Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.238
Filter
1.
Rev Med Inst Mex Seguro Soc ; 62(1): 1-7, 2024 Jan 08.
Article in Spanish | MEDLINE | ID: mdl-39110839

ABSTRACT

Background: Primary breast tumors with neuroendocrine (NE) differentiation are a heterogeneous tumor group with diversity of biological behavior, with poorly defined prevalence and prognosis. Objective: To evaluate the chromogranin, synaptophysin, CD56, INSM1 markers expression prevalence and the association between NE differentiation and tumor molecular type. Material and methods: Observational, cross-sectional study which included 110 breast tissue samples with primary invasive carcinoma. Immunohistochemistry was performed for chromogranin, synaptophysin, CD56 and INMS1 markers. NE differentiation was considered with 10-90% positive cells, and NE tumor with > 90% positive cells. Results: 26.3% showed neuroendocrine differentiation. Out of these, 48.2% were luminal-A type, 24.1% luminal-B, 11.5% HER2neu, 17.2% triple-negative; 1.8% were NE tumors. Tumors were marker positive, and out of these to chromogranin in 24.5%, synaptophysin in 28.2%, CD56 in 2.7%, INSM1 in 16.4%. Synaptophysin was expressed in 17.3% luminal-A type, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negative. NE differentiation showed association with synaptophysin expression (r = 0.586, p = 0.0001). Conclusion: The NE differentiation prevalence was 26.3% in primary invasive breast cancers, with luminal-A molecular type predominance.


Introducción: los tumores primarios de mama con diferenciación neuroendócrina (NEBC por sus siglas en inglés) son un grupo heterogéneo de tumores con diversidad de comportamiento biológico, con prevalencia y pronóstico poco definido. Objetivo: evaluar la prevalencia de la expresión los marcadores cromogranina, sinaptofisina, CD56, INSM1 y la asociación entre la diferenciación neuroendócrina y el tipo molecular del tumor. Material y métodos: estudio observacional, transversal que incluyó 110 muestras de tejido mamario con carcinoma invasor primario. Se realizó inmunohistoquímica para los marcadores cromogranina, sinaptofisina, CD56 y INMS1. La presencia 10-90% de células positivas se consideró diferenciación neuroendócrina y tumor neuroendócrino con > 90% de células positivas. Resultados: el 26.3% mostró diferenciación neuroendócrina. De estos, 48.2% fueron tipo luminal-A, 24.1% luminal-B, 11.5% HER2neu y 17.2% triple-negativo; 1.8% resultaron tumores neuroendócrinos. Los tumores presentaron marcadores positivos y de estos, 24.5% fueron a cromogranina, 28.2% a sinaptofisina, 2.7% a CD56 y 16.4% a INSM1. La sinaptofisina se expresó en 17.3% del tipo luminal-A, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negativo. La diferenciación neuroendócrina mostró asociación con la expresión de sinaptofisina (r = 0.586, p = 0.0001). Conclusión: la prevalencia de la diferenciación neuroendócrina fue del 26.3% en los cánceres invasores primarios de mama, con predominio en el tipo molecular luminal-A.


Subject(s)
Biomarkers, Tumor , Synaptophysin , Humans , Female , Cross-Sectional Studies , Biomarkers, Tumor/metabolism , Middle Aged , Adult , Synaptophysin/metabolism , Aged , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD56 Antigen/metabolism , Immunohistochemistry , Repressor Proteins/metabolism , Chromogranins/metabolism , Receptor, ErbB-2/metabolism , Aged, 80 and over
2.
Am J Reprod Immunol ; 92(2): e13912, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39113660

ABSTRACT

PROBLEM: There is a higher incidence of irritable bowel syndrome with miscarriages, and recurrent miscarriages of otherwise normal embryos have been linked to subnormal expression of the immune checkpoint inhibitor CD200L. We sought to determine if alterations in the expression of the CD200 immune checkpoint inhibitor occur in colonic tissue in IBS-D patients. METHOD OF STUDY: Quantitative immunohistochemical staining of biopsies from proximal and distal colon or rectum for the inhibitory CD200L and CD200S molecules was done. CD56 cells were also enumerated as they play a role in recurrent miscarriages and may express CD200S. RESULTS: CD200L was decreased and CD200S was unchanged in epithelium but not stroma of 3 IBS-D cases. One case had an increase in both CD200L and CD200S. CD56 cells were also stained for CD200S. Degranulation was assessed by the percentage of extracellular CD200S that was increased as epithelial CD200L decreased. CONCLUSIONS: This pilot study was promising and warrants a larger sample to determine if a correlation between uterine implantation site CD200L and CD200S expression in normal and failing implantation sites is needed. Colonic epithelial CD200L may then provide useful information about the pathogenesis of the spontaneous miscarriage in individual cases.


Subject(s)
Abortion, Habitual , Antigens, CD , Diarrhea , Irritable Bowel Syndrome , Humans , Female , Irritable Bowel Syndrome/immunology , Irritable Bowel Syndrome/metabolism , Abortion, Habitual/immunology , Abortion, Habitual/metabolism , Antigens, CD/metabolism , Adult , Diarrhea/immunology , Pregnancy , Pilot Projects , Immune Tolerance , Signal Transduction , CD56 Antigen/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Colon/pathology , Colon/immunology , Colon/metabolism
3.
Medicine (Baltimore) ; 103(33): e39123, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151492

ABSTRACT

RATIONALE: Uterine tumors resembling ovarian sex cord tumors (UTROSCT) with rhabdoid features are uncommon mesenchymal neoplasms exhibiting diverse histological patterns, including significant rhabdoid morphology. A thorough comprehension of their clinicopathologic features is crucial for precise diagnosis and effective management. PATIENT CONCERNS: This study presents 4 cases of UTROSCT with rhabdoid features, diagnosed in patients aged 31 to 58. Varied recurrence patterns were observed, including similar recurrent lesions to the primary tumors with subsequent mortality, initial invasion and lymph node metastasis, and presence of only primary tumor. DIAGNOSES: Histopathological examination revealed diverse morphological patterns, prominently featuring rhabdoid differentiation. Immunohistochemical analysis showed expression of hormone receptors, sex cord, smooth muscle, and epithelial markers, notably WT1, CD56, and CD99. Molecular analysis identified ESR1-NCOA2 fusions and ESR1 and NCOA2/3 rearrangements, indicating a potential association between these genetic alterations and extensive rhabdoid differentiation. INTERVENTIONS: Various treatments were administered post-recurrence, including chemotherapy and targeted therapies. However, poor clinical outcomes were observed in all cases. OUTCOMES: Despite aggressive treatments, including chemotherapy and targeted therapies, poor clinical outcomes were observed, highlighting the aggressive nature of UTROSCT with significant rhabdoid differentiation. LESSONS: This case series emphasizes the importance of detailed pathological reporting, comprehensive molecular testing, and thorough tumor staging in UTROSCT cases with rhabdoid features. Enhanced understanding of the clinicopathologic characteristics of UTROSCT with rhabdoid differentiation is crucial for accurate diagnosis, prognostication, and management strategies.


Subject(s)
Ovarian Neoplasms , Sex Cord-Gonadal Stromal Tumors , Uterine Neoplasms , Humans , Female , Adult , Sex Cord-Gonadal Stromal Tumors/pathology , Sex Cord-Gonadal Stromal Tumors/genetics , Sex Cord-Gonadal Stromal Tumors/diagnosis , Middle Aged , Uterine Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/diagnosis , Diagnosis, Differential , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Rhabdoid Tumor/genetics , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/pathology , Nuclear Receptor Coactivator 2/genetics , CD56 Antigen/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Biomarkers, Tumor/genetics , 12E7 Antigen/genetics , 12E7 Antigen/metabolism , WT1 Proteins/genetics
4.
Clin Neuropathol ; 43(4): 113-126, 2024.
Article in English | MEDLINE | ID: mdl-39120072

ABSTRACT

Glioma is the most common brain tumor, accounting for a large majority of cancer-related deaths. ß-galactoside α2, 6 sialyltranferase 1 (ST6Gal1), the primary enzyme responsible for the conjugation of α2, 6 sialic acids to protein and lipid targets, is strongly associated with the occurrence and development of several brain tumor types. Still, the expression, targets, and functions of ST6Gal1 in glioma patients remain undetermined. As sialylation of the Ig-like cell adhesion family molecules have prominent roles in the latter's regulation in other biological contexts, we screened for members that have potential to be regulated by ST6Gal1 in silico and examined co-expressed protein modules using data derived from the Cancer Genome Atlas (TCGA) database, and we identified neural cell adhesion molecule (NCAM1) as a major ST6Gal1-interacting target. Bioinformatic binding analysis confirmed the interaction of ST6Gal1 and NCAM1. Immunohistochemistry was then used to evaluate post-operative samples from 156 patients with gliomas. ST6Gal1 and NCAM1 were co-expressed in gliomas, and their expression correlated significantly (p = 0.002) by univariate analysis. Our study also found that the expression levels of both ST6Gal1 and NCAM1 corresponded negatively with glioma grade, isocitrate dehydrogenase (IDH) mutation, and proliferation index (Ki67). Consistently, Kaplan-Meier survival curves showed that lower ST6Gal1 and NCAM1 protein levels are linked to unfavorable outcomes in glioma patients (p = 0.018 and p < 0.001, respectively). Our data indicate that ST6Gal1 may participate in the inhibition of oncogenesis and malignant progression via interacting with and targeting NCAM1 in glioma, thus presenting a novel strategy for intervention.


Subject(s)
Brain Neoplasms , Glioma , Sialyltransferases , Humans , Glioma/pathology , Glioma/genetics , Glioma/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Male , Female , Adult , Middle Aged , Antigens, CD/metabolism , CD56 Antigen/metabolism , Aged , Biomarkers, Tumor/metabolism , beta-D-Galactoside alpha 2-6-Sialyltransferase
5.
Sci Rep ; 14(1): 16580, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39020008

ABSTRACT

Vitiligo and halo nevus are immune-mediated skin diseases that have a similar pathogenesis and involve cellular cytotoxicity mechanisms that are not yet fully understood. In this study, we investigated the expression patterns of the cytolytic molecule granulysin (GNLY) in different cytotoxic cells in skin samples of vitiligo and halo nevus. Skin biopsies were taken from perilesional and lesional skin of ten vitiligo patients, eight patients with halo nevus and ten healthy controls. We analysed the expression of GNLY by immunohistochemistry in CD8+ and CD56+ NK cells. A significantly higher accumulation of GNLY+, CD8+ GNLY+ and fewer CD56+ GNLY+ cells was found in the lesional skin of vitiligo and halo nevus than in the healthy skin. These cells were localised in the basal epidermis and papillary dermis, suggesting that GNLY may be involved in the immune response against melanocytes. Similarly, but to a lesser extent, upregulation of GNLY+ and CD8+ GNLY+ cells was observed in the perilesional skin of vitiligo and halo nevus compared to healthy controls. In this study, we demonstrated for the first time an increased expression of CD8+ GNLY+ T lymphocytes and CD56+ GNLY+ NK cells in lesions of vitiligo and halo nevus, indicating the role of GNLY in the pathogenesis of both diseases.


Subject(s)
Antigens, Differentiation, T-Lymphocyte , Killer Cells, Natural , Nevus, Halo , Vitiligo , Humans , Vitiligo/metabolism , Vitiligo/pathology , Antigens, Differentiation, T-Lymphocyte/metabolism , Male , Nevus, Halo/metabolism , Nevus, Halo/pathology , Female , Adult , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Middle Aged , Skin/metabolism , Skin/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Melanocytes/metabolism , Melanocytes/pathology , Young Adult , CD56 Antigen/metabolism , Case-Control Studies
6.
Nat Immunol ; 25(8): 1445-1459, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956379

ABSTRACT

The functional diversity of natural killer (NK) cell repertoires stems from differentiation, homeostatic, receptor-ligand interactions and adaptive-like responses to viral infections. In the present study, we generated a single-cell transcriptional reference map of healthy human blood- and tissue-derived NK cells, with temporal resolution and fate-specific expression of gene-regulatory networks defining NK cell differentiation. Transfer learning facilitated incorporation of tumor-infiltrating NK cell transcriptomes (39 datasets, 7 solid tumors, 427 patients) into the reference map to analyze tumor microenvironment (TME)-induced perturbations. Of the six functionally distinct NK cell states identified, a dysfunctional stressed CD56bright state susceptible to TME-induced immunosuppression and a cytotoxic TME-resistant effector CD56dim state were commonly enriched across tumor types, the ratio of which was predictive of patient outcome in malignant melanoma and osteosarcoma. This resource may inform the design of new NK cell therapies and can be extended through transfer learning to interrogate new datasets from experimental perturbations or disease conditions.


Subject(s)
Killer Cells, Natural , Lymphocytes, Tumor-Infiltrating , Neoplasms , Transcriptome , Tumor Microenvironment , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Gene Expression Profiling , Single-Cell Analysis , Gene Regulatory Networks , CD56 Antigen/metabolism , Gene Expression Regulation, Neoplastic , Cell Differentiation
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000234

ABSTRACT

Juvenile Dermatomyositis (JDM) is the most common inflammatory myopathy in pediatrics. This study evaluates the role of Natural Killer (NK) cells in Juvenile Dermatomyositis (JDM) pathophysiology. The study included 133 untreated JDM children with an NK cell count evaluation before treatment. NK cell subsets (CD56low/dim vs. CD 56bright) were examined in 9 untreated children. CD56 and perforin were evaluated in situ in six untreated JDM and three orthopedic, pediatric controls. 56% of treatment-naive JDM had reduced circulating NK cell counts, designated "low NK cell". This low NK group had more active muscle disease compared to the normal NK cell group. The percentage of circulating CD56low/dim NK cells was significantly lower in the NK low group than in controls (0.55% vs. 4.6% p < 0.001). Examination of the untreated JDM diagnostic muscle biopsy documented an increased infiltration of CD56 and perforin-positive cells (p = 0.023, p = 0.038, respectively). Treatment-naive JDM with reduced circulating NK cell counts exhibited more muscle weakness and higher levels of serum muscle enzymes. Muscle biopsies from treatment-naive JDM displayed increased NK cell infiltration, with increased CD56 and perforin-positive cells.


Subject(s)
CD56 Antigen , Dermatomyositis , Killer Cells, Natural , Muscle Weakness , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Dermatomyositis/immunology , Dermatomyositis/blood , Dermatomyositis/pathology , Male , Child , Muscle Weakness/blood , Female , CD56 Antigen/metabolism , Child, Preschool , Perforin/metabolism , Adolescent , Lymphocyte Count
8.
Ann Hematol ; 103(9): 3723-3735, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38862793

ABSTRACT

Natural killer (NK) cells are equipped with anti-Epstein-Barr virus (EBV) function, however, whether EBV infection will affect NK cells reconstitution after allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains unclear. To identify the characteristics of NK cells, we prospectively enrolled 11 patients who occurred EBV reactivation post allo-HSCT and 11 patients without EBV infection as control. We found that that EBV infection induced the expansion of CD56bright and NKG2A+KIR- NK subsets,and decreased the cytotoxicity function of NK cells. The frequency of NKG2A+KIR- NK cells were higher in patients who progressed into post-transplant lymphoproliferative disorder (PTLD) than EBV viremia patients, which also correlated with decreased proliferation and cytotoxic function. By screening the activation receptors of NK cells, we found the DNAM-1+CD56bright NK cells is significantly increased after EBV stimulation, further we demonstrated that DNAM-1 is essential for EBV induced NK cells activation as the cytokine release against EBV-transformed lymphoblastoid cell lines(EBV-LCLs) of CD56bright NK cells were significantly decreased after DNAM-1 blockade. NK cells infusion suppressed the progression of EBV-related tumor mice model. A prospective cohort indicated that old donor age was an independent risk factor for EBV infection. Rapid CD56bri expansion and high expression of DNAM-1 on CD56bri NK cells in response to EBV reactivation correlated with rapid EBV clearance post allo-HSCT in patients with younger donors. In summary, our data showed that high expression of DNAM-1 receptors on NK cell may participate protective CD56bri NK cells response to EBV infection after allo-HSCT.


Subject(s)
CD56 Antigen , Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human , Killer Cells, Natural , Virus Activation , Humans , Killer Cells, Natural/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , CD56 Antigen/metabolism , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Female , Male , Adult , Middle Aged , Animals , Mice , Prospective Studies , Adolescent , Young Adult , Transplantation, Homologous/adverse effects , Allografts , Antigens, Differentiation, T-Lymphocyte
9.
Am J Dermatopathol ; 46(9): 572-580, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38842366

ABSTRACT

ABSTRACT: Abnormalities in the expression of cytokeratins or adhesion molecules have been associated with hair disorders. The expression patterns of these molecules in the hair follicles of developing human fetuses are not obvious. We aimed to investigate the expression patterns of some cytokeratins and adhesion molecules in the hair follicle of human fetuses and compared them with adults. Forty-eight fetuses of >16 gestational weeks and 22 adult cases with total excisions of benign nevi or cysts were enrolled. The skin samples were taken from both the scalp and back of the fetuses. The histopathologically normal skin areas were evaluated in adults. CK19, CK20, CAM5.2, high-molecular-weight cytokeratin, E-cadherin, ß-catenin, and CD56 immunohistochemical stainings were performed. In the fetus group, the staining scores declined in the third trimester but elevated and reached the highest level in adults, except for CD56, which did not stain any adult samples. All stainings were mostly observed in the outer root sheath, except CD56 that stained the perifollicular dermal sheath only in fetuses. E-cadherin, ß-catenin, and high-molecular-weight cytokeratin strongly and diffusely stained all adult samples. CAM5.2 and CK19 scores were correlated in fetuses (scalp scores: r s = 0.405, P = 0.004; back scores: r s = 0.422, P = 0.003) and adults (back scores: r s = 0.562, P = 0.046). CD56 negativity indicated the immune-privilege feature of adult hair follicles. As CK19, CAM5.2 may be used to find the regions of stem cells or transient amplifying cells.


Subject(s)
CD56 Antigen , Cadherins , Fetus , Hair Follicle , Immunohistochemistry , Keratins , beta Catenin , Humans , beta Catenin/metabolism , beta Catenin/analysis , Adult , Cadherins/metabolism , Cadherins/analysis , Keratins/analysis , Keratins/metabolism , Female , Hair Follicle/metabolism , CD56 Antigen/analysis , CD56 Antigen/metabolism , Fetus/metabolism , Male , Middle Aged , Young Adult , Antigens, CD/metabolism , Biomarkers/analysis , Biomarkers/metabolism
10.
Mol Immunol ; 172: 9-16, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850777

ABSTRACT

BACKGROUND: HIV/HCV coinfection is associated with a rapid progression to liver damage. Specifically, NK cell population dysregulation is of particular interest, as these cells have been shown to block HCV replication effectively and have an anti-fibrogenic activity. The NKp30 receptor is linked to tumor cell lysis and has a crucial role during viral infections. In the present study, we determined the subpopulations of NK cells based on CD56 and CD16 expression, NKp30 receptor expression, its isoforms A, B, and C, along with the cytotoxicity molecules in patients with HIV/HCV. RESULTS: evidenced by the APRI and FIB-4 indices, the HCV-infected patients presented greater liver damage than the HIV and HIV/HCV groups. The HCV group presented a decreased expression of NKp30 isoform A, and NK cell frequency was not different between groups; however, CD56brigth subpopulation, NKp30 receptor, and CD247 adaptor chain were decreased in HIV/HCV patients; further, we described increased levels of soluble IL-8, IL-10, IL-12, and IL-23 in the serum of HIV/HCV patients. CONCLUSIONS: HCV and HIV/HCV patients have multiple parameters of non-fitness status in NK cells; awareness of these dysfunctional immunological parameters in HIV/HCV and HCV patients can elucidate possible novel therapeutics directed towards the improvement of NK cell fitness status, in order to improve their function against liver damage.


Subject(s)
Coinfection , HIV Infections , Hepatitis C , Killer Cells, Natural , Natural Cytotoxicity Triggering Receptor 3 , Protein Isoforms , Humans , Natural Cytotoxicity Triggering Receptor 3/metabolism , Natural Cytotoxicity Triggering Receptor 3/immunology , Killer Cells, Natural/immunology , HIV Infections/immunology , Male , Coinfection/immunology , Female , Adult , Middle Aged , Protein Isoforms/immunology , Hepatitis C/immunology , Hepacivirus/immunology , CD56 Antigen/metabolism , CD56 Antigen/immunology , Receptors, IgG/metabolism , Receptors, IgG/immunology
11.
Sci Rep ; 14(1): 13133, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849432

ABSTRACT

The short-lived nature and heterogeneity of Natural Killer (NK) cells limit the development of NK cell-based therapies, despite their proven safety and efficacy against cancer. Here, we describe the biological basis, detailed phenotype and function of long-lived anti-tumour human NK cells (CD56highCD16+), obtained without cell sorting or feeder cells, after priming of peripheral blood cells with Bacillus Calmette-Guérin (BCG). Further, we demonstrate that survival doses of a cytokine combination, excluding IL18, administered just weekly to BCG-primed NK cells avoids innate lymphocyte exhaustion and leads to specific long-term proliferation of innate cells that exert potent cytotoxic function against a broad range of solid tumours, mainly through NKG2D. Strikingly, a NKG2C+CD57-FcεRIγ+ NK cell population expands after BCG and cytokine stimulation, independently of HCMV serology. This strategy was exploited to rescue anti-tumour NK cells even from the suppressor environment of cancer patients' bone marrow, demonstrating that BCG confers durable anti-tumour features to NK cells.


Subject(s)
Cell Proliferation , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Humans , Cell Proliferation/drug effects , Neoplasms/immunology , Neoplasms/drug therapy , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , Mycobacterium bovis/immunology , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Interleukins/metabolism , CD56 Antigen/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism
12.
Nat Immunol ; 25(7): 1183-1192, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38872000

ABSTRACT

Natural killer (NK) cells function by eliminating virus-infected or tumor cells. Here we identified an NK-lineage-biased progenitor population, referred to as early NK progenitors (ENKPs), which developed into NK cells independently of common precursors for innate lymphoid cells (ILCPs). ENKP-derived NK cells (ENKP_NK cells) and ILCP-derived NK cells (ILCP_NK cells) were transcriptionally different. We devised combinations of surface markers that identified highly enriched ENKP_NK and ILCP_NK cell populations in wild-type mice. Furthermore, Ly49H+ NK cells that responded to mouse cytomegalovirus infection primarily developed from ENKPs, whereas ILCP_NK cells were better IFNγ producers after infection with Salmonella and herpes simplex virus. Human CD56dim and CD56bright NK cells were transcriptionally similar to ENKP_NK cells and ILCP_NK cells, respectively. Our findings establish the existence of two pathways of NK cell development that generate functionally distinct NK cell subsets in mice and further suggest these pathways may be conserved in humans.


Subject(s)
Cell Differentiation , Killer Cells, Natural , Killer Cells, Natural/immunology , Animals , Mice , Humans , Cell Differentiation/immunology , Mice, Inbred C57BL , Immunity, Innate , CD56 Antigen/metabolism , Muromegalovirus/immunology , Cell Lineage/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice, Knockout , Cells, Cultured
13.
PLoS Pathog ; 20(6): e1012315, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38889192

ABSTRACT

Invasive aspergillosis causes significant morbidity and mortality in immunocompromised patients. Natural killer (NK) cells are pivotal for antifungal defense. Thus far, CD56 is the only known pathogen recognition receptor on NK cells triggering potent antifungal activity against Aspergillus fumigatus. However, the underlying cellular mechanisms and the fungal ligand of CD56 have remained unknown. Using purified cell wall components, biochemical treatments, and ger mutants with altered cell wall composition, we herein found that CD56 interacts with the A. fumigatus cell wall carbohydrate galactosaminogalactan (GAG). This interaction induced NK-cell activation, degranulation, and secretion of immune-enhancing chemokines and cytotoxic effectors. Supernatants from GAG-stimulated NK cells elicited antifungal activity and enhanced antifungal effector responses of polymorphonuclear cells. In conclusion, we identified A. fumigatus GAG as a ligand of CD56 on human primary NK cells, stimulating potent antifungal effector responses and activating other immune cells.


Subject(s)
Aspergillosis , Aspergillus fumigatus , CD56 Antigen , Killer Cells, Natural , Humans , Aspergillus fumigatus/immunology , Killer Cells, Natural/immunology , CD56 Antigen/metabolism , CD56 Antigen/immunology , Aspergillosis/immunology , Aspergillosis/microbiology , Lymphocyte Activation/immunology , Polysaccharides/metabolism , Polysaccharides/immunology , Cell Wall/immunology , Cell Wall/metabolism
14.
J Nanobiotechnology ; 22(1): 313, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840120

ABSTRACT

Adoptive cellular immunotherapy as a promising and alternative cancer therapy platform is critical for future clinical applications. Natural killer (NK) cells have attracted attention as an important type of innate immune regulatory cells that can rapidly kill multiple adjacent cancer cells. However, these cells are significantly less effective in treating solid tumors than in treating hematological tumors. Herein, we report the synthesis of a Fe3O4-PEG-CD56/Avastin@Ce6 nanoprobe labeled with NK-92 cells that can be used for adoptive cellular immunotherapy, photodynamic therapy and dual-modality imaging-based in vivo fate tracking. The labeled NK-92 cells specifically target the tumor cells, which increases the amount of cancer cell apoptosis in vitro. Furthermore, the in vivo results indicate that the labeled NK-92 cells can be used for tumor magnetic resonance imaging and fluorescence imaging, adoptive cellular immunotherapy, and photodynamic therapy after tail vein injection. These data show that the developed multifunctional nanostructure is a promising platform for efficient innate immunotherapy, photodynamic treatment and noninvasive therapeutic evaluation of breast cancer.


Subject(s)
Breast Neoplasms , CD56 Antigen , Killer Cells, Natural , Photochemotherapy , Polyethylene Glycols , Breast Neoplasms/therapy , Humans , Female , Animals , Photochemotherapy/methods , Mice , Polyethylene Glycols/chemistry , Cell Line, Tumor , CD56 Antigen/metabolism , Immunotherapy, Adoptive/methods , Apoptosis/drug effects , Magnetic Resonance Imaging/methods , Mice, Inbred BALB C , Mice, Nude
15.
Eur J Immunol ; 54(8): e2451046, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38778501

ABSTRACT

Chronic hepatitis B (CHB) virus infection, which can be divided into immune-tolerant (IT), immune-active (IA), inactive carrier (IC) phases, and HBeAg-negative hepatitis (ENEG), can induce liver cirrhosis and eventually hepatocellular carcinoma (HCC). CD3+CD56+ NKT-like cells play an important role in antiviral immune response. However, the mechanism of NKT-like cells to mediate immune tolerance remains largely elusive. In this study, we observed circulating NKT-like cells from IC and IT CHB patients were phenotypically and functionally impaired, manifested by increased expression of inhibitory receptor TIGIT and decreased capacity of secreting antiviral cytokines. Besides, TIGIT+ NKT-like cells of IC and IT CHB patients expressed lower levels of cytotoxic cytokines than the TIGIT- subset. Furthermore, increased expression of CD155, the ligand of TIGIT, on plasmacytoid dendritic cells (pDCs) was detected in IC and IT CHB patients. Importantly, the co-culture of NKT-like cells and pDCs showed that NKT-like cells restored their antiviral ability after TIGIT blockade upon HBV peptide stimulation in IC and IT CHB patients. In conclusion, our findings suggest that the TIGIT pathway may mediate immune tolerance in IT CHB patients and lead to functional impairment in IC patients, indicating that TIGIT may be a potential therapeutic checkpoint for immunotherapy of CHB patients.


Subject(s)
CD3 Complex , CD56 Antigen , Dendritic Cells , Hepatitis B virus , Hepatitis B, Chronic , Immune Tolerance , Natural Killer T-Cells , Receptors, Immunologic , Humans , Receptors, Immunologic/metabolism , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , CD56 Antigen/metabolism , Male , Hepatitis B virus/immunology , Female , Natural Killer T-Cells/immunology , Adult , Dendritic Cells/immunology , Immune Tolerance/immunology , CD3 Complex/immunology , CD3 Complex/metabolism , Middle Aged , Receptors, Virus/metabolism , Receptors, Virus/immunology , Cytokines/metabolism , Cytokines/immunology
16.
J Immunol ; 213(1): 40-51, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38809096

ABSTRACT

NK cells are innate immune effectors that kill virally infected or malignant cells. NK cell deficiency (NKD) occurs when NK cell development or function is impaired and variants in MCM4, GINS1, MCM10, and GINS4 result in NKD. Although NK cells are strongly impacted by mutational deficiencies in helicase proteins, the mechanisms underlying this specific susceptibility are poorly understood. In this study, we induced replication stress in activated NK cells or T cells by chemical and genetic methods. We found that the CD56bright subset of NK cells accumulates more DNA damage and replication stress during activation than do CD56dim NK cells or T cells. Aphidicolin treatment increases apoptosis of CD56bright NK cells through increased pan-caspase expression and decreases perforin expression in surviving cells. These findings show that sensitivity to replication stress affects NK cell survival and function and contributes to NKD.


Subject(s)
Apoptosis , Killer Cells, Natural , Lymphocyte Activation , Humans , Killer Cells, Natural/immunology , Apoptosis/immunology , Lymphocyte Activation/immunology , DNA Damage , DNA Replication , CD56 Antigen/metabolism , Stress, Physiological/immunology , T-Lymphocytes/immunology , Cells, Cultured
17.
J Cutan Pathol ; 51(8): 614-617, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38725374

ABSTRACT

Mycosis fungoides (MF) represents the most common type of primary cutaneous T-cell lymphoma. Recognition of MF variants with divergent immunophenotypes is important for accurate diagnosis and appropriate management, as they can be confused with other lymphoma subtypes. We present a case of a 49-year-old male previously diagnosed with a cutaneous lymphoproliferative disorder with an unusual NK/T-cell phenotype. He presented with a 10-year history of pelvic girdle rash involving the right hip and upper thigh. The lesions were characterized as atrophic patches concentrated in sun-protected areas and involving 10% of the body surface area. Shave biopsies revealed an atypical epidermotropic infiltrate composed of hyperchromatic small to medium-sized lymphocytes with perinuclear halos and "tagging" along the dermal-epidermal junction. The immunophenotype was unusual in that the neoplastic lymphocytes showed complete loss of pan T-cell antigens along with expression of CD56, cytotoxic markers, and weak CD20. All other B-cell markers were negative. The combination of clinical findings, in addition to the histopathologic and immunophenotypic profile, were diagnostic of null T-cell phenotype MF with aberrant expression of CD56 and CD20. Null T-cell phenotype MF is very uncommon, can be diagnostically challenging, and can mislead the diagnosis of aggressive lymphoma subtypes.


Subject(s)
Antigens, CD20 , CD56 Antigen , Mycosis Fungoides , Skin Neoplasms , Humans , Mycosis Fungoides/pathology , Mycosis Fungoides/diagnosis , Mycosis Fungoides/metabolism , Male , Middle Aged , CD56 Antigen/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Antigens, CD20/metabolism , Immunophenotyping/methods , Phenotype , T-Lymphocytes/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Diagnosis, Differential , Biomarkers, Tumor/metabolism
18.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731828

ABSTRACT

HLA-DR-positive NK cells, found in both healthy individuals and patients with different inflammatory diseases, are characterized as activated cells. However, data on their capacity for IFNγ production or cytotoxic response vary between studies. Thus, more precise investigation is needed of the mechanisms related to the induction of HLA-DR expression in NK cells, their associations with NK cell differentiation stage, and functional or metabolic state. In this work, HLA-DR-expressing NK cell subsets were investigated using transcriptomic analysis, metabolic activity assays, and analysis of intercellular signaling cascades. We demonstrated that HLA-DR+CD56bright NK cells were characterized by a proliferative phenotype, while HLA-DR+CD56dim NK cells exhibited features of adaptive cells and loss of inhibitory receptors with increased expression of MHC class II trans-activator CIITA. The activated state of HLA-DR-expressing NK cells was confirmed by higher levels of ATP and mitochondrial mass observed in this subset compared to HLA-DR- cells, both ex vivo and after stimulation in culture. We showed that HLA-DR expression in NK cells in vitro can be induced both through stimulation by exogenous IL-2 and IL-21, as well as through auto-stimulation by NK-cell-produced IFNγ. At the intracellular level, HLA-DR expression depended on the activation of STAT3- and ERK1/2-mediated pathways, with subsequent activation of isoform 3 of the transcription factor CIITA. The obtained results broaden the knowledge about HLA-DR-positive NK cell appearance, diversity, and functions, which might be useful in terms of understanding the role of this subset in innate immunity and assessing their possible implications in NK cell-based therapy.


Subject(s)
Cell Differentiation , HLA-DR Antigens , Interferon-gamma , Killer Cells, Natural , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Humans , HLA-DR Antigens/metabolism , HLA-DR Antigens/genetics , Interferon-gamma/metabolism , CD56 Antigen/metabolism , Lymphocyte Activation/immunology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Cells, Cultured , Nuclear Proteins , Trans-Activators
19.
Sci Rep ; 14(1): 12188, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806640

ABSTRACT

Natural killer (NK) cells are closely associated with malignant tumor progression and metastasis. However, studies on their relevance in colorectal cancer (CRC) are limited. We aimed to comprehensively analyze the absolute counts, phenotypes, and function of circulating NK cells in patients with CRC using multiparametric flow cytometry. The distribution of NK cell subsets in the peripheral circulation of patients with CRC was significantly altered relative to the control group. This is shown by the decreased frequency and absolute count of CD56dimCD16+ NK cells with antitumor effects, contrary to the increased frequency of CD56bright NK and CD56dimCD16- NK cells with poor or ineffective antitumor effects. NK cells in patients with CRC were functionally impaired, with decreased intracellular interferon (IFN)-γ secretion and a significantly lower percentage of cell surface granzyme B and perforin expression. In addition, IFN-γ expression decreased significantly with the tumor stage progression. Based on a comprehensive analysis of the absolute counts, phenotypes, and functional markers of NK cells, we found an altered subset distribution and impaired function of circulating NK cells in patients with CRC.


Subject(s)
Colorectal Neoplasms , Granzymes , Interferon-gamma , Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/blood , Male , Female , Middle Aged , Interferon-gamma/metabolism , Aged , Granzymes/metabolism , Perforin/metabolism , CD56 Antigen/metabolism , Flow Cytometry , Adult
20.
Cell Calcium ; 121: 102894, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728789

ABSTRACT

TRPV2 voltage-insensitive, calcium-permeable ion channels play important roles in cancer progression, immune response, and neuronal development. Despite TRPV2's physiological impact, underlying endogenous proteins mediating TRPV2 responses and affected signaling pathways remain elusive. Using quantitative peroxidase-catalyzed (APEX2) proximity proteomics we uncover dynamic changes in the TRPV2-proximal proteome and identify calcium signaling and cell adhesion factors recruited to the molecular channel neighborhood in response to activation. Quantitative TRPV2 proximity proteomics further revealed activation-induced enrichment of protein clusters with biological functions in neural and cellular projection. We demonstrate a functional connection between TRPV2 and the neural immunoglobulin cell adhesion molecules NCAM and L1CAM. NCAM and L1CAM stimulation robustly induces TRPV2 [Ca2+]I flux in neuronal PC12 cells and this TRPV2-specific [Ca2+]I flux requires activation of the protein kinase PKCα. TRPV2 expression directly impacts neurite lengths that are modulated by NCAM or L1CAM stimulation. Hence, TRPV2's calcium signaling plays a previously undescribed, yet vital role in cell adhesion, and TRPV2 calcium flux and neurite development are intricately linked via NCAM and L1CAM cell adhesion proteins.


Subject(s)
Calcium , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules , Neuronal Outgrowth , Proteome , TRPV Cation Channels , Animals , Humans , Rats , Calcium/metabolism , Calcium Signaling , Cell Adhesion , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , PC12 Cells , Protein Kinase C-alpha/metabolism , Proteome/metabolism , TRPV Cation Channels/metabolism , CD56 Antigen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL