Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 893
1.
Arch Dermatol Res ; 316(6): 290, 2024 May 29.
Article En | MEDLINE | ID: mdl-38809465

Enz_MoriL is a naturally occurring substance extracted from the leaves of Morus alba L. through enzymatic conversion. Historically, M. alba L. has been recognized for its potential to promote hair regrowth. However, the precise mechanism by which Enz_MoriL affects human hair follicle dermal papilla cells (hDPCs) remains unclear. The aim of this study was to investigate the molecular basis of Enz_MoriL's effect on hair growth in hDPCs. Interferon-gamma (IFN-γ) was used to examine the effects of Enz_MoriL on hDPCs during the anagen and catagen phases, as well as under conditions mimicking alopecia areata (AA). Enz_MoriL demonstrated the ability to promote cell proliferation in both anagen and catagen stages. It increased the levels of active ß-catenin in the catagen stage induced by IFN-γ, leading to its nuclear translocation. This effect was achieved by increasing the phosphorylation of GSK3ß and decreasing the expression of DKK-1. This stimulation induced proliferation in hDPCs and upregulated the expression of the Wnt family members 3a, 5a, and 7a at the transcript level. Additionally, Enz_MoriL suppressed JAK1 and STAT3 phosphorylation, contrasting with IFN-γ, which induced them in the catagen stage. In conclusion, Enz_MoriL directly induced signals for anagen re-entry into hDPCs by affecting the Wnt/ß-catenin pathway and enhancing the production of growth factors. Furthermore, Enz_MoriL attenuated and reversed the interferon-induced AA-like environment by blocking the JAK-STAT pathway in hDPCs.


Alopecia Areata , Cell Proliferation , Hair Follicle , Interferon-gamma , Wnt Signaling Pathway , beta Catenin , Humans , Hair Follicle/drug effects , Hair Follicle/cytology , Hair Follicle/metabolism , Cell Proliferation/drug effects , Wnt Signaling Pathway/drug effects , Interferon-gamma/metabolism , beta Catenin/metabolism , Alopecia Areata/metabolism , Alopecia Areata/drug therapy , Alopecia Areata/pathology , Cells, Cultured , Glycogen Synthase Kinase 3 beta/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Janus Kinases/metabolism , Dermis/cytology , Dermis/drug effects , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Hair/drug effects , Hair/growth & development , Wnt-5a Protein/metabolism , Janus Kinase 1/metabolism , Signal Transduction/drug effects , STAT Transcription Factors/metabolism
2.
Zhongguo Zhong Yao Za Zhi ; 49(8): 2188-2196, 2024 Apr.
Article Zh | MEDLINE | ID: mdl-38812234

This study aims to investigate the protective effect of salidroside(SAL) on renal damage in diabetic nephropathy(DN) mice based on the receptor for advanced glycation end products/janus activated kinase 1/signal transduction and activator of transcription 3(RAGE/JAK1/STAT3) signaling pathway. The mouse DN model was established by high-fat/high-sucrose diets combined with intraperitoneal injection of streptozocin(STZ). Mice were randomly divided into normal group, model group, low-dose SAL group(20 mg·kg~(-1)), high-dose SAL group(100 mg·kg~(-1)), and metformin group(140 mg·kg~(-1)), with 12 mice in each group. After establishing the DN model, mice were given drugs or solvent intragastrically, once a day for consecutive 10 weeks. Body weight, daily water intake, and fasting blood glucose(FBG) were measured every two weeks. After the last dose, the glucose tolerance test was performed, and the samples of 24-hour urine, serum, and kidney tissue were collected. The levels of 24 hours urinary total protein(24 h-UTP), serum creatinine(Scr), blood urea nitrogen(BUN), triglyceride(TG), total cholesterol(TC), low density lipoprotein cholesterol(LDL-C), and high density lipoprotein cholesterol(HDL-C) were detected by biochemical tests. Periodic acid-schiff(PAS) staining was used to observe the pathological changes in the kidney tissue. The protein expressions of α-smooth muscle actin(α-SMA), vimentin, and advanced glycation end products(AGEs) in kidneys were detected by immunohistochemical staining. The activities of superoxide dismutase(SOD), catalase(CAT), glutathione peroxidase(GSH-PX), and the level of malondialdehyde(MDA) in kidneys were detected by using a corresponding detection kit. Enzyme-linked immunosorbent assay(ELISA) was used to detect the levels of AGEs, carboxymethyllysine(CML), and carboxyethyllysine(CEL) in serum. The protein expressions of RAGE and the phosphorylation level of JAK1 and STAT3 in kidneys were detected by Western blot. Compared with the normal group, the levels of FBG, the area under the curve of glucose(AUCG), water intake, kidney index, 24 h-UTP, tubular injury score, extracellular matrix deposition ratio of the renal glomerulus, the serum levels of Scr, BUN, TG, LDL-C, AGEs, CEL, and CML, the level of MDA, the protein expressions of α-SMA, vimentin, AGEs, and RAGE, and the phosphorylation level of JAK1 and STAT3 in kidney tissue were increased significantly(P<0.01), while the level of HDL-C in serum and the activity of SOD, CAT, and GSH-PX in kidney tissue were decreased significantly(P<0.01). Compared with the model group, the above indexes of the high-dose SAL group were reversed significantly(P<0.05 or P<0.01). In conclusion, this study suggests that SAL can alleviate oxidative stress and renal fibrosis by inhibiting the activation of AGEs-mediated RAGE/JAK1/STAT3 signaling axis, thus playing a potential role in the treatment of DN.


Diabetic Nephropathies , Glucosides , Janus Kinase 1 , Kidney , Phenols , Receptor for Advanced Glycation End Products , STAT3 Transcription Factor , Signal Transduction , Animals , Mice , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Glucosides/pharmacology , Glucosides/administration & dosage , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Signal Transduction/drug effects , Male , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Phenols/pharmacology , Janus Kinase 1/metabolism , Janus Kinase 1/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Protective Agents/pharmacology , Protective Agents/administration & dosage , Humans , Mice, Inbred C57BL , Blood Glucose/metabolism , Blood Glucose/drug effects
3.
Cell Transplant ; 33: 9636897241254678, 2024.
Article En | MEDLINE | ID: mdl-38798038

Chronic graft-versus-host disease (cGVHD) is a potentially life-threatening complication after allogeneic hematopoietic stem cell transplantation. Standard steroid first-line treatment could not satisfy therapeutic needs due to limited efficacy. As a highly selective Janus kinase (JAK) 1 inhibitor, SHR0302 exhibits a reduced inhibition effect on JAK2 and might have less effect on hematopoiesis. This phase I clinical trial investigated the tolerability and safety of SHR0302 in combination with prednisone, and its early efficacy evidence as a potential first-line treatment to moderate/severe cGVHD. The standard 3 + 3 dose escalation was implemented to find the optimal dose of SHR0302. And prednisone was concurrently administrated with a dose of 1 mg/kg/d and then gradually tapered after 2 weeks. Eighteen patients were enrolled into the study. Grade ≥ 3 treatment-related adverse events were observed in 38.9% of patients. Only one patient developed DLT (grade ≥ 3 hypercholesterolemia) in the highest dose-level group who had pre-existing hypercholesterolemia. The maximum tolerated dose was not reached. No patient discontinued treatment due to AEs. Sixteen out of 18 patients were evaluable for responses, the ORR at week 4 and week 24 were 94.4 and 87.5%, respectively. Overall, the treatment of SHR0302 combined with prednisone was safe and well-tolerated, preliminary clinical results presented a high response for previously untreated cGVHD and a significant reduction in prednisone use in this study. A phase II trial will be conducted to further investigate its therapeutic effects clinically.


Graft vs Host Disease , Janus Kinase 1 , Prednisone , Humans , Graft vs Host Disease/drug therapy , Prednisone/therapeutic use , Male , Female , Adult , Middle Aged , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Chronic Disease , Young Adult , Hematopoietic Stem Cell Transplantation/adverse effects , Aged , Drug Therapy, Combination , Bronchiolitis Obliterans Syndrome
4.
Ann Clin Lab Sci ; 54(2): 156-159, 2024 Mar.
Article En | MEDLINE | ID: mdl-38802165

OBJECTIVE: There has been no significant improvement in remission rate in inflammatory bowel disease (IBD) despite several new drugs being introduced in the past two decades. Post-treatment biopsies sometimes show histologic healing in some areas of the intestine while other areas within the same intestine continue to show active inflammation. The aim of this short descriptive study was to determine whether heterogeneous treatment response in IBD may be caused by heterogeneous expression of treatment targets within the same intestine. METHODS: Six cases of Crohn's disease and five cases of ulcerative colitis in which moderate to severe active inflammation was present in at least two biopsies from the same intestine obtained during the same endoscopy procedure were entered in the study. Sections were stained for TNFα and phospho-JAK1 (p-JAK1) using immunohistochemistry. Expression of TNFα and p-JAK1 was recorded as high when the staining intensity was moderate or high, or low when there was no or week staining. The number of eosinophils per high power field was counted in the area of peak density. RESULTS: Different sites within the same intestine from IBD patients with moderate to severe active inflammation may express different levels of TNFα and p-JAK1. For example, in one patient with Crohn's disease with histologically moderate to severe activity in biopsies from the ileum (site 1) and cecum (site 2), there was high expression of p-JAK1 and low TNFα in the ileum biopsy with the exact opposite in the cecum biopsy (low p-JAK1 and high TNFα expression). In this example neither small molecule drug targeting JAK1 nor anti-TNFα biologic given as single agent therapy would be expected to induce histologic remission in both actively inflamed sites in this patient. CONCLUSIONS: The heterogeneous expression of treatment targets within the same intestine may explain why some patients with IBD may not have complete remission on single drug. Studies are needed to determine whether assay for target expression in mucosal biopsies from IBD patients can help to optimize treatment selection.


Inflammatory Bowel Diseases , Janus Kinase 1 , Tumor Necrosis Factor-alpha , Humans , Tumor Necrosis Factor-alpha/metabolism , Janus Kinase 1/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/drug therapy , Male , Female , Adult , Crohn Disease/metabolism , Crohn Disease/pathology , Crohn Disease/drug therapy , Middle Aged , Biopsy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colitis, Ulcerative/drug therapy
5.
Int J Mol Sci ; 25(10)2024 May 20.
Article En | MEDLINE | ID: mdl-38791600

Lead (Pb) is a common pollutant that is not biodegradable and gravely endangers the environment and human health. Annona squamosa fruit has a wide range of medicinal uses owing to its phytochemical constituents. This study evaluated the effect of treatment with A. squamosa fruit extract (ASFE) on testicular toxicity induced in male rats by lead acetate. The metal-chelating capacity and phytochemical composition of ASFE were determined. The LD50 of ASFE was evaluated by probit analysis. Molecular docking simulations were performed using Auto Dock Vina. Forty male Sprague Dawley rats were equally divided into the following groups: Gp1, a negative control group; Gp2, given ASFE (350 mg/kg body weight (b. wt.)) (1/10 of LD50); Gp3, given lead acetate (PbAc) solution (100 mg/kg b. wt.); and Gp4, given PbAc as in Gp3 and ASFE as in Gp2. All treatments were given by oro-gastric intubation daily for 30 days. Body weight changes, spermatological parameters, reproductive hormone levels, oxidative stress parameters, and inflammatory biomarkers were evaluated, and molecular and histopathological investigations were performed. The results showed that ASFE had promising metal-chelating activity and phytochemical composition. The LD50 of ASFE was 3500 mg/kg b. wt. The docking analysis showed that quercetin demonstrated a high binding affinity for JAK-1 and STAT-3 proteins, and this could make it a more promising candidate for targeting the JAK-1/STAT-3 pathway than others. The rats given lead acetate had defective testicular tissues, with altered molecular, biochemical, and histological features, as well as impaired spermatological characteristics. Treatment with ASFE led to a significant mitigation of these dysfunctions and modulated the JAK-1/STAT-3/SOCS-1 axis in the rats.


Annona , Fruit , Janus Kinase 1 , Molecular Docking Simulation , Organometallic Compounds , Plant Extracts , Rats, Sprague-Dawley , STAT3 Transcription Factor , Signal Transduction , Testis , Animals , Male , Plant Extracts/pharmacology , Plant Extracts/chemistry , STAT3 Transcription Factor/metabolism , Rats , Annona/chemistry , Testis/drug effects , Testis/metabolism , Testis/pathology , Fruit/chemistry , Signal Transduction/drug effects , Janus Kinase 1/metabolism , Oxidative Stress/drug effects
6.
Chem Pharm Bull (Tokyo) ; 72(5): 498-506, 2024 May 25.
Article En | MEDLINE | ID: mdl-38735699

Using (S)-decursinol isolated from root of Angelica gigas Nakai (AGN), we semi-synthesized and evaluated a series of both enantiomerically pure decursin derivatives for their antiproliferative activities against A549 human lung cancer cells. All synthesized compounds showed a broad spectrum of inhibitory activities against the growth of A549 cells. Especially, compound (S)-2d with (E)-(furan-3-yl)acryloyl group showed the most potent activity (IC50: 14.03 µM) against A549 cancer cells as compared with the reference compound, decursin (IC50: 43.55 µM) and its enantiomer, (R)-2d (IC50: 151.59 µM). Western blotting assays indicated that (S)-2d more strongly inhibited Janus kinase 1 (JAK1) and signal transducer and activator of transcription activation 3 (STAT3) phosphorylation than decursin in a dose-dependent manner, while having no effect on CXCR7 overexpression and total STAT3 level. In addition, (S)-2d induced cell cycle arrest at G1 phase and subsequent apoptotic cell death in A549 cancer cells. Our combined analysis of molecular docking studies and biological data suggests that the inhibition of JAK1 with (S)-2d resulted in loss of STAT3 phosphorylation and inhibition of cell growth in A549 cancer cells. These overall results strongly suggest that (S)-2d (MRC-D-004) as a novel JAK1 inhibitor may have therapeutic potential in the treatment of A549 human lung cancers by targeting the JAK1/STAT3 signaling pathway.


Apoptosis , Benzopyrans , Butyrates , Cell Proliferation , Drug Screening Assays, Antitumor , Molecular Docking Simulation , STAT3 Transcription Factor , Humans , Cell Proliferation/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Benzopyrans/pharmacology , Benzopyrans/chemistry , Benzopyrans/chemical synthesis , Butyrates/pharmacology , Butyrates/chemistry , Butyrates/chemical synthesis , Apoptosis/drug effects , A549 Cells , Stereoisomerism , Dose-Response Relationship, Drug , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Molecular Structure , Angelica/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry
7.
Cell Rep ; 43(5): 114202, 2024 May 28.
Article En | MEDLINE | ID: mdl-38733583

Interleukin-6 (IL-6)-class inflammatory cytokines signal through the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription (STAT) pathway and promote the development of pancreatic ductal adenocarcinoma (PDAC); however, the functions of specific intracellular signaling mediators in this process are less well defined. Using a ligand-controlled and pancreas-specific knockout in adult mice, we demonstrate in this study that JAK1 deficiency prevents the formation of KRASG12D-induced pancreatic tumors, and we establish that JAK1 is essential for the constitutive activation of STAT3, whose activation is a prominent characteristic of PDAC. We identify CCAAT/enhancer binding protein δ (C/EBPδ) as a biologically relevant downstream target of JAK1 signaling, which is upregulated in human PDAC. Reinstating the expression of C/EBPδ was sufficient to restore the growth of JAK1-deficient cancer cells as tumorspheres and in xenografted mice. Collectively, the findings of this study suggest that JAK1 executes important functions of inflammatory cytokines through C/EBPδ and may serve as a molecular target for PDAC prevention and treatment.


Carcinoma, Pancreatic Ductal , Janus Kinase 1 , Pancreatic Neoplasms , STAT3 Transcription Factor , Animals , Janus Kinase 1/metabolism , Janus Kinase 1/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Humans , Mice , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , STAT3 Transcription Factor/metabolism , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Disease Progression , Signal Transduction , Cell Line, Tumor , Mice, Knockout
8.
Cytokine ; 179: 156620, 2024 07.
Article En | MEDLINE | ID: mdl-38701735

PURPOSE: The emergence of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, but these drugs can also cause severe immune-related adverse effects (irAEs), including myocarditis. Researchers have become interested in exploring ways to mitigate this side effect, and one promising avenue is the use of baricitinib, a Janus kinase inhibitor known to have anti-inflammatory properties. This study aimed to examine the potential mechanism by which baricitinib in ICIs-related myocarditis. METHODS: To establish an ICIs-related myocarditis model, BALB/c mice were administered murine cardiac troponin I (cTnI) peptide and anti-mouse programmed death 1 (PD-1) antibodies. Subsequently, baricitinib was administered to the mice via intragastric administration. Echocardiography, HE staining, and Masson staining were performed to evaluate myocardial functions, inflammation, and fibrosis. Immunofluorescence was used to detect macrophages in the cardiac tissue of the mice.In vitro experiments utilized raw264.7 cells to induce macrophage polarization using anti-PD-1 antibodies. Different concentrations of baricitinib were applied to assess cell viability, and the release of pro-inflammatory cytokines was measured. The activation of the JAK1/STAT3 signaling pathway was evaluated through western blot analysis. RESULTS: Baricitinib demonstrated its ability to improve cardiac function and reduce cardiac inflammation, as well as fibrosis induced by ICIs. Mechanistically, baricitinib treatment promoted the polarization of macrophages towards the M2 phenotype. In vitro and in vivo experiments showed that anti-PD-1 promoted the release of inflammatory factors. However, treatment with baricitinib significantly inhibited the phosphorylation of JAK1 and STAT3. Additionally, the use of RO8191 reversed the effects of baricitinib, further confirming our findings. CONCLUSION: Baricitinib demonstrated its potential as a protective agent against ICIs-related myocarditis by modulating macrophage polarization. These findings provide a solid theoretical foundation for the development of future treatments for ICIs-related myocarditis.


Azetidines , Janus Kinase 1 , Macrophages , Mice, Inbred BALB C , Myocarditis , Purines , Pyrazoles , STAT3 Transcription Factor , Sulfonamides , Animals , Male , Mice , Azetidines/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Janus Kinase 1/metabolism , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Myocarditis/chemically induced , Myocarditis/drug therapy , Myocarditis/pathology , Myocarditis/metabolism , Purines/pharmacology , Pyrazoles/pharmacology , RAW 264.7 Cells , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Troponin I/metabolism
9.
Nat Commun ; 15(1): 4484, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802340

Deciphering the intricate dynamic events governing type I interferon (IFN) signaling is critical to unravel key regulatory mechanisms in host antiviral defense. Here, we leverage TurboID-based proximity labeling coupled with affinity purification-mass spectrometry to comprehensively map the proximal human proteomes of all seven canonical type I IFN signaling cascade members under basal and IFN-stimulated conditions. This uncovers a network of 103 high-confidence proteins in close proximity to the core members IFNAR1, IFNAR2, JAK1, TYK2, STAT1, STAT2, and IRF9, and validates several known constitutive protein assemblies, while also revealing novel stimulus-dependent and -independent associations between key signaling molecules. Functional screening further identifies PJA2 as a negative regulator of IFN signaling via its E3 ubiquitin ligase activity. Mechanistically, PJA2 interacts with TYK2 and JAK1, promotes their non-degradative ubiquitination, and limits the activating phosphorylation of TYK2 thereby restraining downstream STAT signaling. Our high-resolution proximal protein landscapes provide global insights into the type I IFN signaling network, and serve as a valuable resource for future exploration of its functional complexities.


Interferon Type I , Janus Kinase 1 , Receptor, Interferon alpha-beta , STAT2 Transcription Factor , Signal Transduction , TYK2 Kinase , Ubiquitination , Humans , HEK293 Cells , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Janus Kinase 1/metabolism , Phosphorylation , Proteome/metabolism , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , TYK2 Kinase/metabolism , Ubiquitin-Protein Ligases/metabolism
10.
Birth Defects Res ; 116(5): e2345, 2024 May.
Article En | MEDLINE | ID: mdl-38716582

BACKGROUND: Abrocitinib is a Janus kinase (JAK) 1 selective inhibitor approved for the treatment of atopic dermatitis. Female reproductive tissues were unaffected in general toxicity studies, but an initial female rat fertility study resulted in adverse effects at all doses evaluated. A second rat fertility study was conducted to evaluate lower doses and potential for recovery. METHODS: This second study had 4 groups of 20 females each administered abrocitinib (0, 3, 10, or 70 mg/kg/day) 2 weeks prior to cohabitation through gestation day (GD) 7. In addition, 2 groups of 20 rats (0 or 70 mg/kg/day) were dosed for 3 weeks followed by a 4-week recovery period before mating. All mated females were evaluated on GD 14. RESULTS: No effects were observed at ≤10 mg/kg/day. At 70 mg/kg/day (29x human exposure), decreased pregnancy rate, implantation sites, and viable embryos were observed. All these effects reversed 4 weeks after the last dose. CONCLUSIONS: Based on these data and literature on the potential role of JAK signaling in implantation, we hypothesize that these effects may be related to JAK1 inhibition and, generally, that peri-implantation effects such as these, in the absence of cycling or microscopic changes in nonpregnant female reproductive tissues, are anticipated to be reversible.


Fertility , Janus Kinase 1 , Pyrimidines , Sulfonamides , Female , Animals , Pregnancy , Rats , Fertility/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Rats, Sprague-Dawley , Embryo Implantation/drug effects , Janus Kinase Inhibitors/pharmacology , Pregnancy Rate
11.
J Ethnopharmacol ; 329: 118138, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38565410

ETHNOPHARMACOLOGICAL RELEVANCE: Phoenix dactylifera L. (date palm) seed is widely used in Arabian traditional medicine to alleviate several health problems including inflammatory conditions. The herbal tea of date palm seed has been consumed by rheumatoid patients to relief their symptoms. AIM OF THE STUDY: The purpose of this study was to investigate the claimed beneficial use of P. dactylifera L. (Sewy variety) seed (PDS) in the treatment of rheumatoid arthritis (RA) and its mechanism of action as well as to study its phytoconstituents. MATERIALS AND METHODS: The anti-inflammatory and anti-oxidative properties of the non-polar and the polar extracts of PDS were studied using Complete Freund's adjuvant (CFA)-induced arthritis rat model. Paw edema, body weight, total nitrate/nitrite NOX content and cytokine markers were evaluated to monitor the progress of arthritis. Also, histological examination and thermal analysis were conducted. The phytoconstituent profiles of non-polar and polar extracts of PDS were investigated using liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). The multiple reactions monitoring mode (MRM) of liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) was used to quantify phenolic phytoconstituents in both extracts. RESULTS: According to the findings, the polar and non-polar PDS extracts kept body weight comparable to those of healthy individuals while considerably lowering paw swelling, edema, and neutrophil infiltration. It also reduced the levels of Nuclear Factor Kappa B (NF-κB), Tumor Necrosis Factor Alpha (TNF-α), Interleukin 22, Interleukin 23, Interferon (IFN), Interleukin 17, Interleukin 1ß, Interleukin 6, Interleukin 36, Janus Kinase 1 (JAK1), and Signal Transducer and Activator of Transcription 3 (STAT3). They also reduced the degenerative alterations caused by RA. Thermal research gave additional support for these findings. 83 phytoconstituents were identified in the non-polar PDS extract and 86 phytoconstituents were identified in the polar PDS extract. 74 of the identified phytoconstituents were common in both extracts. 33 phytoconstituents were identified here from P. dactylifera for the first time as far as we know. In MRM-LC-ESI-MS/MS analysis, the major phenolics in both extracts were chlorogenic acid, naringenin, and vanillin. Catechin was only detected in the non-polar PDS extract. On the other hand, apigenin, kaempferol, and hesperetin were only detected in the polar PDS extract. Generally, the polar PDS extract showed higher concentrations of the identified phenolics than the non-polar extract. CONCLUSIONS: The PDS extracts especially the non-polar extract showed significant anti-inflammatory and anti-oxidative properties in the CFA-induced arthritis rat model. PDS might be used to produce RA medicines.


Anti-Inflammatory Agents , Arthritis, Experimental , Cytokines , Freund's Adjuvant , Janus Kinase 1 , Phoeniceae , Plant Extracts , STAT3 Transcription Factor , Seeds , Animals , Phoeniceae/chemistry , STAT3 Transcription Factor/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Plant Extracts/pharmacology , Plant Extracts/chemistry , Janus Kinase 1/metabolism , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Seeds/chemistry , Male , Antirheumatic Agents/pharmacology , Antirheumatic Agents/isolation & purification , Rats , Phytochemicals/analysis , Phytochemicals/pharmacology , Signal Transduction/drug effects , Rats, Wistar , Rats, Sprague-Dawley , Antioxidants/pharmacology
12.
Phys Chem Chem Phys ; 26(17): 13420-13431, 2024 May 01.
Article En | MEDLINE | ID: mdl-38647171

Autoimmune inflammatory diseases, such as rheumatoid arthritis (RA) and ulcerative colitis, are associated with an uncontrolled production of cytokines leading to the pronounced inflammatory response of these disorders. Their therapy is currently focused on the inhibition of cytokine receptors, such as the Janus kinase (JAK) protein family. Tofacitinib and peficitinib are JAK inhibitors that have been recently approved to treat rheumatoid arthritis. In this study, an in-depth analysis was carried out through quantum biochemistry to understand the interactions involved in the complexes formed by JAK1 and tofacitinib or peficitinib. Computational analyses provided new insights into the binding mechanisms between tofacitinib or peficitinib and JAK1. The essential amino acid residues that support the complex are also identified and reported. Additionally, we report new interactions, such as van der Waals; hydrogen bonds; and alkyl, pi-alkyl, and pi-sulfur forces, that stabilize the complexes. The computational results revealed that peficitinib presents a similar affinity to JAK1 compared to tofacitinib based on their interaction energies.


Adamantane/analogs & derivatives , Janus Kinase 1 , Niacinamide , Niacinamide/analogs & derivatives , Piperidines , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Piperidines/therapeutic use , Niacinamide/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 1/chemistry , Humans , Quantum Theory , Autoimmune Diseases/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Hydrogen Bonding , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Adamantane/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Molecular Docking Simulation
13.
J Ethnopharmacol ; 330: 118223, 2024 Aug 10.
Article En | MEDLINE | ID: mdl-38642624

ETHNOPHARMACOLOGICAL RELEVANCE: Leonurus japonicus Houtt. (Labiatae), commonly known as Chinese motherwort, is a herbaceous flowering plant that is native to Asia. It is widely acknowledged in traditional medicine for its diuretic, hypoglycemic, antiepileptic properties and neuroprotection. Currently, Leonurus japonicus (Leo) is included in the Pharmacopoeia of the People's Republic of China. Traditional Chinese Medicine (TCM) recognizes Leo for its myriad pharmacological attributes, but its efficacy against ICH-induced neuronal apoptosis is unclear. AIMS OF THE STUDY: This study aimed to identify the potential targets and regulatory mechanisms of Leo in alleviating neuronal apoptosis after ICH. MATERIALS AND METHODS: The study employed network pharmacology, UPLC-Q-TOF-MS technique, molecular docking, pharmacodynamic studies, western blotting, and immunofluorescence techniques to explore its potential mechanisms. RESULTS: Leo was found to assist hematoma absorption, thus improving the neurological outlook in an ICH mouse model. Importantly, molecular docking highlighted JAK as Leo's potential therapeutic target in ICH scenarios. Further experimental evidence demonstrated that Leo adjusts JAK1 and STAT1 phosphorylation, curbing Bax while augmenting Bcl-2 expression. CONCLUSION: Leo showcases potential in mitigating neuronal apoptosis post-ICH, predominantly via the JAK/STAT mechanism.


Apoptosis , Cerebral Hemorrhage , Leonurus , Molecular Docking Simulation , Network Pharmacology , Neurons , Animals , Apoptosis/drug effects , Leonurus/chemistry , Neurons/drug effects , Neurons/metabolism , Mice , Male , Cerebral Hemorrhage/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/chemistry , Janus Kinase 1/metabolism , STAT1 Transcription Factor/metabolism , Disease Models, Animal
14.
Molecules ; 29(8)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38675621

Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly effective, well-established treatment for patients with various hematologic malignancies and non-malignant diseases. The therapeutic benefits of allo-HCT are mediated by alloreactive T cells in donor grafts. However, there is a significant risk of graft-versus-host disease (GvHD), in which the donor T cells recognize recipient cells as foreign and attack healthy organs in addition to malignancies. We previously demonstrated that targeting JAK1/JAK2, mediators of interferon-gamma receptor (IFNGR) and IL-6 receptor signaling, in donor T cells using baricitinib and ruxolitinib results in a significant reduction in GvHD after allo-HCT. Furthermore, we showed that balanced inhibition of JAK1/JAK2 while sparing JAK3 is important for the optimal prevention of GvHD. Thus, we have generated novel JAK1/JAK2 inhibitors, termed WU derivatives, by modifying baricitinib. Our results show that WU derivatives have the potential to mitigate GvHD by upregulating regulatory T cells and immune reconstitution while reducing the frequencies of antigen-presenting cells (APCs) and CD80 expression on these APCs in our preclinical mouse model of allo-HCT. In addition, WU derivatives effectively downregulated CXCR3 and T-bet in primary murine T cells. In summary, we have generated novel JAK inhibitors that could serve as alternatives to baricitinib or ruxolitinib.


Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Pyrazoles , Transplantation, Homologous , Animals , Mice , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Azetidines/pharmacology , Disease Models, Animal , Graft vs Host Disease/prevention & control , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Mice, Inbred C57BL , Purines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects
15.
J Exp Clin Cancer Res ; 43(1): 67, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38429845

BACKGROUND: Docetaxel resistance represents a significant obstacle in the treatment of prostate cancer. The intricate interplay between cytokine signalling pathways and transcriptional control mechanisms in cancer cells contributes to chemotherapeutic resistance, yet the underlying molecular determinants remain only partially understood. This study elucidated a novel resistance mechanism mediated by the autocrine interaction of interleukin-11 (IL-11) and its receptor interleukin-11 receptor subunit alpha(IL-11RA), culminating in activation of the JAK1/STAT4 signalling axis and subsequent transcriptional upregulation of the oncogene c-MYC. METHODS: Single-cell secretion profiling of prostate cancer organoid was analyzed to determine cytokine production profiles associated with docetaxel resistance.Analysis of the expression pattern of downstream receptor IL-11RA and enrichment of signal pathway to clarify the potential autocrine mechanism of IL-11.Next, chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) was performed to detect the nuclear localization and DNA-binding patterns of phosphorylated STAT4 (pSTAT4). Coimmunoprecipitation and reporter assays were utilized to assess interaction between pSTAT4 and the cotranscription factor CREB-binding protein (CBP) as well as their role in c-MYC transcriptional activity. RESULTS: Autocrine secretion of IL-11 was markedly increased in docetaxel-resistant prostate cancer cells. IL-11 stimulation resulted in robust activation of JAK1/STAT4 signalling. Upon activation, pSTAT4 translocated to the nucleus and associated with CBP at the c-MYC promoter region, amplifying its transcriptional activity. Inhibition of the IL-11/IL-11RA interaction or disruption of the JAK1/STAT4 pathway significantly reduced pSTAT4 nuclear entry and its binding to CBP, leading to downregulation of c-MYC expression and restoration of docetaxel sensitivity. CONCLUSION: Our findings identify an autocrine loop of IL-11/IL-11RA that confers docetaxel resistance through the JAK1/STAT4 pathway. The pSTAT4-CBP interaction serves as a critical enhancer of c-MYC transcriptional activity in prostate cancer cells. Targeting this signalling axis presents a potential therapeutic strategy to overcome docetaxel resistance in advanced prostate cancer.


Drug Resistance, Neoplasm , Interleukin-11 , Prostatic Neoplasms , Humans , Male , Docetaxel/pharmacology , Gene Expression Regulation , Interleukin-11/genetics , Interleukin-11/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Signal Transduction , STAT4 Transcription Factor/metabolism , Drug Resistance, Neoplasm/genetics
16.
Int J Mol Sci ; 25(5)2024 Mar 04.
Article En | MEDLINE | ID: mdl-38474223

The Janus kinase (JAK) family is a small group of protein tyrosine kinases that represent a central component of intracellular signaling downstream from a myriad of cytokine receptors. The JAK3 family member performs a particularly important role in facilitating signal transduction for a key set of cytokine receptors that are essential for immune cell development and function. Mutations that impact JAK3 activity have been identified in a number of human diseases, including somatic gain-of-function (GOF) mutations associated with immune cell malignancies and germline loss-of-function (LOF) mutations associated with immunodeficiency. The structure, function and impacts of both GOF and LOF mutations of JAK3 are highly conserved, making animal models highly informative. This review details the biology of JAK3 and the impact of its perturbation in immune cell-related diseases, including relevant animal studies.


Immunologic Deficiency Syndromes , Neoplasms , Animals , Humans , Janus Kinase 3/metabolism , Signal Transduction , Janus Kinases/metabolism , Receptors, Cytokine/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism
18.
J Biol Chem ; 300(3): 105779, 2024 Mar.
Article En | MEDLINE | ID: mdl-38395305

The newly discovered zoonotic coronavirus swine acute diarrhea syndrome coronavirus (SADS-CoV) causes acute diarrhea, vomiting, dehydration, and high mortality rates in newborn piglets. Although SADS-CoV uses different strategies to evade the host's innate immune system, the specific mechanism(s) by which it blocks the interferon (IFN) response remains unidentified. In this study, the potential of SADS-CoV nonstructural proteins (nsp) to inhibit the IFN response was detected. The results determined that nsp1 was a potent antagonist of IFN response. SADS-CoV nsp1 efficiently inhibited signal transducer and activator of transcription 1 (STAT1) phosphorylation by inducing Janus kinase 1 (JAK1) degradation. Subsequent research revealed that nsp1 induced JAK1 polyubiquitination through K11 and K48 linkages, leading to JAK1 degradation via the ubiquitin-proteasome pathway. Furthermore, SADS-CoV nsp1 induced CREB-binding protein degradation to inhibit IFN-stimulated gene production and STAT1 acetylation, thereby inhibiting STAT1 dephosphorylation and blocking STAT1 transport out of the nucleus to receive antiviral signaling. In summary, the results revealed the novel mechanisms by which SADS-CoV nsp1 blocks the JAK-STAT signaling pathway via the ubiquitin-proteasome pathway. This study yielded valuable findings on the specific mechanism of coronavirus nsp1 in inhibiting the JAK-STAT signaling pathway and the strategies of SADS-CoV in evading the host's innate immune system.


Alphacoronavirus , Coronavirus Infections , Proteasome Endopeptidase Complex , Swine Diseases , Viral Nonstructural Proteins , Animals , Acetylation , Alphacoronavirus/physiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Swine , Ubiquitins/metabolism , Swine Diseases/metabolism , Swine Diseases/virology , HEK293 Cells , Vero Cells , Humans , Chlorocebus aethiops , Viral Nonstructural Proteins/metabolism
19.
J Pharmacol Exp Ther ; 389(1): 40-50, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38336380

B-cell acute lymphoblastic leukemia (B-ALL) is the most prevalent type of cancer in young children and is associated with high levels of reactive oxygen species (ROS). The antioxidant N-acetylcysteine (NAC) was tested for its ability to alter disease progression in a mouse model of B-ALL. Mb1-CreΔPB mice have deletions in genes encoding PU.1 and Spi-B in B cells and develop B-ALL at 100% incidence. Treatment of Mb1-CreΔPB mice with NAC in drinking water significantly reduced the frequency of CD19+ pre-B-ALL cells infiltrating the thymus at 11 weeks of age. However, treatment with NAC did not reduce leukemia progression or increase survival by a median 16 weeks of age. NAC significantly altered gene expression in leukemias in treated mice. Mice treated with NAC had increased frequencies of activating mutations in genes encoding Janus kinases 1 and 3. In particular, frequencies of Jak3 R653H mutations were increased in mice treated with NAC compared with control drinking water. NAC opposed oxidization of PTEN protein ROS in cultured leukemia cells. These results show that NAC alters leukemia progression in this mouse model, ultimately selecting for leukemias with high Jak3 R653H mutation frequencies. SIGNIFICANCE STATEMENT: In a mouse model of precursor B-cell acute lymphoblastic leukemia associated with high levels of reactive oxygen species, treatment with N-acetylcysteine did not delay disease progression but instead selected for leukemic clones with activating R653H mutations in Janus kinase 3.


Drinking Water , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Mice , Animals , Child, Preschool , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Janus Kinases , Mutation Rate , Reactive Oxygen Species/metabolism , Precursor Cells, B-Lymphoid/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Mutation , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Disease Progression
20.
Oncotarget ; 15: 65-75, 2024 Feb 05.
Article En | MEDLINE | ID: mdl-38319731

Multiple myeloma (MM) is the most common primary malignancy of the bone marrow. No established curative treatment is currently available for patients diagnosed with MM. In recent years, new and more effective drugs have become available for the treatment of this B-cell malignancy. These new drugs have often been evaluated together and in combination with older agents. However, even these novel combinations eventually become ineffective; and, thus, novel therapeutic approaches are necessary to help overcome resistance to these treatments. Recently, the Janus Kinase (JAK) family of tyrosine kinases, specifically JAK1 and JAK2, has been shown to have a role in the pathogenesis of MM. Preclinical studies have demonstrated a role for JAK signaling in direct and indirect growth of MM and downregulation of anti-tumor immune responses in these patients. Also, inhibition of JAK proteins enhances the anti-MM effects of other drugs used to treat MM. These findings have been confirmed in clinical studies which have further demonstrated the safety and efficacy of JAK inhibition as a means to overcome resistance to currently available anti-MM therapies. Additional studies will provide further support for this promising new therapeutic approach for treating patients with MM.


Janus Kinase Inhibitors , Multiple Myeloma , Pyrazoles , Humans , Multiple Myeloma/pathology , Janus Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Nitriles/therapeutic use , Janus Kinases/metabolism , Janus Kinase 2/metabolism , Janus Kinase 1/metabolism , Protein Kinase Inhibitors/pharmacology
...