Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.189
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Front Immunol ; 15: 1427810, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39351227

RESUMEN

It has been demonstrated previously that human leukocyte antigen class I (HLA-I) and class II (HLA-II) alleles may modulate JAK2 V617F and CALR mutation (CALRmut)-associated oncogenesis in myeloproliferative neoplasms (MPNs). However, the role of immunogenetic factors in MPNs remains underexplored. We aimed to investigate the potential involvement of HLA genes in CALRmut+ MPNs. High-resolution genotyping of HLA-I and -II loci was conducted in 42 CALRmut+ and 158 JAK2 V617F+ MPN patients and 1,083 healthy controls. A global analysis of the diversity of HLA-I genotypes revealed no significant differences between CALRmut+ patients and controls. However, one HLA-I allele (C*06:02) showed an inverse correlation with presence of CALR mutation. A meta-analysis across independent cohorts and healthy individuals from the 1000 Genomes Project confirmed an inverse correlation between the presentation capabilities of the HLA-I loci for JAK2 V617F and CALRmut-derived peptides in both patients and healthy individuals. scRNA-Seq analysis revealed low expression of TAP1 and CIITA genes in CALRmut+ hematopoietic stem and progenitor cells. In conclusion, the HLA-I genotype differentially restricts JAK2 V617F and CALRmut-driven oncogenesis potentially explaining the mutual exclusivity of the two mutations and differences in their presentation latency. These findings have practical implications for the development of neoantigen-based vaccines in MPNs.


Asunto(s)
Calreticulina , Genotipo , Janus Quinasa 2 , Mutación , Trastornos Mieloproliferativos , Humanos , Janus Quinasa 2/genética , Calreticulina/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Adulto , Carcinogénesis/genética , Alelos , Antígenos de Histocompatibilidad Clase I/genética , Anciano de 80 o más Años
2.
Pathologica ; 116(4): 242-248, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39377506

RESUMEN

Breast implant-associated (BIA) lymphoma is a rare malignancy, typically originating from T-cells; however, few cases of diffuse large B-cell lymphoma (LBCL) have been recently described. These cases share major features: Epstein-Barr virus positivity and a favorable prognosis with surgical intervention alone, hinting at a potential link to fibrin-associated LBCL (FA-LBCL). This study presents the first case of BIA-FA-LBCL in Italy and one of the few assessed from a molecular standpoint so far. We identified two pathogenic mutations in DNMT3A and a variant of uncertain significance (VUS) in JAK2. These findings suggest that dysfunctional epigenetic mechanisms and constitutive activation of the JAK-STAT pathway may underpin BIA-FA-LBCL lymphomagenesis. Finally, we summarized all the previously reported cases in alignment with the updated WHO-HAEM5 classification, shedding further light on the nature of this new entity. This report highlights the rarity of BIA-FA-LBCL and underscores the importance of comprehensive capsule sampling and reporting to national databases for accurate characterization and management of these lymphomas. The study supports the classification of BIA-FA-LBCL within the spectrum of FA-LBCL, emphasizing the need for further research to elucidate its molecular underpinnings and improve clinical outcomes.


Asunto(s)
Implantes de Mama , Fibrina , Linfoma de Células B Grandes Difuso , Humanos , Femenino , Implantes de Mama/efectos adversos , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/virología , Fibrina/metabolismo , Fibrina/análisis , Janus Quinasa 2/genética , Mutación , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/diagnóstico , Persona de Mediana Edad , ADN Metiltransferasa 3A
3.
Clin Transl Med ; 14(10): e70040, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39358917

RESUMEN

BACKGROUND AND AIMS: Myofibroblasts, the primary producers of extracellular matrix, primarily originate from hepatic stellate cells (HSCs), and their activation plays a pivotal role in liver fibrosis. This study aimed to investigate the function of CXC motif ligand 14 (CXCL14) in the progression of liver fibrosis. APPROACH AND RESULTS: CXCL14 knockdown significantly reduced the extent of liver fibrosis. Using Ingenuity pathway analysis and qRT-PCR, activating transcription factor 3 (ATF3) was identified as a key upstream regulator of CXCL14 expression. Mechanistically, ATF3 was shown to bind to the CXCL14 promoter, promoting its transactivation by TGF-ß in HSCs. Notably, both global CXCL14 deletion (CXCL14-/-) and HSC/myofibroblast-specific CXCL14 knockdown significantly attenuated liver fibrosis in mice. RNA-seq comparisons between CXCL14-/- and WT mice highlighted Jak2 as the most significantly downregulated gene, implicating its role in the antifibrotic effects of CXCL14 suppression on HSC inactivation. Moreover, Jak2 overexpression reversed the inhibition of liver fibrosis caused by CXCL14 knockdown in vivo. CONCLUSIONS: These findings unveil a novel ATF3/CXCL14/Jak2 signalling axis in liver fibrosis, presenting potential therapeutic targets for the disease.


Asunto(s)
Factor de Transcripción Activador 3 , Quimiocinas CXC , Células Estrelladas Hepáticas , Cirrosis Hepática , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Animales , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Masculino , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Humanos , Activación Transcripcional/genética , Ratones Endogámicos C57BL
4.
Int J Mol Sci ; 25(20)2024 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-39457093

RESUMEN

A high-altitude, low-pressure hypoxic environment has severe effects on the health and work efficiency of its residents, and inadequate preventive measures and adaptive training may lead to the occurrence of AMS. Acute exposure to hypoxia conditions can have a less-favorable physiological effect on the human immune system. However, the regulation of the immune system in high-altitude environments is extremely complex and remains elusive. This study integrated system bioinformatics methods to screen for changes in immune cell subtypes and their associated targets. It also sought potential therapeutically effective natural compound candidates. The present study observed that monocytes, M1 macrophages and NK cells play a crucial role in the inflammatory response in AMS. IL15RA, CD5, TNFSF13B, IL21R, JAK2 and CXCR3 were identified as hub genes, and JAK2 was positively correlated with monocytes; TNFSF13B was positively correlated with NK cells. The natural compound monomers of jasminoidin and isoliquiritigenin exhibited good binding affinity with JAK2, while dicumarol and artemotil exhibited good binding affinity with TNFSF13B, and all are expected to become a potential therapeutic agents.


Asunto(s)
Mal de Altura , Biomarcadores , Simulación del Acoplamiento Molecular , Humanos , Mal de Altura/genética , Mal de Altura/tratamiento farmacológico , Mal de Altura/sangre , Perfilación de la Expresión Génica/métodos , Transcriptoma , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Inflamación/sangre , Inflamación/genética
5.
Endocrinology ; 165(11)2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39378329

RESUMEN

In rodents, loss of growth hormone (GH) or its receptor is associated with extended lifespan. We aimed to determine the signaling process resulting in this longevity using GH receptor (GHR)-mutant mice with key signaling pathways deleted and correlate this with cancer incidence and expression of genes associated with longevity. GHR uses both canonical janus kinase (JAK)2-signal transducer and activator of transcription (STAT) signaling as well as signaling via the LYN-ERK1/2 pathway. We used C57BL/6 mice with loss of key receptor tyrosines and truncation resulting in 1) loss of most STAT5 response to GH; 2) total inability to generate STAT5 to GH; 3) loss of Box1 to prevent activation of JAK2 but not LYN kinase; or 4) total knockout of the receptor. For each mutant we analyzed lifespan, histopathology to determine likely cause of death, and hepatic gene and protein expression. The extended lifespan is evident in the Box1-mutant males (retains Lyn activation), which have a median lifespan of 1016 days compared to 890 days for the Ghr-/- males. In the females, GhrBox1-/- mice have a median lifespan of 970 days compared to 911 days for the knockout females. Sexually dimorphic GHR-STAT5 is repressive for longevity, since its removal results in a median lifespan of 1003 days in females compared to 734 days for wild-type females. Numerous transcripts related to insulin sensitivity, oxidative stress response, and mitochondrial function are regulated by GHR-STAT5; however, LYN-responsive genes involve DNA repair, cell cycle control, and anti-inflammatory response. There appears to be a yin-yang relationship between JAK2 and LYN that determines lifespan.


Asunto(s)
Hormona del Crecimiento , Janus Quinasa 2 , Longevidad , Factor de Transcripción STAT5 , Transducción de Señal , Familia-src Quinasas , Animales , Femenino , Masculino , Ratones , Hormona del Crecimiento/metabolismo , Incidencia , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Longevidad/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/metabolismo , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Transducción de Señal/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT5/genética
6.
Georgian Med News ; (352-353): 101-105, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39441278

RESUMEN

A high level of hemoglobin and hematocrit is one of the most important laboratory indicators of polycythemia vera (PV). Nevertheless, in some cases, these indicators may be normal or below the norm. This form of the disease is called latent or masked PV (LPV). It has been found that thrombohemorrhagic complications (THC) are more common in LPV patients than in classic PV (CPV) patients. The relationship between JAK2 gene mutation allele burden, thrombocytosis, leukocytosis level and the occurrence of thrombosis during LPV was analysed in different studies. The results some of them were conflicted. It is also possible that this situation occurs due to the delay in diagnosis and treatment of patients with LPV. AIM: investigate the laboratory and clinical features of the latent form of PV. MATERIALS AND METHODS: An analysis of PV patients registered in 2019-2020 was conducted. Out of them patients with LPV were distinguished. During diagnosis of the disease, general blood analysis, trepanobiopsy and histological examination of bone marrow, molecular genetic examination of peripheral blood and bone marrow were performed. All numerical indicators obtained in the course of the research were statistically analyzed taking according to the modern recommendations. Indicators in the groups were arranged in the order of variation, and the average indicator, standard error of this indicator, confidence interval for the 95% confidence level (CI-confidence interval) were calculated for each order. For comparing the groups, the integrity criterion p was taken into account. Calculations were performed using ONE-WAY-ANOVA calculation software. RESULTS: We study 101 patients. Out of them 36 patients with latent polycythemia were identified. The clinical and laboratory parameters of patients with LPV and CPV were compared. In latent PV the complaints of patients were less intense; the size of the spleen was smaller; thrombotic complications were more often; hemoglobin, hematocrit, erythrocytes count was lower; the number of platelets was higher; leukocytes count and JAK2М617F gene allel burden were not statistically different. The most part of LPV patients, in contrast to CPV patients, was in a high-risk group of THC. CONCLUSION: According to the obtained results, it can be concluded that timely and correct diagnosis of LPV is very important. Despite the fact that disease passes in a latent, masked form, THC are more likely to occur. This can be attributed to the high platelet count in the blood and the lack of timely treatment of the disease.


Asunto(s)
Janus Quinasa 2 , Policitemia Vera , Humanos , Policitemia Vera/sangre , Policitemia Vera/genética , Policitemia Vera/diagnóstico , Policitemia Vera/patología , Janus Quinasa 2/genética , Masculino , Persona de Mediana Edad , Femenino , Hemoglobinas/análisis , Hematócrito , Mutación , Anciano , Adulto , Trombosis/sangre , Trombosis/genética , Trombosis/patología , Trombosis/diagnóstico
8.
J Clin Invest ; 134(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352379

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) has emerged as a previously unrecognized, potent, age-related, and common risk factor for atherosclerosis. Somatic mutations in certain known leukemia driver genes give rise to clones of mutant cells in peripheral blood. The increased risk of developing hematologic malignancy does not, on its own, explain excess mortality in individuals with CHIP. Cardiovascular disease accounts for much of this gap. Experimental evidence supports the causality of certain CHIP mutations in accelerated atherosclerosis. CHIP due to mutations in different driver genes varies in their promotion of atherosclerotic events and in the region of augmented atherosclerotic involvement. For example, CHIP due to mutations in DNMT3a appears less atherogenic than CHIP that arises from TET2 or JAK2, forms of CHIP that incite inflammation. The recognition of certain CHIP mutations as promoters of atherosclerotic risk has opened new insights into understanding of the pathophysiology of this disease. The accentuated cardiovascular risk and involvement of distinct pathways of various forms of CHIP also inform novel approaches to allocation of targeted therapies, affording a step toward personalized medicine.


Asunto(s)
Aterosclerosis , Hematopoyesis Clonal , ADN Metiltransferasa 3A , Dioxigenasas , Mutación , Humanos , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Hematopoyesis Clonal/genética , ADN Metiltransferasa 3A/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Animales , Janus Quinasa 2/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Riesgo
9.
BMC Neurol ; 24(1): 386, 2024 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-39395952

RESUMEN

BACKGROUND: Cerebral venous sinus thrombosis (CVST) is a rare but potentially life-threatening subtype of stroke. Prompt and appropriate anticoagulation is crucial for improving the prognosis of CVST and preventing its recurrence. Identifying the underlying cause of CVST is decisive for guiding anticoagulant selection and determining treatment duration. CASE PRESENTATION: A 50-year-old man presented with a 35-day history of headache, nausea, vomiting, and blurred vision. Digital subtraction angiography performed at another facility revealed CVST. A contrast-enhanced black-blood MRI at our center confirmed the diagnosis, which was supported by a high intracranial pressure of 330mmH2O. Laboratory tests showed elevated leukocytes and platelet counts, raising suspicion of an underlying myeloproliferative neoplasms (MPNs). A bone marrow biopsy demonstrated increased megakaryocytes and granulocytes, and genetic testing identified the presence of the Janus kinase 2 V617F (JAK2 V617F) mutation, leading to a diagnosis of pre-primary myelofibrosis (pre-PMF). During hospitalization, anticoagulation with nadroparin calcium and fibrinolytic therapy were initiated. Upon discharge, rivaroxaban and aspirin were prescribed to prevent CVST recurrence and arterial thrombosis. CONCLUSION: This case highlights the importance of recognizing dynamic changes in routine blood tests that may link CVST to underlying hematological disorders. The JAK2 mutation is not only associated with MPNs but also increases the risk of thrombosis, including CVST. Further investigation is warranted to better understand the mechanisms by which JAK2 mutations contribute to thrombosis and to explore the potential benefits of JAK2 inhibitors in reducing this risk.


Asunto(s)
Janus Quinasa 2 , Mielofibrosis Primaria , Trombosis de los Senos Intracraneales , Humanos , Masculino , Janus Quinasa 2/genética , Persona de Mediana Edad , Trombosis de los Senos Intracraneales/genética , Trombosis de los Senos Intracraneales/tratamiento farmacológico , Trombosis de los Senos Intracraneales/diagnóstico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/tratamiento farmacológico , Mutación
10.
J Cell Mol Med ; 28(20): e70138, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39431551

RESUMEN

Myeloproliferative neoplasms (MPNs) are characterized by an increased production of blood cells due to the acquisition of mutations such as JAK2V617F. TGF-ß, whose secretion is increased in MPN patients, is known to negatively regulate haematopoietic stem cell (HSC) proliferation. Using an isogenic JAK2V617F or JAK2 wild-type UT-7 cell line we observed that JAK2V617F cells resist to TGF-ß antiproliferative activity. Although TGF-ß receptors and SMAD2/3 expressions are similar in both cell types, TGF-ß-induced phosphorylation of SMAD2/3 is reduced in UT-7 JAK2V617F cells compared with JAK2 WT cells. We confirmed that JAK2V617F mutated cells are resistant to the antiproliferative effect of TGF-ß in a competitive assay as we observed a positive selection of JAK2V617F cells when exposed to TGF-ß. Using cell lines, CD34-positive cells from MPN patients and bone marrow cells from JAK2V617F knock-in mice we identified a down regulation of the SHP-1 phosphatase, which is required for the regulation of HSC quiescence by TGF-ß. The transduction of SHP-1 cDNA (but not a phosphatase inactive cDNA) restores the antiproliferative effect of TGF-ß in JAK2V617F mutated cells. Finally, SC-1, a known agonist of SHP-1, antagonized the selection of JAK2V617F mutated cells in the presence of TGF-ß. In conclusion, we show a JAK2-dependent down regulation of SHP-1 in MPN patients' cells which is related to their resistance to the antiproliferative effect of TGF-ß. This may participate in the clonal selection of cancer cells in MPNs.


Asunto(s)
Proliferación Celular , Janus Quinasa 2 , Trastornos Mieloproliferativos , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Factor de Crecimiento Transformador beta , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Ratones , Línea Celular Tumoral , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación hacia Abajo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína Smad2/genética , Mutación/genética
11.
Clin Lab ; 70(10)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39382932

RESUMEN

BACKGROUND: This study aimed to explore the clinical significance of ruxolitinib and its effects on the proliferation and apoptosis of human erythroleukemia (HEL) cells and the expression of immune checkpoint molecules programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), and regulatory T cells (Tregs) in HEL cells and JAK2 V617F-positive patients with myeloproliferative neoplasms (MPNs). METHODS: JAK2 V617F-positive patients with MPNs admitted to the Baoding No. 1 Hospital from January 2016 to September 2023 were recruited, including 30 patients for the newly diagnosed group and 10 for the treatment group. Additionally, 15 healthy volunteers were selected as the control group. JAK2 V617F mutation was detected by using fluorescence quantitative PCR, and the expression levels of phosphorylated JAK2 (p-JAK2), PD-1, and PD-L1 in fresh bone marrow were examined by immunohistochemistry. HEL cells were treated with ruxolitinib at different concentrations (0, 50, 100, 250, 500, and 1,000 nmol/L). Cell viability was detected by CCK-8 assay. The mRNA expression levels of JAK2, PD-1, and PD-L1 were determined by using fluorescence quantitative PCR. The protein expression of p-JAK2 was detected by Western blot and those of PD-1 and PD-L1 were evaluated by flow cytometry. The expression of PD-1, PD-L1, and Tregs after the 48-hour co-culture of primary bone marrow cells and HEL cells were also analyzed by flow cytometry. RESULTS: In the newly diagnosed group, the bone marrow myeloid cells highly expressed p-JAK2, PD-1, and PD-L1. The Tregs expression in their peripheral blood increased and was significantly higher than those in the treatment and control groups (all p < 0.05). Ruxolitinib at different concentrations could inhibit the proliferation of HEL cells and was positively correlated with treatment time and dose. Additionally, ruxolitinib could reduce p-JAK2, PD-1, and PD-L1 expression in HEL cells and Tregs expression. CONCLUSIONS: Ruxolitinib reduces the expression of p-JAK2, PD-1, and PD-L1 in JAK2 V617F-positive cells by specifically inhibiting the JAK2 signaling pathway, thereby suppressing the progression of MPNs.


Asunto(s)
Antígeno B7-H1 , Janus Quinasa 2 , Trastornos Mieloproliferativos , Nitrilos , Pirazoles , Pirimidinas , Humanos , Pirazoles/farmacología , Pirazoles/uso terapéutico , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Nitrilos/farmacología , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/metabolismo , Adulto , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Anciano , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Mutación
13.
Sci Rep ; 14(1): 23725, 2024 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390116

RESUMEN

The persistent infection of high-risk human papillomavirus (HPV) and the progression of cervical cancer necessitate the involvement of microenvironmental immunity. As cervical lesions advance, there is an observed increase in the infiltration of type 2 (M2) macrophages. However, the precise mechanism driving this increased infiltration of M2 macrophages remains unclear. In this study, we investigated the role of exosomes in polarising M2 macrophages in cervical lesions associated with HPV E6. Through the analysis of bioinformatics data and clinical specimens, we discovered a positive correlation between HPV E6/E7 mRNA copy number and the level of M2 macrophage infiltration. Exosomes derived from HPV E6 overexpressed (HPV E6+) cervical squamous cell carcinoma (CESC) cells were found to induce the polarisation of macrophages towards M2 type. Specifically, miR-204-5p, enriched in HPV E6 + CESC exosomes, was transported into macrophages and triggered M2 macrophage polarisation by inhibiting JAK2. The clinical relevance of exosomal miR-204-5p in the progression of cervical lesions was validated through serum samples from 35 cases. Exosomal miR-204-5p emerges as a critical factor influencing M2 macrophage polarisation and is correlated with the severity of cervical lesions. Consequently, miR-204-5p could be used as a potential treatment and a candidate biomarker for cervical lesions.


Asunto(s)
Exosomas , Macrófagos , MicroARNs , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Microambiente Tumoral , Neoplasias del Cuello Uterino , Femenino , Humanos , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Exosomas/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/virología , MicroARNs/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
14.
J Nanobiotechnology ; 22(1): 632, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39415256

RESUMEN

Neutrophil polarization contributes to inflammation and its resolution, but the role of neutrophil polarization in myocardial ischemia/reperfusion (I/R) injury remains unknown. Cardiomyocytes (CMs) participate in cardiac inflammation by secreting extracellular vesicles (EVs). Therefore, we investigated the role of neutrophil polarization in myocardial I/R injury and the mechanism by which CM-derived EVs regulated neutrophil polarization. In the present study, our data showed that N1 neutrophil polarization enlarged cardiac infarct size and exacerbated cardiac dysfunction at the early stage of myocardial I/R. Further, CM-EV-derived miR-9-5p was identified as a mediator inducing neutrophils to the N1 phenotype. Mechanistically, miR-9-5p directly suppressed SOCS5 and SIRT1 expression, resulting in activating JAK2/STAT3 and NF-κB signaling pathways in neutrophils. Importantly, we confirmed that serum EV-derived miR-9-5p levels were independently associated with cardiovascular mortality in patients with ST-segment elevation myocardial infarction undergoing percutaneous coronary intervention. These findings suggest neutrophil polarization is a promising therapeutic target against myocardial I/R-induced inflammation and injury, and serum EV-derived miR-9-5p is a promising prognostic biomarker for cardiovascular mortality in patients with ST-segment elevation myocardial infarction undergoing percutaneous coronary intervention.


Asunto(s)
Vesículas Extracelulares , Ratones Endogámicos C57BL , MicroARNs , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Neutrófilos , MicroARNs/genética , MicroARNs/metabolismo , Animales , Miocitos Cardíacos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Vesículas Extracelulares/metabolismo , Neutrófilos/metabolismo , Ratones , Masculino , Transducción de Señal , Sirtuina 1/metabolismo , Sirtuina 1/genética , Humanos , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , FN-kappa B/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/genética
15.
Rinsho Ketsueki ; 65(8): 810-818, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-39231712

RESUMEN

Treatment selection for patients with polycythemia vera (PV) is based on patient age and history of thrombosis. The standard treatment is low-dose aspirin and phlebotomy for low-risk PV, with cytoreductive therapy added for high-risk PV. Thrombotic events and disease progression due to PV clone expansion affect the prognosis of PV. Although phlebotomy is effective in controlling hematocrit level, it has no effect on disease progression or PV-related symptoms. In Western countries, interferon (IFN) has been used as a cytoreductive therapy for PV. Long-term IFN therapy has been shown to result in sustained hematologic remission and molecular responses. Ropeginterferon-α-2b (ropeg-IFN), which is administered every two weeks, has recently become available. Clinical trials in patients with PV have shown that ropeg-IFN treatment is safe and efficacious, reducing JAK2V617F allele burden. Ropeg-IFN could ultimately affect long-term hematologic remission and molecular response in younger patients with low-risk PV, and may even offer a cure.


Asunto(s)
Policitemia Vera , Policitemia Vera/terapia , Humanos , Janus Quinasa 2/genética , Interferón-alfa/uso terapéutico , Interferón-alfa/administración & dosificación , Factores de Riesgo
16.
Exp Hematol ; 139: 104636, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39237052

RESUMEN

CREB3L1, a gene encoding the endoplasmic reticulum stress transducer, is specifically overexpressed in platelet RNA from patients with myeloproliferative neoplasms (MPNs). However, the pathophysiological roles of CREB3L1 overexpression remain unclear. In the present study, we aimed to study CREB3L1 messenger RNA (mRNA) expression in the red blood cells (RBCs) of patients with MPN and its role in erythrocytosis. Elevated expression of CREB3L1 was exclusively observed in the RBCs of patients with polycythemia vera (PV) harboring JAK2 exon 12 mutations, but not in those harboring JAK2 V617F mutation or control subjects. In erythropoiesis, CREB3L1 expression was sharply induced in erythroblasts of bone marrow cells collected from patients with JAK2 exon 12 mutation. This was also evident when erythropoiesis was induced in vitro using hematopoietic stem and progenitor cells (HSPCs) with JAK2 exon 12 mutation. Interestingly, overexpression of CREB3L1 in RBCs was observed in patients with reactive erythrocytosis whose serum erythropoietin (EPO) levels exceeded 100 mIU/mL. Elevated CREB3L1 expression was also observed in the erythroblasts of a patient with acute erythroid leukemia. EPO-dependent induction of CREB3L1 was evident in erythroblasts differentiated from HSPCs in vitro, regardless of driver mutation status or MPN pathogenesis. These data strongly suggest that CREB3L1 overexpression in RBCs is associated with hyperactivation of the EPO receptor and its downstream molecule, JAK2. Short hairpin RNA (shRNA) knockdown of CREB3L1 expression in HSPCs blocked erythroblast formation in vitro. These results suggest that CREB3L1 is required for erythropoiesis in the presence of JAK2 exon 12 mutation or high level of EPO, possibly by antagonizing cellular stress.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Eritropoyesis , Exones , Janus Quinasa 2 , Humanos , Eritropoyesis/genética , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Exones/genética , Masculino , Femenino , Eritroblastos/metabolismo , Eritroblastos/patología , Persona de Mediana Edad , Eritropoyetina/metabolismo , Eritropoyetina/genética , Mutación , Policitemia/genética , Policitemia/metabolismo , Policitemia/sangre , Policitemia/patología , Policitemia Vera/genética , Policitemia Vera/metabolismo , Policitemia Vera/sangre , Policitemia Vera/patología , Anciano , Proteínas del Tejido Nervioso
17.
J Thromb Haemost ; 22(11): 3199-3208, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39260744

RESUMEN

BACKGROUND: Myeloproliferative neoplasms (MPNs) are characterized by a high rate of thrombotic complications that contribute to morbidity and mortality. MPN-related thrombogenesis is assumed to be multifactorial, involving both procoagulant and proinflammatory processes. Whether impaired fibrinolysis also participates in the prothrombotic phenotype of MPN has been poorly investigated. OBJECTIVES: We determined whether MPN, particularly JAK2V617F-positive MPN, is associated with fibrinolytic changes. METHODS: Tissue-type plasminogen activator (tPA)-mediated fibrinolysis was evaluated both in whole blood and plasma from mice with a hematopoietic-restricted Jak2V617F expression compared with wild-type (WT) mice (Jak2WT) using (1) halo clot lysis, (2) front lysis, and (3) plasmin generation assays. tPA clot lysis assay was performed in the plasma from 65 MPN patients (JAK2V617F mutation, n = 50; CALR mutations, n = 9) compared with 28 healthy controls. RESULTS: In whole blood from Jak2V617F mice, we observed a decreased fibrinolysis characterized by a significantly lower halo clot lysis rate compared with Jak2WT (95 ± 22 vs 147 ± 39 AU/min; P < .05). Similar results were observed in plasma (halo clot lysis rate, 130 ± 27 vs 186 ± 29 AU/min; front lysis rate, 2.8 ± 1.6 vs 6.1 ± 1.2 µm.min-1; P < .05). Plasmin generation was significantly decreased both in plasma clots and standardized fibrin clots from Jak2V617F mice compared with Jak2WT mice. Among MPN patients, impaired tPA-related fibrinolysis with prolonged clot lysis time was observed in JAK2V617F and CALR patients. Plasminogen activator inhibitor-1 and α2-antiplasmin were significantly increased in plasma from JAK2V617F patients compared with controls. CONCLUSION: Our results suggest that impaired tPA-mediated fibrinolysis represents an important prothrombotic mechanism in MPN patients that requires confirmation in larger studies.


Asunto(s)
Fibrinólisis , Janus Quinasa 2 , Trastornos Mieloproliferativos , Activador de Tejido Plasminógeno , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Animales , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/genética , Humanos , Activador de Tejido Plasminógeno/sangre , Trombosis/sangre , Trombosis/genética , Mutación , Masculino , Fibrinolisina/metabolismo , Femenino , Ratones , Persona de Mediana Edad , Ratones Endogámicos C57BL , Estudios de Casos y Controles , Calreticulina/genética , Anciano , Fenotipo , Ratones Transgénicos , Coagulación Sanguínea
19.
Circ Res ; 135(9): 890-909, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39263750

RESUMEN

BACKGROUND: Salt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP. METHODS: We performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function. RESULTS: We found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha). CONCLUSIONS: Our findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.


Asunto(s)
Presión Sanguínea , Hipertensión , Janus Quinasa 2 , Ratones Noqueados , Factor de Transcripción STAT3 , Cloruro de Sodio Dietético , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Animales , Humanos , Ratones , Cloruro de Sodio Dietético/efectos adversos , Masculino , Factor de Transcripción STAT3/metabolismo , Hipertensión/metabolismo , Proteína smad3/metabolismo , Proteína smad3/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Células Mieloides/enzimología , Femenino , Monocitos/metabolismo , Monocitos/efectos de los fármacos
20.
Cell Genom ; 4(10): 100659, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39317187

RESUMEN

Evidence from clinical trials suggests that CXCR4 antagonists enhance immunotherapy effectiveness in several cancers. However, the specific mechanisms through which CXCR4 contributes to immune cell phenotypes are not fully understood. Here, we employed single-cell transcriptomic analysis and identified CXCR4 as a marker gene in T cells, with CD8+PD-1high exhausted T (Tex) cells exhibiting high CXCR4 expression. By blocking CXCR4, the Tex phenotype was attenuated in vivo. Mechanistically, CXCR4-blocking T cells mitigated the Tex phenotype by regulating the JAK2-STAT3 pathway. Single-cell RNA/TCR/ATAC-seq confirmed that Cxcr4-deficient CD8+ T cells epigenetically mitigated the transition from functional to exhausted phenotypes. Notably, clinical sample analysis revealed that CXCR4+CD8+ T cells showed higher expression in patients with a non-complete pathological response. Collectively, these findings demonstrate the mechanism by which CXCR4 orchestrates CD8+ Tex cells and provide a rationale for combining CXCR4 antagonists with immunotherapy in clinical trials.


Asunto(s)
Linfocitos T CD8-positivos , Janus Quinasa 2 , Receptores CXCR4 , Factor de Transcripción STAT3 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Animales , Ratones , Humanos , Transducción de Señal , Fenotipo , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA