Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 79
Filtrer
1.
Toxicol Ind Health ; 40(12): 637-642, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39222320

RÉSUMÉ

Arsenic and its inorganic compounds affect numerous organs and systemic functions, such as the nervous and hematopoietic systems, liver, kidneys, and skin. Despite a large number of studies on arsenic toxicity, rare reports have investigated the leukopenia incidence in workers exposed to arsenic. In workplaces, the main source of workers' exposure is the contaminated air by the inorganic arsenic in mines, arsenic or copper smelter industries, and chemical factories. Erythropoiesis inhibition is one of the arsenic effects and it is related to regulatory factor GATA-1. This factor is necessary for the normal differentiation of early erythroid progenitors. JAK-STAT is an important intracellular signal transduction pathway responsible for the mediating normal functions of several cytokines related to cell proliferation and hematopoietic systems development and regulation. Arsenic inactivates JAK-STAT by inhibiting JAK tyrosine kinase and using the IFNγ pathway. The intravascular hemolysis starts after the absorption phase when arsenic binds to the globin of hemoglobin in erythrocytes and is transported into the body, which increases the oxidation of sulfhydryl groups in hemoglobin. So, this article intends to highlight the potential leukopenia risk via inhalation for workers exposed to arsenic and suggests a possible mechanism for this leukopenia through the JAK-signal transducer and activator of transcription (STAT) pathway inhibition.


Sujet(s)
Arsenic , Leucopénie , Exposition professionnelle , Humains , Arsenic/toxicité , Janus kinases/métabolisme , Leucopénie/induit chimiquement , Leucopénie/épidémiologie , Exposition professionnelle/effets indésirables , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription STAT/métabolisme
2.
Front Immunol ; 15: 1385473, 2024.
Article de Anglais | MEDLINE | ID: mdl-38720890

RÉSUMÉ

Interferons (IFNs) are a family of cytokines that activate the JAK-STAT signaling pathway to induce an antiviral state in cells. Interleukin 27 (IL-27) is a member of the IL-6 and/or IL-12 family that elicits both pro- and anti-inflammatory responses. Recent studies have reported that IL-27 also induces a robust antiviral response against diverse viruses, both in vitro and in vivo, suggesting that IFNs and IL-27 share many similarities at the functional level. However, it is still unknown how similar or different IFN- and IL-27-dependent signaling pathways are. To address this question, we conducted a comparative analysis of the transcriptomic profiles of human monocyte-derived macrophages (MDMs) exposed to IL-27 and those exposed to recombinant human IFN-α, IFN-γ, and IFN-λ. We utilized bioinformatics approaches to identify common differentially expressed genes between the different transcriptomes. To verify the accuracy of this approach, we used RT-qPCR, ELISA, flow cytometry, and microarrays data. We found that IFNs and IL-27 induce transcriptional changes in several genes, including those involved in JAK-STAT signaling, and induce shared pro-inflammatory and antiviral pathways in MDMs, leading to the common and unique expression of inflammatory factors and IFN-stimulated genes (ISGs)Importantly, the ability of IL-27 to induce those responses is independent of IFN induction and cellular lineage. Additionally, functional analysis demonstrated that like IFNs, IL-27-mediated response reduced chikungunya and dengue viruses replication in MDMs. In summary, IL-27 exhibits properties similar to those of all three types of human IFN, including the ability to stimulate a protective antiviral response. Given this similarity, we propose that IL-27 could be classified as a distinct type of IFN, possibly categorized as IFN-pi (IFN-π), the type V IFN (IFN-V).


Sujet(s)
Fièvre chikungunya , Dengue , Interleukine-27 , Janus kinases , Macrophages , Transduction du signal , Humains , Cellules cultivées , Fièvre chikungunya/immunologie , Fièvre chikungunya/virologie , Virus du chikungunya/immunologie , Dengue/immunologie , Dengue/virologie , Virus de la dengue/physiologie , Virus de la dengue/immunologie , Interférons/métabolisme , Interleukine-27/métabolisme , Interleukines/immunologie , Interleukines/pharmacologie , Janus kinases/métabolisme , Macrophages/immunologie , Macrophages/virologie , Transduction du signal/génétique , Facteurs de transcription STAT/métabolisme , Transcriptome , Réplication virale
3.
J Am Chem Soc ; 146(19): 13317-13325, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38700457

RÉSUMÉ

We describe the synthesis and biological testing of ruthenium-bipyridine ruxolitinib (RuBiRuxo), a photoreleasable form of ruxolitinib, a JAK inhibitor used as an antitumoral agent in cutaneous T-cell lymphomas (CTCL). This novel caged compound is synthesized efficiently, is stable in aqueous solution at room temperature, and is photoreleased rapidly by visible light. Irradiation of RuBiRuxo reduces cell proliferation and induces apoptosis in a light- and time-dependent manner in a CTCL cell line. This effect is specific and is mediated by a decreased phosphorylation of STAT proteins. Our results demonstrate the potential of ruthenium-based photocompounds and light-based therapeutic approaches for the potential treatment of cutaneous lymphomas and other pathologies.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Nitriles , Pyrazoles , Pyrimidines , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Nitriles/composition chimique , Nitriles/pharmacologie , Nitriles/synthèse chimique , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Pyrimidines/synthèse chimique , Apoptose/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie , Pyrazoles/composition chimique , Pyrazoles/synthèse chimique , Lignée cellulaire tumorale , Inhibiteurs des Janus kinases/pharmacologie , Inhibiteurs des Janus kinases/composition chimique , Inhibiteurs des Janus kinases/synthèse chimique , Ruthénium/composition chimique , Ruthénium/pharmacologie , Lumière , Structure moléculaire , Janus kinases/antagonistes et inhibiteurs , Janus kinases/métabolisme
4.
Rev Assoc Med Bras (1992) ; 70(3): e20231167, 2024.
Article de Anglais | MEDLINE | ID: mdl-38656003

RÉSUMÉ

OBJECTIVE: The aim of this study was to analyze possible alterations (morphological and inflammatory) in the ocular cells of fetuses from mothers with insulin resistance exposed to saturated fatty acids through the period of pregnancy. METHODS: Wistar female rats were induced to develop insulin resistance before pregnancy. Fetuses' skulls were collected on the 20th day of intrauterine life. The rats were separated on the first day of management into two groups according to the diet applied: control group (C): diet containing soybean oil as a source of fat; and saturated fatty acid group (S): diet containing butter as a source of fat. RESULTS: Histological and immunohistochemical analyses have been conducted. The immunohistochemical analyses of interleukin 6, suppressor of cytokine signaling, 3 and signal transducer and activator of transcription 3 did not demonstrate alterations in the expression of proteins in the fetuses of mothers fed with a saturated fatty diet. Moreover, no histopathological changes were noticed between groups. CONCLUSION: The saturated fatty diet does not induce tissue changes or activate the Janus kinase/signal transducer and activator of transcription signaling pathway during eye development in the fetuses of mothers with insulin resistance.


Sujet(s)
Insulinorésistance , Janus kinases , Rat Wistar , Transduction du signal , Animaux , Femelle , Grossesse , Transduction du signal/effets des médicaments et des substances chimiques , Insulinorésistance/physiologie , Janus kinases/métabolisme , Acides gras/analyse , Matières grasses alimentaires/pharmacologie , Matières grasses alimentaires/effets indésirables , Foetus/effets des médicaments et des substances chimiques , Immunohistochimie , Facteur de transcription STAT-3/métabolisme , Interleukine-6/analyse , Interleukine-6/métabolisme , Rats , Oeil/embryologie , Oeil/effets des médicaments et des substances chimiques
5.
Eur J Med Chem ; 263: 115935, 2024 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-37989057

RÉSUMÉ

A series of hybrid inhibitors, combining pharmacophores of known kinase inhibitors bearing anilino-purines (ruxolitinib, ibrutinib) and benzohydroxamate HDAC inhibitors (nexturastat A), were generated in the present study. The compounds have been synthesized and tested against solid and hematological tumor cell lines. Compounds 4d-f were the most promising in cytotoxicity assays (IC50 ≤ 50 nM) vs. hematological cells and displayed moderate activity in solid tumor models (EC50 = 9.3-21.7 µM). Compound 4d potently inhibited multiple kinase targets of interest for anticancer effects, including JAK2, JAK3, HDAC1, and HDAC6. Molecular dynamics simulations showed that 4d has stable interactions with HDAC and members of the JAK family, with differences in the hinge binding energy conferring selectivity for JAK3 and JAK2 over JAK1. The kinase inhibition profile of compounds 4d-f allows selective cytotoxicity, with minimal effects on non-tumorigenic cells. Moreover, these compounds have favorable pharmacokinetic profiles, with high stability in human liver microsomes (e.g., see t1/2: >120 min for 4f), low intrinsic clearance, and lack of significant inhibition of four major CYP450 isoforms.


Sujet(s)
Antinéoplasiques , Tumeurs , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Janus kinases , Purines/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire
6.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-37834225

RÉSUMÉ

Breast cancer remains the most frequently diagnosed cancer in women worldwide. Tumors that express hormone receptors account for 75% of all cases. Understanding alternative signaling cascades is important for finding new therapeutic targets for hormone receptor-positive breast cancer patients. JAK-STAT signaling is commonly activated in hormone receptor-positive breast tumors, inducing inflammation, proliferation, migration, and treatment resistance in cancer cells. In hormone receptor-positive breast cancer, the JAK-STAT cascade is stimulated by hormones and cytokines, such as prolactin and IL-6. In normal cells, JAK-STAT is inhibited by the action of the adaptor protein, LNK. However, the role of LNK in breast tumors is not fully understood. This review compiles published reports on the expression and activation of the JAK-STAT pathway by IL-6 and prolactin and potential inhibition of the cascade by LNK in hormone receptor-positive breast cancer. Additionally, it includes analyses of available datasets to determine the level of expression of LNK and various members of the JAK-STAT family for the purpose of establishing associations between expression and clinical outcomes. Together, experimental evidence and in silico studies provide a better understanding of the potential implications of the JAK-STAT-LNK loop in hormone receptor-positive breast cancer progression.


Sujet(s)
Tumeurs du sein , Transduction du signal , Humains , Femelle , Transduction du signal/physiologie , Janus kinases/métabolisme , Prolactine/métabolisme , Interleukine-6/métabolisme , Facteurs de transcription STAT/métabolisme
7.
Int J Mol Sci ; 24(13)2023 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-37446365

RÉSUMÉ

The Krüppel-like factor 13 (KLF13) has emerged as an important transcription factor involved in essential processes of the central nervous system (CNS). It predominantly functions as a transcriptional repressor, impacting the activity of several signaling pathways with essential roles in the CNS, including the JAK/STAT pathway, which is the canonical mediator of growth hormone (GH) signaling. It is now recognized that GH has important actions as a neurotrophic factor. Therefore, we analyzed the effects of KLF13 on the activity of the JAK/STAT signaling pathway in the hippocampus-derived cell line HT22. Results showed that KLF13 directly regulates the expression of several genes involved in the JAK-STAT pathway, including Jak1, Jak2, Jak3, and Socs1, by associating with their proximal gene promoters. In addition, it was found that in KLF13-deficient HT22 neurons, the expression of Jak1, Stat3, Socs1, Socs3, and Igf1 was dysregulated, exhibiting mRNA levels that went up to 7-fold higher than the control cell line. KLF13 displayed a differential effect on the GH-induced JAK/STAT pathway activity, decreasing the STAT3 branch while enhancing the STAT5 branch. In KLF13-deficient HT22 cells, the activity of the STAT3 branch was enhanced, mediating the GH-dependent augmented expression of the JAK/STAT output genes Socs1, Socs3, Igf1, and Bdnf. Furthermore, GH treatment increased both the nuclear content of KLF13 and Klf13 mRNA levels, suggesting that KLF13 could be part of the mechanisms that maintain the homeostatic state of this pathway. These findings support the notion that KLF13 is a regulator of JAK/STAT activity.


Sujet(s)
Janus kinases , Transduction du signal , Janus kinases/génétique , Janus kinases/métabolisme , Facteurs de transcription STAT/génétique , Facteurs de transcription STAT/métabolisme , Protéines SOCS/métabolisme , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme , ARN messager/métabolisme
8.
Int J Mol Sci ; 24(12)2023 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-37373437

RÉSUMÉ

Several cytokines with major biological functions in inflammatory diseases exert their functions through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signal transduction pathway. JAKs phosphorylate the cytoplasmic domain of the receptor, inducing the activation of its substrates, mainly the proteins known as STATs. STATs bind to these phosphorylated tyrosine residues and translocate from the cytoplasm to the nucleus, further regulating the transcription of several genes that regulate the inflammatory response. The JAK/STAT signaling pathway plays a critical role in the pathogenesis of inflammatory diseases. There is also increasing evidence indicating that the persistent activation of the JAK/STAT signaling pathway is related to several inflammatory bone (osteolytic) diseases. However, the specific mechanism remains to be clarified. JAK/STAT signaling pathway inhibitors have gained major scientific interest to explore their potential in the prevention of the destruction of mineralized tissues in osteolytic diseases. Here, our review highlights the importance of the JAK/STAT signaling pathway in inflammation-induced bone resorption and presents the results of clinical studies and experimental models of JAK inhibitors in osteolytic diseases.


Sujet(s)
Janus kinases , Facteurs de transcription STAT , Humains , Janus kinases/métabolisme , Facteurs de transcription STAT/métabolisme , Transduction du signal/physiologie , Cytokines/métabolisme , Inflammation/traitement médicamenteux
9.
Genes (Basel) ; 14(6)2023 05 24.
Article de Anglais | MEDLINE | ID: mdl-37372319

RÉSUMÉ

The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway constitutes a rapid signaling module from the cell surface to the nucleus, and activates different cellular responses, such as proliferation, survival, migration, invasion, and inflammation. When the JAK/STAT pathway is altered, it contributes to cancer progression and metastasis. STAT proteins play a central role in developing cervical cancer, and inhibiting the JAK/STAT signaling may be necessary to induce tumor cell death. Several cancers show continuous activation of different STATs, including cervical cancer. The constitutive activation of STAT proteins is associated with a poor prognosis and overall survival. The human papillomavirus (HPV) oncoproteins E6 and E7 play an essential role in cervical cancer progression, and they activate the JAK/STAT pathway and other signals that induce proliferation, survival, and migration of cancer cells. Moreover, there is a crosstalk between the JAK/STAT signaling cascade with other signaling pathways, where a plethora of different proteins activate to induce gene transcription and cell responses that contribute to tumor growth. Therefore, inhibition of the JAK/STAT pathway shows promise as a new target in cancer treatment. In this review, we discuss the role of the JAK/STAT pathway components and the role of the HPV oncoproteins associated with cellular malignancy through the JAK/STAT proteins and other signaling pathways to induce tumor growth.


Sujet(s)
Infections à papillomavirus , Tumeurs du col de l'utérus , Femelle , Humains , Janus kinases/génétique , Janus kinases/métabolisme , Transduction du signal/physiologie , Tumeurs du col de l'utérus/génétique , Infections à papillomavirus/génétique , Facteurs de transcription STAT/génétique , Facteurs de transcription STAT/métabolisme
10.
An Bras Dermatol ; 98(5): 656-677, 2023.
Article de Anglais | MEDLINE | ID: mdl-37230920

RÉSUMÉ

The JAK-STAT signaling pathway mediates important cellular processes such as immune response, carcinogenesis, cell differentiation, division and death. Therefore, drugs that interfere with different JAK-STAT signaling patterns have potential indications for various medical conditions. The main dermatological targets of JAK-STAT pathway inhibitors are inflammatory or autoimmune diseases such as psoriasis, vitiligo, atopic dermatitis and alopecia areata; however, several dermatoses are under investigation to expand this list of indications. As JAK-STAT pathway inhibitors should gradually occupy a relevant space in dermatological prescriptions, this review presents the main available drugs, their immunological effects, and their pharmacological characteristics, related to clinical efficacy and safety, aiming to validate the best dermatological practice.


Sujet(s)
Dermatologie , Inhibiteurs des Janus kinases , Vitiligo , Humains , Inhibiteurs des Janus kinases/pharmacologie , Inhibiteurs des Janus kinases/usage thérapeutique , Janus kinases/métabolisme , Janus kinases/pharmacologie , Transduction du signal , Facteurs de transcription STAT/métabolisme , Facteurs de transcription STAT/pharmacologie , Vitiligo/traitement médicamenteux
11.
Acta Cir Bras ; 38: e381023, 2023.
Article de Anglais | MEDLINE | ID: mdl-37132754

RÉSUMÉ

PURPOSE: To investigate the role of cyanidin-3-O-glucoside (C3G) in renal ischemia/reperfusion (I/R) injury and the potential mechanisms. METHODS: Mouse models were established by clamping the left renal vessels, and in vitro cellular models were established by hypoxic reoxygenation. RESULTS: Renal dysfunction and tissue structural damage were significantly higher in the I/R group. After treatment with different concentrations of C3G, the levels of renal dysfunction and tissue structural damage decreased at different levels. And its protective effect was most pronounced at 200 mg/kg. The use of C3G reduced apoptosis as well as the expression of endoplasmic reticulum stress (ERS)-related proteins. Hypoxia/reoxygenation (H/R)-induced apoptosis and ERS are dependent on oxidative stress in vitro. In addition, both AG490 and C3G inhibited the activation of JAK/STAT pathway and attenuated oxidative stress, ischemia-induced apoptosis and ERS. CONCLUSIONS: The results demonstrated that C3G blocked renal apoptosis and ERS protein expression by preventing reactive oxygen species (ROS) production after I/R via the JAK/STAT pathway, suggesting that C3G may be a potential therapeutic agent for renal I/R injury.


Sujet(s)
Maladies du rein , Lésion d'ischémie-reperfusion , Souris , Animaux , Janus kinases/métabolisme , Janus kinases/pharmacologie , Janus kinases/usage thérapeutique , Transduction du signal , Facteurs de transcription STAT/métabolisme , Facteurs de transcription STAT/pharmacologie , Facteurs de transcription STAT/usage thérapeutique , Lésion d'ischémie-reperfusion/métabolisme , Apoptose , Ischémie , Glucosides/pharmacologie
12.
Inflamm Res ; 72(6): 1257-1274, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37212867

RÉSUMÉ

OBJECTIVE AND DESIGN: The discovery of new inflammatory pathways and the mechanism of action of inflammatory, autoimmune, genetic, and neoplastic diseases led to the development of immunologically driven drugs. We aimed to perform a narrative review regarding the rising of a new class of drugs capable of blocking important and specific intracellular signals in the maintenance of these pathologies: the small molecules. MATERIALS/METHODS: A total of 114 scientific papers were enrolled in this narrative review. RESULTS: We describe in detail the families of protein kinases-Janus Kinase (JAK), Src kinase, Syk tyrosine kinase, Mitogen-Activated Protein Kinase (MAPK), and Bruton Tyrosine Kinase (BTK)-their physiologic function and new drugs that block these pathways of intracellular signaling. We also detail the involved cytokines and the main metabolic and clinical implications of these new medications in the field of dermatology. CONCLUSIONS: Despite having lower specificity compared to specific immunobiological therapies, these new drugs are effective in a wide variety of dermatological diseases, especially diseases that had few therapeutic options, such as psoriasis, psoriatic arthritis, atopic dermatitis, alopecia areata, and vitiligo.


Sujet(s)
Dermatologie , Psoriasis , Vitiligo , Humains , Auto-immunité , Psoriasis/traitement médicamenteux , Inflammation/traitement médicamenteux , Janus kinases/métabolisme
13.
Drugs ; 83(4): 299-314, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36913180

RÉSUMÉ

In recent years, better knowledge of the pathophysiology of inflammatory bowel diseases (IBD) has led to a relevant expansion of the therapeutic arsenal for these conditions. Janus kinase (JAK) inhibitors are a family of small molecules that block one or more of the intracellular tyrosine kinases, including JAK-1, JAK-2, JAK-3 and TYK-2. Tofacitinib, a non-selective small molecule JAK inhibitor, and upadacitinib and filgotinib, which are selective JAK-1 inhibitors, have been approved by the US Food and Drug Administration (FDA) for moderate-to-severe active ulcerative colitis. Compared to biological drugs, JAK inhibitors have a short half-life, rapid onset of action, and no immunogenicity. Both clinical trials and real-world evidence support the use of JAK inhibitors in the treatment of IBD. However, these therapies have been linked with multiple adverse events (AEs) including infection, hypercholesterolemia, venous thromboembolism, major adverse cardiovascular events, and malignancy. While early studies recognized several potential AEs, post-marketing trials have shown that tofacitinib may increase the risk of thromboembolic diseases and major cardiovascular events. The latter are seen in patients aged 50 years or older with cardiovascular risk factors. Hence, the benefits of treatment and risk stratification need to be considered when positioning tofacitinib. Novel JAK inhibitors with a more selective effect on JAK-1 have proven to be effective in both Crohn's disease and ulcerative colitis, offering a potentially safer and efficacious therapeutic option to patients, including those with previous non-response to other therapies such as biologics. Nevertheless, long-term effectiveness and safety data are required.


Sujet(s)
Maladies cardiovasculaires , Rectocolite hémorragique , Maladie de Crohn , Maladies inflammatoires intestinales , Inhibiteurs des Janus kinases , Humains , Inhibiteurs des Janus kinases/effets indésirables , Rectocolite hémorragique/traitement médicamenteux , Maladies inflammatoires intestinales/traitement médicamenteux , Maladie de Crohn/traitement médicamenteux , Maladies cardiovasculaires/traitement médicamenteux , Janus kinases
14.
Int J Biochem Cell Biol ; 158: 106409, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36997057

RÉSUMÉ

Extracellular matrix protein 2 (ECM2), which regulates cell proliferation and differentiation, has recently been reported as a prognostic indicator for multiple cancers, but its value in lower grade glioma (LGG) remains unknown. In this study, LGG transcriptomic data of 503 cases in The Cancer Genome Atlas (TCGA) database and 403 cases in The Chinese Glioma Genome Atlas (CGGA) database were collected to analyze ECM2 expression patterns and the relationship with clinical characteristics, prognosis, enriched signaling pathways, and immune-related markers. In addition, a total of 12 laboratory samples were used for experimental validation. Wilcoxon or Kruskal-Wallis tests demonstrated highly expressed ECM2 in LGG was positively associated with malignant histological features and molecular features such as recurrent LGG and isocitrate dehydrogenase (IDH) wild-type. Also, Kaplan-Meier (KM) curves proved high ECM2 expression could predict shorter overall survival in LGG patients, as multivariate analysis and meta-analysis claimed ECM2 was a deleterious factor for LGG prognosis. In addition, the enrichment of immune-related pathways for ECM2, for instance JAK-STAT pathway, was obtained by Gene Set Enrichment Analysis (GSEA) analysis. Furthermore, positive relationships between ECM2 expression with immune cells infiltration and cancer-associated fibroblasts (CAFs), iconic markers (CD163), and immune checkpoints (CD274, encoding PD-L1) were proved by Pearson correlation analysis. Finally, laboratory experiments of RT-qPCR and immunohistochemistry showed high expression of ECM2, as well as CD163 and PD-L1 in LGG samples. This study identifies ECM2, for the first time, as a subtype marker and prognostic indicator for LGG. ECM2 could also provide a reliable guarantee for further personalized therapy, synergizing with tumor immunity, to break through the current limitations and thus reinvigorating immunotherapy for LGG. AVAILABILITY OF DATA AND MATERIALS: Raw data from all public databases involved in this study are stored in the online repository (chengMD2022/ECM2 (github.com)).


Sujet(s)
Tumeurs du cerveau , Gliome , Humains , Antigène CD274 , Janus kinases , Pronostic , Facteurs de transcription STAT , Transduction du signal , Gliome/génétique , Gliome/thérapie , Immunothérapie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/thérapie
15.
Acta cir. bras ; Acta cir. bras;38: e381023, 2023. ilus, graf
Article de Anglais | LILACS, VETINDEX | ID: biblio-1439116

RÉSUMÉ

Purpose: To investigate the role of cyanidin-3-O-glucoside (C3G) in renal ischemia/reperfusion (I/R) injury and the potential mechanisms. Methods: Mouse models were established by clamping the left renal vessels, and in vitro cellular models were established by hypoxic reoxygenation. Results: Renal dysfunction and tissue structural damage were significantly higher in the I/R group. After treatment with different concentrations of C3G, the levels of renal dysfunction and tissue structural damage decreased at different levels. And its protective effect was most pronounced at 200 mg/kg. The use of C3G reduced apoptosis as well as the expression of endoplasmic reticulum stress (ERS)-related proteins. Hypoxia/reoxygenation (H/R)-induced apoptosis and ERS are dependent on oxidative stress in vitro. In addition, both AG490 and C3G inhibited the activation of JAK/STAT pathway and attenuated oxidative stress, ischemia-induced apoptosis and ERS. Conclusions: The results demonstrated that C3G blocked renal apoptosis and ERS protein expression by preventing reactive oxygen species (ROS) production after I/R via the JAK/STAT pathway, suggesting that C3G may be a potential therapeutic agent for renal I/R injury.


Sujet(s)
Animaux , Souris , Lésion d'ischémie-reperfusion , Système de signalisation des MAP kinases , Janus kinases , Atteinte rénale aigüe/physiopathologie , Ischémie , Anthocyanes/analyse
16.
Neurol Sci ; 43(11): 6271-6278, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-35849199

RÉSUMÉ

Multiple sclerosis (MS) is a chronic demyelinating autoimmune disease that affects the central nervous system (CNS), varying from relatively benign to severely disabling. Although the roles of several cytokines and chemokines in MS are well established, their roles in MS lesions and evolution remain a matter of debate. Soluble CD40L (sCD40L) is a ligand that induces lymphocyte proinflammatory activity by stimulating the activation and maturation of B cells, promoting isotype switching and affinity hypermutation. Circulating sCD40L levels reflect activation of the CD40-CD40L complex. The interaction between CD40 and CD40L is of fundamental importance, suggesting their role in MS pathogenesis. Interleukin-31 (IL-31) is a proinflammatory cytokine that plays a role in allergies, autoimmune diseases, and is a major factor in several chronic inflammatory diseases. IL-31 triggers the JAK-STAT pathway in several different cell types, to induce proliferation and tissue remodeling in fibroblasts, epithelial cells, and endothelial cells. Some studies have described a correlation between these two cytokines and decreased serum levels of sCD40L and IL-31 after MS treatment, accompanied by a lower inflammatory response. In this review, we emphasize the possible correlation and positive feedback between IL31 and sCD40L in the MS proinflammatory response. We also describe the justification for this hypothesis and whether it is possible to investigate these cytokines as biomarkers of MS.


Sujet(s)
Ligand de CD40 , Sclérose en plaques , Humains , Ligand de CD40/métabolisme , Cellules endothéliales/métabolisme , Janus kinases/métabolisme , Facteurs de transcription STAT/métabolisme , Transduction du signal , Marqueurs biologiques , Interleukines , Cytokines
17.
Med Res Rev ; 42(6): 2126-2167, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35762498

RÉSUMÉ

The rising pandemic caused by a coronavirus, resulted in a scientific quest to discover some effective treatments against its etiologic agent, the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). This research represented a significant scientific landmark and resulted in many medical advances. However, efforts to understand the viral mechanism of action and how the human body machinery is subverted during the infection are still ongoing. Herein, we contributed to this field with this compilation of the roles of both viral and human enzymes in the context of SARS-CoV-2 infection. In this sense, this overview reports that proteases are vital for the infection to take place: from SARS-CoV-2 perspective, the main protease (Mpro ) and papain-like protease (PLpro ) are highlighted; from the human body, angiotensin-converting enzyme-2, transmembrane serine protease-2, and cathepsins (CatB/L) are pointed out. In addition, the influence of the virus on other enzymes is reported as the JAK/STAT pathway and the levels of lipase, enzymes from the cholesterol metabolism pathway, amylase, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and glyceraldehyde 3-phosphate dehydrogenase are also be disturbed in SARS-CoV-2 infection. Finally, this paper discusses the importance of detailed enzymatic studies for future treatments against SARS-CoV-2, and how some issues related to the syndrome treatment can create opportunities in the biotechnological market of enzymes and the development of new drugs.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Alanine transaminase/pharmacologie , Amylases/pharmacologie , Angiotensines/pharmacologie , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Aspartate aminotransferases/pharmacologie , Cathepsines/pharmacologie , Cholestérol , Corps humain , Humains , Janus kinases/pharmacologie , Lactate dehydrogenases , Triacylglycerol lipase/pharmacologie , Papaïne/pharmacologie , SARS-CoV-2 , Facteurs de transcription STAT/pharmacologie , Protéases à sérine/pharmacologie , Transduction du signal
18.
Brasília; CONITEC; jun. 2022.
Non conventionel de Portugais | LILACS, Coleciona SUS | ID: biblio-1378051

RÉSUMÉ

CONTEXTO: Mielofibrose é uma neoplasia maligna rara que pode se desenvolver como doença primária, sendo uma doença mieloproliferativa crônica caracterizada pela falha da medula óssea e proliferação clonal de células mieloides associada com excesso de fibras de reticulina e/ou colágeno, e algum grau de atipia no megacariócito. O quadro clínico pode evoluir com esplenomegalia, anemia, sintomas constitucional (fadiga, sudorese noturna, febre), caquexia, dor óssea, infarto esplênico, prurido, trombose e sangramentos. A incidência na União Europeia e EUA é de 0,3 casos por 100.000 habitantes. Não há dados epidemiológicos robustos no Brasil. Ruxolitinibe é um inibidor seletivo das Janus Quinases associadas (JAKs) ­ JAK1 e JAK2. A desregulação da via JAK-STAT tem sido associada a vários tipos de câncer e aumento da proliferação e sobrevida de células malignas. TECNOLOGIA: Ruxolitinibe. PERGUNTA: O uso de ruxolitinibe no tratamento da mielofibrose risco intermediário-2 ou alto (classificação IPSS), em adultos, com contagem plaquetária acima de 100.000/mm3 é eficaz e seguro quando comprado ao


Sujet(s)
Humains , Janus kinases/antagonistes et inhibiteurs , Myélofibrose primitive/traitement médicamenteux , Système de Santé Unifié , Brésil , Analyse coût-bénéfice/économie
19.
Brasília; CONITEC; mar. 2022.
Non conventionel de Portugais | LILACS, BRISA/RedTESA | ID: biblio-1368853

RÉSUMÉ

INTRODUÇÃO: O baricitinbe, um imunomodulador que atua sobre a atividade da IL-6 (citocina pró-inflamatória), pode representar uma estratégia para o tratamento de pacientes com COVID-19 que tiveram comprometimento pulmonar devido a resposta hiperinflamátoria desencadeada pela tempestade de citocinas característica na infecção causada pelo vírus SARS-COV2. TECNOLOGIA: Baricitinibe (Olumiant®). EVIDÊNCIAS CLÍNICAS: Para seleção das evidências clínicas foi conduzida uma revisão sistemática da literatura em busca de ensaios clínicos randomizados (ECR), estudos observacionais (mundo real) e revisões sistemáticas que avaliassem os efeitos do baricitinibe como monoterapia ou associado aos cuidados usuais - definidos aqui como 'terapia padrão' (corticoesteróides sistêmicos, anticoagulantes, antimicrobianos/antivirais) no tratamento de pacientes adultos com COVID-19, hospitalizados e que necessitam de suplementação de oxigênio (máscara ou cateter nasal, alto fluxo de oxigênio ou ventilação não invasiva). As buscas eletrônicas foram realizadas nas bases de dados: the Cochrane Library, MEDLINE via Pubmed, Literatura Latino-Americana e do Caribe em Ciências da Saúde (LILACS), EMBASE e Centre for Reviews and Dissemination (CRD). O risco de viés dos estudos primários incluídos foi avaliado pelas ferramentas Risk of Bias versão 2 da Cochrane (para ECR) ou ROBINS-I (para estudos observacionais), e a qualidade metodológica das revisões sistemáticas foi avaliada pela ferramenta AMSTAR-2. A qualidade da evidência foi avaliada pelo sistema GRADE. Seis artigos foram incluídos na presente revisão, sendo dois deles referentes a um ensaio clínico randomizado (ECR), um estudo observacional e três revisões sistemáticas com meta-análise (RSMA), sendo uma


Sujet(s)
Humains , Oxygénothérapie/instrumentation , Janus kinases/antagonistes et inhibiteurs , Ventilation non effractive/instrumentation , SARS-CoV-2/effets des médicaments et des substances chimiques , COVID-19/traitement médicamenteux , Immunosuppresseurs/antagonistes et inhibiteurs , Système de Santé Unifié , Brésil , Analyse coût-bénéfice/économie , Patients hospitalisés
20.
Lima; IETSI; jun. 2021.
Non conventionel de Espagnol | BRISA/RedTESA | ID: biblio-1358225

RÉSUMÉ

INTRODUCCIÓN: El presente dictamen expone la evaluación de la eficacia y seguridad de tofacitinib, comparado con la mejor terapia de soporte, para el tratamiento de pacientes adultos con diagnóstico de colitis ulcerativa (CU) moderada a severa con falla o intolerancia a infliximab. La colitis ulcerativa (CU) es una enfermedad inflamatoria crónica, de causa desconocida y poco frecuente. La CU se caracteriza por episodios recurrentes y remitentes de inflamación que afectan a la capa mucosa del colon; produciendo episodios de diarrea que pueden estar acompañadas de sangrado. Aunque la CU puede presentarse a cualquier edad, la mayor incidencia ocurre entre los 15 y 30 años. Aunque se desconoce la incidencia de CU en Perú, el Hospital Nacional Edgardo Rebagliati Martins reportó 2.1 casos por año entre los años 2001 y 2003 y el Hospital Nacional Guillermo Almenara Irigoyen reportó 81 casos entre los años 2004 y 2014.  La severidad clínica de la CU es medida por índices de actividad y severidad de la enfermedad. Estos índices clasifican la inflamación a través de criterios clínicos y endoscópicos. El índice Mayo es el más utilizado en los ensayos clínicos y puede ser utilizado en el seguimiento de los pacientes durante el tratamiento. El puntaje del índice Mayo varía entre los 0 y 12 puntos (los puntajes más altos indican mayor gravedad). El tratamiento de la CU puede ser quirúrgico (colectomía) o farmacológico. Aunque el tratamiento quirúrgico puede ser curativo, este presenta un impacto negativo en la mortalidad, morbilidad y calidad de vida del paciente. El manejo farmacológico con inmunomoduladores tiene por objetivo lograr la mejora sintomática de la CU y la curación de la mucosa. En los casos de CU moderada a severa, se recomienda - además de los inmunomoduladores- la terapia biológica con inhibidores de TNFα (e.g., infliximab). En EsSalud, los pacientes con CU moderada a severa son tratados con infliximab; sin embargo, algunos de ellos presentan falla o intolerancia al tratamiento. Dado que estos pacientes con falla o intolerancia a infliximab no tienen otra alternativa de tratamiento, los especialistas de EsSalud sugieren el uso de tofacitinib para el tratamiento de estos pacientes. METODOLOGÍA: Se llevó a cabo una búsqueda sistemática de la literatura con el objetivo de identificar la mejor evidencia disponible sobre la eficacia y seguridad de tofacitinib, comparado con la mejor terapia de soporte, para el tratamiento de pacientes adultos con diagnóstico de CU moderada a severa con falla o intolerancia a infliximab. Se realizó una búsqueda sistemática en las bases de datos: The Cochrane Library, PubMed y LILACS. Adicionalmente, se realizó una búsqueda manual dentro de las bases de datos pertenecientes a grupos que realizan evaluación de tecnologías sanitarias (ETS) y guías de práctica clínica (GPC) incluyendo: el National Institute for Health and Care Excellence (NICE), la Canadian Agency for Drugs and Technologies in Health (CADTH), el Scottish Medicines Consortium (SMC), el Institute for Clinical and Economic Review (ICER), el Institute for Quality and Efficiency in Healthcare (IQWiG por sus siglas en alemán), la Base regional de informes de evaluación de tecnologías en salud de las Américas (BRISA), el Instituto de Evaluación de Tecnologías en Salud e Investigación (IETSI) y las páginas web de organizaciones especializadas en el manejo de CU. Finalmente, se realizó una búsqueda adicional en la página web de ClinicalTrials.gov para identificar EC en curso y cuyos resultados no hayan sido publicados. RESULTADOS: La presente sinopsis describe la evidencia disponible sobre la eficacia y seguridad de tofacitinib como tratamiento de pacientes adultos con CU moderada a severa y con falla o intolerancia a infliximab, según el tipo de publicación. CONCLUSIONES: El presente dictamen preliminar tuvo por objetivo evaluar la mejor evidencia disponible sobre la eficacia y seguridad de tofacitinib comparado con la mejor terapia de soporte para el tratamiento de pacientes con diagnóstico de CU moderada a severa con falla o intolerancia a infliximab. Luego de la búsqueda sistemática de la literatura se identificaron dos GPC (AGA, BSG), 4 ETS desarrolladas por IQWiG, NICE, SMC y CADTH y una publicación de Sandborn que presenta los resultados de los ECA OCTAVE 1 y OCTAVE 2. Las GPC de la AGA y la BSG recomiendan el uso de tofacitinib para el tratamiento de pacientes con CU moderada a severa y con falla a infliximab. Debido a las limitaciones de los análisis realizados por estas GPC (la AGA realizó un metaanálisis en red y la BSG realizó análisis de subgrupos), sus recomendaciones deben tomarse con precaución. Con respecto a las ETS de NICE, SMC, CADTH e IQWiG, las tres primeras recomendaron el uso de tofacitinib condicional a un descuento en el precio de lista; mientras que IQWiG concluyó que no es posible determinar si tofacitinib ofrece algún beneficio adicional. Pero, la recomendación de uso de tofacitinib fue condicionada a un descuento del precio de lista. Esto sugiere que el beneficio observado con tofacitinib no justifica el precio propuesto por el fabricante. Los resultados de los ECA OCTAVE 1 y OCTAVE 2 sugieren que, en el corto plazo (ocho semanas), tofacitinib sería más eficaz que placebo en el subgrupo de pacientes con falla o intolerancia a inhibidores de TNFα y el perfil de seguridad de tofacitinib sería similar al del placebo. Para el presente dictamen se tomaron en cuenta los siguientes aspectos: i) Dado que la CU es una enfermedad infrecuente, la población de interés del dictamen (pacientes con CU moderada a severa y falla o intolerancia a infliximab) sería muy pequeña; ii) en EsSalud, estos pacientes no cuentan con una alternativa de tratamiento para el control de la CU (vacío terapéutico); iii) las dos GPC evaluadas recomiendan el uso de tofacitinib en los pacientes con CU moderado a severo con falla o intolerancia a inhibidores de TNFα; iv) las ETS de NICE, SMC y CADTH, mediante metaanálisis en red y análisis de subgrupos, concluyen que tofacitinib es más eficaz e igual de seguro que placebo; aunque condicionaron su uso a la reducción del precio de lista de tofacitinib; v) la ETS de IQWiG concluyó que la evidencia disponible con seguimiento de al menos 12 meses es insuficiente para determinar si tofacitinib ofrecía mayor beneficio que otras terapias biológicas; vi) los resultados a corto plazo (ocho semanas) de los ECA OCTAVE 1 y OCTAVE 2 sugieren que tofacitinib tendría mayor eficacia (remisión y curación de la mucosa) y similar perfil de seguridad que placebo en el tratamiento de pacientes con CU moderada a severa con falla o intolerancia inhibidores de TNFα; y vii) aunque los EA relevantes presentados con tofacitinib (infecciones severas, cáncer de piel no melanoma y eventos cardiovasculares) son similares a los esperados con la mejor terapia de soporte, su aparición en un corto periodo de seguimiento (ocho semanas) nos obliga a mantener un adecuado seguimiento de los pacientes que reciben tofacitinib. Por lo expuesto, el IETSI aprueba el uso de tofacitinib en pacientes con diagnóstico de CU moderada a severa con falla o intolerancia a infliximab como producto farmacéutico no incluido en el Petitorio Farmacológico de EsSalud. La vigencia del presente dictamen es de un año, según lo establecido en el Anexo N° 1. Así, la continuación de dicha aprobación estará sujeta a la evaluación de los resultados obtenidos y de mayor evidencia que pueda surgir en el tiempo.


Sujet(s)
Humains , Rectocolite hémorragique/traitement médicamenteux , Janus kinases/antagonistes et inhibiteurs , Infliximab/effets indésirables , Efficacité en Santé Publique , Analyse coût-bénéfice
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE