cytokine transcription in the splenocytes. Remarkably, a robust polyfunctional T cell response, with inhibition of Th2 profile, is characterized by an increase in the expression of il-6, il-10, il-12, tnf-α, ifn-γ and tgf-ß genes. These findings suggest that hydatid fluid-induced autophagy in dendritic cells plays a crucial role in shaping the subsequent T cell responses, which is important for a better understanding of host-parasite interactions in cystic echinococcosis.


Asunto(s)
Autofagia , Células Dendríticas , Equinococosis , Echinococcus granulosus , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Echinococcus granulosus/inmunología , Autofagia/inmunología , Equinococosis/inmunología , Equinococosis/parasitología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones , Lectinas Tipo C/metabolismo , Citocinas/metabolismo , Femenino , Autofagosomas/inmunología , Autofagosomas/metabolismo
14.
J Thromb Haemost ; 22(6): 1749-1757, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811291

RESUMEN

BACKGROUND: An iron overload status induces ferroptosis, an iron-dependent nonapoptotic cell death, in various pathological conditions. We previously reported that hemin (heme), protoporphyrin-IX with ferric iron, activates platelets via C-type lectin-like receptor-2 (CLEC-2) and glycoprotein VI/FcRγ, but protoporphyrin-IX alone blocks CLEC-2-dependent platelet activation. Therefore, we hypothesized that free iron has the ability to activate platelets. OBJECTIVES: This study aimed to elucidate platelet activation mechanisms of iron (ferric chloride), including the identification of signaling pathways and receptors, and to examine whether platelets regulate ferroptosis. METHODS: Platelet aggregometry, platelet activation marker expression, and protein phosphorylation were examined in ferric chloride-stimulated human and murine platelets. Inhibitors of platelet activation signaling pathways and receptor-deleted platelets were utilized to identify the responsible signaling pathway and receptor. The effect of platelets on ferroptosis of endothelial cells was investigated in vitro. RESULTS: Ferric chloride induced platelet activation dependent on Src family kinase pathways in humans and mice. Ferric chloride-induced platelet aggregation was almost lost in CLEC-2-depleted murine platelets and wild-type platelets preincubated with recombinant CLEC-2 proteins. Furthermore, coculture of wild-type platelets, but not CLEC-2-deficient platelets, attenuated ferroptosis of endothelial cells in vitro. CONCLUSION: Ferric chloride activates platelets via CLEC-2 and Src family kinase pathways, and platelets have a protective role in the ferroptosis of endothelial cells dependent on CLEC-2.


Asunto(s)
Plaquetas , Cloruros , Compuestos Férricos , Ferroptosis , Lectinas Tipo C , Ratones Endogámicos C57BL , Activación Plaquetaria , Agregación Plaquetaria , Transducción de Señal , Animales , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Compuestos Férricos/farmacología , Humanos , Activación Plaquetaria/efectos de los fármacos , Lectinas Tipo C/metabolismo , Cloruros/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Familia-src Quinasas/metabolismo , Fosforilación , Ratones Noqueados , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Ratones , Células Endoteliales de la Vena Umbilical Humana/metabolismo
15.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38731839

RESUMEN

CLEC4G, a glycan-binding receptor, has previously been demonstrated to inhibit Aß generation, yet its brain localization and functions in Alzheimer's disease (AD) are not clear. We explored the localization, function, and regulatory network of CLEC4G via experiments and analysis of RNA-seq databases. CLEC4G transcripts and proteins were identified in brain tissues, with the highest expression observed in neurons. Notably, AD was associated with reduced levels of CLEC4G transcripts. Bioinformatic analyses revealed interactions between CLEC4G and relevant genes such as BACE1, NPC1, PILRA, TYROBP, MGAT1, and MGAT3, all displaying a negative correlation trend. We further identified the upstream transcriptional regulators NR2F6 and XRCC4 for CLEC4G and confirmed a decrease in CLEC4G expression in APP/PS1 transgenic mice. This study highlights the role of CLEC4G in protecting against AD progression and the significance of CLEC4G for AD research and management.


Asunto(s)
Enfermedad de Alzheimer , Lectinas Tipo C , Ratones Transgénicos , Neuronas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Neuronas/metabolismo , Ratones , Humanos , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Encéfalo/metabolismo , Encéfalo/patología , Regulación de la Expresión Génica , Modelos Animales de Enfermedad
16.
Front Immunol ; 15: 1385696, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38770013

RESUMEN

Background: Recent studies have demonstrated a strong association between acute kidney injury (AKI) and chronic kidney disease (CKD), while the unresolved inflammation is believed to be a driving force for this chronic transition process. As a transmembrane pattern recognition receptor, Mincle (macrophage-inducible C-type lectin, Clec4e) was identified to participate in the early immune response after AKI. However, the impact of Mincle on the chronic transition of AKI remains largely unclear. Methods: We performed single-cell RNA sequencing (scRNA-seq) with the unilateral ischemia-reperfusion (UIR) murine model of AKI at days 1, 3, 14 and 28 after injury. Potential effects and mechanism of Mincle on renal inflammation and fibrosis were further validated in vivo utilizing Mincle knockout mice. Results: The dynamic expression of Mincle in macrophages and neutrophils throughout the transition from AKI to CKD was observed. For both cell types, Mincle expression was significantly up-regulated on day 1 following AKI, with a second rise observed on day 14. Notably, we identified distinct subclusters of Minclehigh neutrophils and Minclehigh macrophages that exhibited time-dependent influx with dual peaks characterized with remarkable pro-inflammatory and pro-fibrotic functions. Moreover, we identified that Minclehigh neutrophils represented an "aged" mature neutrophil subset derived from the "fresh" mature neutrophil cluster in kidney. Additionally, we observed a synergistic mechanism whereby Mincle-expressing macrophages and neutrophils sustained renal inflammation by tumor necrosis factor (TNF) production. Mincle-deficient mice exhibited reduced renal injury and fibrosis following AKI. Conclusion: The present findings have unveiled combined persistence of Minclehigh neutrophils and macrophages during AKI-to-CKD transition, contributing to unresolved inflammation followed by fibrosis via TNF-α as a central pro-inflammatory cytokine. Targeting Mincle may offer a novel therapeutic strategy for preventing the transition from AKI to CKD.


Asunto(s)
Lesión Renal Aguda , Modelos Animales de Enfermedad , Lectinas Tipo C , Macrófagos , Proteínas de la Membrana , Ratones Noqueados , Neutrófilos , Insuficiencia Renal Crónica , Animales , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Lesión Renal Aguda/etiología , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratones , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Masculino , Inflamación/inmunología , Ratones Endogámicos C57BL , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Fibrosis , Progresión de la Enfermedad
17.
Sci Rep ; 14(1): 10346, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710903

RESUMEN

Mammals are generally resistant to Mycobacterium avium complex (MAC) infections. We report here on a primary immunodeficiency disorder causing increased susceptibility to MAC infections in a canine breed. Adult Miniature Schnauzers developing progressive systemic MAC infections were related to a common founder, and pedigree analysis was consistent with an autosomal recessive trait. A genome-wide association study and homozygosity mapping using 8 infected, 9 non-infected relatives, and 160 control Miniature Schnauzers detected an associated region on chromosome 9. Whole genome sequencing of 2 MAC-infected dogs identified a codon deletion in the CARD9 gene (c.493_495del; p.Lys165del). Genotyping of Miniature Schnauzers revealed the presence of this mutant CARD9 allele worldwide, and all tested MAC-infected dogs were homozygous mutants. Peripheral blood mononuclear cells from a dog homozygous for the CARD9 variant exhibited a dysfunctional CARD9 protein with impaired TNF-α production upon stimulation with the fungal polysaccharide ß-glucan that activates the CARD9-coupled C-type lectin receptor, Dectin-1. While CARD9-deficient knockout mice are susceptible to experimental challenges by fungi and mycobacteria, Miniature Schnauzer dogs with systemic MAC susceptibility represent the first spontaneous animal model of CARD9 deficiency, which will help to further elucidate host defense mechanisms against mycobacteria and fungi and assess potential therapies for animals and humans.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Enfermedades de los Perros , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Complejo Mycobacterium avium , Infección por Mycobacterium avium-intracellulare , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Perros , Infección por Mycobacterium avium-intracellulare/veterinaria , Infección por Mycobacterium avium-intracellulare/genética , Infección por Mycobacterium avium-intracellulare/microbiología , Complejo Mycobacterium avium/genética , Enfermedades de los Perros/genética , Enfermedades de los Perros/microbiología , Eliminación de Secuencia , Linaje , Femenino , Masculino , Secuenciación Completa del Genoma , Homocigoto , Lectinas Tipo C/genética
18.
J Biomed Sci ; 31(1): 53, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38764023

RESUMEN

BACKGROUND: The C-type lectin family 18 (CLEC18) with lipid and glycan binding capabilities is important to metabolic regulation and innate immune responses against viral infection. However, human CLEC18 comprises three paralogous genes with highly similar sequences, making it challenging to distinguish genetic variations, expression patterns, and biological functions of individual CLEC18 paralogs. Additionally, the evolutionary relationship between human CLEC18 and its counterparts in other species remains unclear. METHODS: To identify the sequence variation and evolutionary divergence of human CLEC18 paralogs, we conducted a comprehensive analysis using various resources, including human and non-human primate reference genome assemblies, human pangenome assemblies, and long-read-based whole-genome and -transcriptome sequencing datasets. RESULTS: We uncovered paralogous sequence variants (PSVs) and polymorphic variants (PVs) of human CLEC18 proteins, and identified distinct signatures specific to each CLEC18 paralog. Furthermore, we unveiled a novel segmental duplication for human CLEC18A gene. By comparing CLEC18 across human and non-human primates, our research showed that the CLEC18 paralogy probably occurred in the common ancestor of human and closely related non-human primates, and the lipid-binding CAP/SCP/TAPS domain of CLEC18 is more diverse than its glycan-binding CTLD. Moreover, we found that certain amino acids alterations at variant positions are exclusive to human CLEC18 paralogs. CONCLUSIONS: Our findings offer a comprehensive profiling of the intricate variations and evolutionary characteristics of human CLEC18.


Asunto(s)
Evolución Molecular , Variación Genética , Lectinas Tipo C , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Animales , Primates/genética
19.
Front Immunol ; 15: 1372927, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38742105

RESUMEN

The parasitic helminth Schistosoma mansoni is a potent inducer of type 2 immune responses by stimulating dendritic cells (DCs) to prime T helper 2 (Th2) responses. We previously found that S. mansoni soluble egg antigens (SEA) promote the synthesis of Prostaglandin E2 (PGE2) by DCs through ERK-dependent signaling via Dectin-1 and Dectin-2 that subsequently induces OX40L expression, licensing them for Th2 priming, yet the ligands present in SEA involved in driving this response and whether specific targeting of PGE2 synthesis by DCs could affect Th2 polarization are unknown. We here show that the ability of SEA to bind Dectin-2 and drive ERK phosphorylation, PGE2 synthesis, OX40L expression, and Th2 polarization is impaired upon cleavage of high-mannose glycans by Endoglycosidase H treatment. This identifies high-mannose glycans present on glycoproteins in SEA as important drivers of this signaling axis. Moreover, we find that OX40L expression and Th2 induction are abrogated when microsomal prostaglandin E synthase-1 (mPGES) is selectively inhibited, but not when a general COX-1/2 inhibitor is used. This shows that the de novo synthesis of PGE2 is vital for the Th2 priming function of SEA-stimulated DCs as well as points to the potential existence of other COX-dependent lipid mediators that antagonize PGE2-driven Th2 polarization. Lastly, specific PGE2 inhibition following immunization with S. mansoni eggs dampened the egg-specific Th cell response. In summary, our findings provide new insights in the molecular mechanisms underpinning Th2 induction by S. mansoni and identify druggable targets for potential control of helminth driven-Th2 responses.


Asunto(s)
Antígenos Helmínticos , Células Dendríticas , Dinoprostona , Lectinas Tipo C , Manosa , Polisacáridos , Schistosoma mansoni , Células Th2 , Animales , Schistosoma mansoni/inmunología , Dinoprostona/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/inmunología , Manosa/metabolismo , Manosa/inmunología , Ratones , Polisacáridos/inmunología , Polisacáridos/metabolismo , Antígenos Helmínticos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Óvulo/inmunología , Óvulo/metabolismo , Ratones Endogámicos C57BL , Ligando OX40/metabolismo
20.
J Immunol Res ; 2024: 5582151, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690552

RESUMEN

Unlike T cells in other tissues, uterine T cells must balance strong immune defense against pathogens with tolerance to semiallogeneic fetus. Our previous study fully elucidated the characteristics of γδT cells in nonpregnant uterus and the mechanism modulated by estrogen. However, comprehensive knowledge of the immunological properties of αßT (including CD4+T cells and CD8+T) cells in nonpregnancy uterus has not been acquired. In this study, we fully compared the immunological properties of αßT cells between uterus and blood using mouse and human sample. It showed that most of CD4+T cells and CD8+T cells in murine uterus and human endometrium were tissue resident memory T cells which highly expressed tissue residence markers CD69 and/or CD103. In addition, both CD4+T cells and CD8+T cells in uterus highly expressed inhibitory molecular PD-1 and cytokine IFN-γ. Uterine CD4+T cells highly expressed IL-17 and modulated by transcription factor pSTAT3. Moreover, we compared the similarities and differences between human and murine uterine T cell phenotype. Together, uterine CD4+T cells and CD8+ cells exhibited a unique mixed signature of T cell dysfunction, activation, and effector function which enabled them to balance strong immune defense against pathogens with tolerance to fetus. Our study fully elucidated the unique immunologic properties of uterine CD4+T and CD8+T cells and provided a base for further investigation of functions.


Asunto(s)
Antígenos CD , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Útero , Femenino , Linfocitos T CD8-positivos/inmunología , Animales , Humanos , Ratones , Linfocitos T CD4-Positivos/inmunología , Útero/inmunología , Antígenos CD/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Cadenas alfa de Integrinas/metabolismo , Células T de Memoria/inmunología , Factor de Transcripción STAT3/metabolismo , Interferón gamma/metabolismo , Lectinas Tipo C/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Interleucina-17/metabolismo , Activación de Linfocitos/inmunología , Memoria Inmunológica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA