Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 391
Filter
1.
Elife ; 132024 Oct 14.
Article in English | MEDLINE | ID: mdl-39401071

ABSTRACT

Current treatments for congenital and acquired craniofacial (CF) bone abnormalities are limited and costly. Conventional methods involve surgical correction, short-term stabilization, and long-term bone grafting, which may include problematic allografts and limited autografts. While bone morphogenetic protein 2 (BMP2) has been used for bone regeneration, it can cause bone overgrowth and life-threatening inflammation. Bone marrow-derived mesenchymal stem cell therapies, though promising, are not Food and Drug Administration approved and are resource intensive. Thus, there is a need for effective, affordable, and less side-effect-prone bone regenerative therapies. Previous research demonstrated that JAGGED1 induces osteoblast commitment in murine cranial neural crest cells through a NOTCH-dependent non-canonical pathway involving JAK2-STAT5. We hypothesize that delivery of JAGGED1 and induction of its downstream NOTCH non-canonical signaling in pediatric human osteoblasts constitutes an effective bone regenerative treatment. Delivering pediatric human bone-derived osteoblast-like cells to an in vivo murine bone loss model of a critically sized cranial defect, we identified that JAGGED1 promotes human pediatric osteoblast commitment and bone formation through p70 S6K phosphorylation. This approach highlights the potential of JAGGED1 and its downstream activators as innovative treatments for pediatric CF bone loss.


Subject(s)
Bone Regeneration , Jagged-1 Protein , Osteoblasts , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Humans , Animals , Mice , Osteoblasts/physiology , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Osteogenesis , Child , Craniofacial Abnormalities/therapy , Disease Models, Animal
2.
Int J Mol Sci ; 25(19)2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39408800

ABSTRACT

Localized scleroderma (LS) is an autoimmune disease characterized by inflammation and fibrosis, leading to severe cutaneous manifestations such as skin hardening, tightness, discoloration, and other textural changes that may result in disability. While LS shares similar histopathologic features and immune-fibroblast interactions with systemic sclerosis (SSc), its molecular mechanisms remain understudied. Endothelial cells (EC) are known to play a crucial role in SSc but have not been investigated in LS. Single-cell RNA sequencing (scRNA-seq) now allows for detailed examination of this cell type in the primary organ of interest for scleroderma, the skin. In this study, we analyzed skin-isolated cells from 27 LS patients (pediatric and adult) and 17 healthy controls using scRNA-seq. Given the known role of EC damage as an initial event in SSc and the histologic and clinical skin similarities to LS, we focused primarily on endothelial cells. Our analysis identified eight endothelial subclusters within the dataset, encompassing both disease and healthy samples. Interaction analysis revealed that signaling from diseased endothelial cells was predicted to promote fibrosis through SELE interaction with FGFBP1 and other target genes. We also observed high levels of JAG in arterial endothelial cells and NOTCH in capillary endothelial cells, indicating the activation of a signaling pathway potentially responsible for epidermal abnormalities and contributing to LS pathogenesis. In summary, our scRNA-seq analysis identified potential disease-propagating endothelial cell clusters with upregulated pathways in LS skin, highlighting their importance in disease progression.


Subject(s)
Endothelial Cells , Receptors, Notch , Scleroderma, Localized , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Skin , Humans , Endothelial Cells/metabolism , Endothelial Cells/pathology , Receptors, Notch/metabolism , Receptors, Notch/genetics , Scleroderma, Localized/metabolism , Scleroderma, Localized/pathology , Scleroderma, Localized/genetics , Adult , Skin/metabolism , Skin/pathology , Female , Male , Child , Adolescent , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics
3.
Int J Mol Sci ; 25(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39408921

ABSTRACT

Breast cancer is the most common cancer in women with multiple risk factors including smoking, genetics, environmental factors, and obesity. Smoking and obesity are the top two risk factors for the development of breast cancer. The effect of obesity on adipose tissue mediates the pathogenesis of breast cancer in the context of obesity. Triple-negative breast cancer (TNBC) is a breast cancer subtype within which the cells lack estrogen, progesterone, and HER2 receptors. TNBC is the deadliest breast cancer subtype. The 5-year survival rates for patients with TNBC are 8-16% lower than the 5-year survival rates for patients with estrogen-receptor-positive breast tumors. In addition, TNBC patients have early relapse rates (3-5 years after diagnosis). Obesity is associated with an increased risk for TNBC, larger TNBC tumors, and increased breast cancer metastasis compared with lean women. Thus, novel therapeutic approaches are warranted to treat TNBC in the context of obesity. In this paper, we show that peritumor breast adipose-derived secretome (ADS) from patients with a high (>30) BMI is a stronger inducer of TNBC cell invasiveness and JAG1 expression than peritumor breast ADS from patients with low (<30) BMI. These findings indicate that patient BMI-associated changes in peritumor AT induce changes in peritumor ADS, which in turn acts on TNBC cells to stimulate JAG1 expression and cancer cell invasiveness.


Subject(s)
Adipose Tissue , Body Mass Index , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Female , Adipose Tissue/metabolism , Adipose Tissue/pathology , Neoplasm Invasiveness , Cell Line, Tumor , Obesity/metabolism , Obesity/complications , Obesity/pathology , Middle Aged , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics
4.
Cell Mol Biol (Noisy-le-grand) ; 70(8): 96-103, 2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39262258

ABSTRACT

Resveratrol, a potent anticancer bioactive compound, has been shown to trigger apoptosis in numerous cancer cells. Although Notch signaling promotes breast cancer apoptosis, it is unclear whether resveratrol induces apoptosis in MCF-7 cells via influencing the Notch pathway. This study aimed to evaluate the effect of resveratrol on modulating Notch signaling targets and provide critical information for employing resveratrol in breast cancer therapy. Thus, in this study, we have deciphered the effect of resveratrol against three potent genes (Notch1, Jagged1, and DLL4) of the notch signaling pathway. For mechanistic studies, in silico, and in vitro analysis was executed to investigate the apoptotic-inducing potential of resveratrol against three selected oncogenes involved in the progression of breast cancer. Docking analysis revealed the inhibitory potential of resveratrol against all three selected targets of the Notch pathway (Notch1: -5.0; Jagged-1: -5.9; DLL4: -5.8). In vitro, findings further displayed a significant reduction in cell viability in resveratrol-treated MCF-7 cancer cells, which were concomitantly related to the downregulation of Notch-1, Jagged-1, and DLL4. Moreover, the antiproliferative efficacy of resveratrol was correlated with apoptosis and modulation in the expression of Bax, Bcl-2, cyclin D1, CDK4, p21, and caspase-3 activation. Taken together, these experimental findings suggested that apoptotic inducing potential of resveratrol was mediated through a novel mechanism involving suppression of the Notch signaling pathway.


Subject(s)
Apoptosis , Breast Neoplasms , Jagged-1 Protein , Resveratrol , Signal Transduction , Humans , Resveratrol/pharmacology , Apoptosis/drug effects , MCF-7 Cells , Signal Transduction/drug effects , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Stilbenes/pharmacology , Receptors, Notch/metabolism , Receptors, Notch/genetics , Molecular Docking Simulation , Cell Survival/drug effects , Caspase 3/metabolism , Caspase 3/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
5.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39254648

ABSTRACT

During embryonic development, Wnt signaling influences both proliferation and sensory formation in the cochlea. How this dual nature of Wnt signaling is coordinated is unknown. In this study, we define a novel role for a Wnt-regulated gene, Mybl2, which was already known to be important for proliferation, in determining the size and patterning of the sensory epithelium in the murine cochlea. Using a quantitative spatial analysis approach and analyzing Mybl2 loss-of-function, we show that Mybl2 promoted proliferation in the inner sulcus domain but limited the size of the sensory domain by influencing their adjoining boundary position via Jag1 regulation during development. Mybl2 loss-of-function simultaneously decreased proliferation in the inner sulcus and increased the size of the sensory domain, resulting in a wider sensory epithelium with ectopic inner hair cell formation during late embryonic stages. These data suggest that progenitor cells in the inner sulcus determine boundary formation and pattern the sensory epithelium via MYBL2.


Subject(s)
Cell Proliferation , Cochlea , Jagged-1 Protein , Stem Cells , Animals , Cochlea/embryology , Cochlea/cytology , Cochlea/metabolism , Mice , Epithelium/embryology , Epithelium/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Gene Expression Regulation, Developmental , Wnt Signaling Pathway , Body Patterning/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/cytology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
6.
Sci Rep ; 14(1): 20999, 2024 09 09.
Article in English | MEDLINE | ID: mdl-39251686

ABSTRACT

Skin-resident regulatory T cells (Tregs) play an irreplaceable role in orchestrating cutaneous immune homeostasis and repair, including the promotion of hair regeneration via the Notch signaling ligand Jagged-1 (Jag1). While skin Tregs are indispensable for facilitating tissue repair post-wounding, it remains unknown if Jag1-expressing skin Tregs impact wound healing. Using a tamoxifen inducible Foxp3creERT2Jag1fl/fl model, we show that loss of functional Jag1 in Tregs significantly delays the rate of full-thickness wound closure. Unlike in hair regeneration, skin Tregs do not utilize Jag1 to impact epithelial stem cells during wound healing. Instead, mice with Treg-specific Jag1 ablation exhibit a significant reduction in Ly6G + neutrophil accumulation at the wound site. However, during both homeostasis and wound healing, the loss of Jag1 in Tregs does not impact the overall abundance or activation profile of immune cell targets in the skin, such as CD4+ and CD8+ T cells, or pro-inflammatory macrophages. This collectively suggests that skin Tregs may utilize Jag1-Notch signalling to co-ordinate innate cell recruitment under conditions of injury but not homeostasis. Overall, our study demonstrates the importance of Jag1 expression in Tregs to facilitate adequate wound repair in the skin.


Subject(s)
Jagged-1 Protein , Skin , T-Lymphocytes, Regulatory , Wound Healing , Animals , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Wound Healing/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Mice , Skin/metabolism , Skin/immunology , Signal Transduction , Receptors, Notch/metabolism
7.
Nat Commun ; 15(1): 7424, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198465

ABSTRACT

In fetal development, tissue interaction such as the interplay between blood vessel (BV) and epithelial tissue is crucial for organogenesis. Here we recapitulate the spatial arrangement between liver epithelial tissue and the portal vein to observe the formation of intrahepatic bile ducts (BDs) from human induced pluripotent stem cells (hiPSC). We co-culture hiPSC-liver progenitors on the artificial BV consisting of immature smooth muscle cells and endothelial cells, both derived from hiPSCs. After 3 weeks, liver progenitors within hiPSC-BV-incorporated liver organoids (BVLO) differentiate to cholangiocytes and acquire epithelial characteristics, including intercellular junctions, microvilli on the apical membrane, and secretory functions. Furthermore, liver surface transplanted-BVLO temporarily attenuates cholestatic injury symptoms. Single cell RNA sequence analysis suggests that BD interact with the BV in BVLO through TGFß and Notch pathways. Knocking out JAG1 in hiPSC-BV significantly attenuates bile duct formation, highlighting BVLO potential as a model for Alagille syndrome, a congenital biliary disease. Overall, we develop a novel 3D co-culture method that successfully establishes functional human BDs by emulating liver epithelial-BV interaction.


Subject(s)
Cell Differentiation , Coculture Techniques , Induced Pluripotent Stem Cells , Jagged-1 Protein , Liver , Organoids , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Organoids/cytology , Liver/cytology , Liver/metabolism , Liver/blood supply , Coculture Techniques/methods , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Animals , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Mice , Receptors, Notch/metabolism , Receptors, Notch/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Bile Ducts/cytology , Bile Ducts/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Transforming Growth Factor beta/metabolism
8.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125657

ABSTRACT

Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.


Subject(s)
Cardiovascular Diseases , Metformin , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Humans , Male , Female , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Adult , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Inflammation/genetics , Inflammation/drug therapy , Inflammation/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Middle Aged , Gene Expression Regulation/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Biomarkers , Case-Control Studies
9.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201406

ABSTRACT

Sensorineural hearing loss can be caused by lesions to the inner ear during development. Understanding the events and signaling pathways that drive inner ear formation is crucial for determining the possible causes of congenital hearing loss. We have analyzed the innervation and expression of SOX2, JAGGED1, ß-catenin (CTNNB1), and vitamin D receptor (VDR) in the inner ears of human conceptuses aged 5 to 10 weeks after fertilization (W) using immunohistochemistry. The prosensory domains of the human inner ear displayed SOX2 and JAGGED1 expression throughout the analyzed period, with SOX2 expression being more extensive in all the analyzed timepoints. Innervation of vestibular prosensory domains was present at 6 W and extensive at 10 W, while nerve fibers reached the base of the cochlear prosensory domain at 7-8 W. CTNNB1 and VDR expression was mostly membranous and present during all analyzed timepoints in the inner ear, being the strongest in the non-sensory epithelium. Their expression was stronger in the vestibular region compared to the cochlear duct. CTNNB1 and VDR expression displayed opposite expression trends during the analyzed period, but additional studies are needed to elucidate whether they interact during inner ear development.


Subject(s)
Ear, Inner , Jagged-1 Protein , Receptors, Calcitriol , SOXB1 Transcription Factors , beta Catenin , Humans , beta Catenin/metabolism , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Ear, Inner/metabolism , Ear, Inner/innervation , Ear, Inner/embryology , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Gene Expression Regulation, Developmental , Female
10.
Clin Transl Med ; 14(8): e1793, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39113232

ABSTRACT

INTRODUCTION: Liver fibrosis is primarily driven by the activation of hepatic stellate cells (HSCs), which involves various epigenetic modifications. OBJECTIVES: N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotic cells, influences numerous physiological and pathological processes. Nevertheless, the role of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader gene mediating m6A modifications, in liver fibrosis remains unclear. METHODS AND RESULTS: This study demonstrated that IGF2BP3 knockout reduces liver fibrosis by promoting HSC ferroptosis (FPT) and inactivating HSCs. Multi-omics analysis revealed that HSC-specific IGF2BP3 knockout decreased m6A content in Jagged1 (Jag1), a key component of the Notch signalling pathway. Furthermore, IGF2BP3 deficiency significantly reduced the expression of hairy and enhancer of split-1 (Hes1), a transcription factor in the Notch/Jag1 signalling pathway, with mRNA levels declining to 35%-62% and protein levels to 28%-35%. Additionally, it suppressed glutathione peroxidase 4 (GPX4) (decreased to approximately 31%-38%), a negative regulator of FPT, thereby facilitating HSC FPT progression and reducing profibrotic gene expression. CONCLUSION: These findings uncover a novel IGF2BP3/Notch/Jag1 signalling pathway involving HSC FPT, suggesting promising targets for ameliorating liver fibrosis. KEY POINTS/HIGHLIGHTS: IGF2BP3 deficiency inactivates Jag1 signalling. IGF2BP3 deficiency-mediated m6A modifications promote HSC ferroptosis. IGF2BP3 inhibition facilitates ferroptosis in HSCs via the Hes1/GPX4 axis. IGF2BP3 deficiency inactivates Jag1/Notch1/3/Hes1 signalling pathway inactivation, leading to the decrease in GPX4, which contributes to HSC ferroptosis.


Subject(s)
Ferroptosis , Hepatic Stellate Cells , Jagged-1 Protein , Liver Cirrhosis , RNA-Binding Proteins , Receptors, Notch , Signal Transduction , Ferroptosis/genetics , Hepatic Stellate Cells/metabolism , Animals , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Receptors, Notch/metabolism , Receptors, Notch/genetics , Mice, Knockout , Male , Humans
11.
Nat Cardiovasc Res ; 3(8): 933-950, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39196035

ABSTRACT

Valve remodeling is a process involving extracellular matrix organization and elongation of valve leaflets. Here, through single-cell RNA sequencing of human fetal valves, we identified an elastin-producing valve interstitial cell (VIC) subtype (apolipoprotein E (APOE)+, elastin-VICs) spatially located underneath valve endothelial cells (VECs) sensing unidirectional flow. APOE knockdown in fetal VICs resulted in profound elastogenesis defects. In valves with pulmonary stenosis (PS), we observed elastin fragmentation and decreased expression of APOE along with other genes regulating elastogenesis. Cell-cell interaction analysis revealed that jagged 1 (JAG1) from unidirectional VECs activates elastogenesis in elastin-VICs through NOTCH2. Similar observations were made in VICs cocultured with VECs under unidirectional flow. Notably, a drastic reduction of JAG1-NOTCH2 was also observed in PS valves. Lastly, we found that APOE controls JAG1-induced NOTCH activation and elastogenesis in VICs through the extracellular signal-regulated kinase pathway. Our study suggests important roles of both APOE and NOTCH in regulating elastogenesis during human valve remodeling.


Subject(s)
Apolipoproteins E , Elastin , Endothelial Cells , Jagged-1 Protein , Signal Transduction , Humans , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Elastin/metabolism , Elastin/genetics , Endothelial Cells/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Receptor, Notch2/metabolism , Receptor, Notch2/genetics , Cells, Cultured , Pulmonary Valve/metabolism , Coculture Techniques , Cell Communication/physiology , Heart Valves/embryology , Heart Valves/metabolism
12.
Arthritis Rheumatol ; 76(10): 1475-1487, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38961731

ABSTRACT

Notch ligands and receptors, including JAG1/2, DLL1/4, and Notch1/3, are enriched on macrophages (MΦs), fibroblast-like synoviocytes (FLS), and/or endothelial cells in rheumatoid arthritis (RA) compared with normal synovial tissues (ST). Power Doppler ultrasound-guided ST studies reveal that the Notch family is highly involved in early active RA, especially during neovascularization. In contrast, the Notch family is not implicated during the erosive stage, evidenced by their lack of correlation with radiographic damage in RA ST. Toll-like receptors and tumor necrosis factor (TNF) are the common inducers of Notch expression in RA MΦs, FLS, and endothelial cells. Among Notch ligands, JAG1 and/or DLL4 are most inducible by inflammatory responses in RA MΦs or endothelial cells and transactivate their receptors on RA FLS. TNF plays a central role on Notch ligands, as anti-TNF good responders display JAG1/2 and DLL1/4 transcriptional downregulation in RA ST myeloid cells. In in vitro studies, TNF increases Notch3 expression in MΦs, which is further amplified by RA FLS addition. Specific disease-modifying antirheumatic drugs reduced JAG1 and Notch3 expression in MΦ and RA FLS cocultures. Organoids containing FLS and endothelial cells have increased expression of JAG1 and Notch3. Nonetheless, Methotrexate, interleukin-6 receptor (IL-6R) antibodies, and B cell blockers are mostly ineffective at decreasing Notch family expression. NF-κB, MAPK, and AKT pathways are involved in Notch signaling, whereas JAK/STATs are not. Although Notch blockade has been effective in RA preclinical studies, its small molecule inhibitors have failed in phase I and II studies, suggesting that alternative strategies may be required to intercept their function.


Subject(s)
Arthritis, Rheumatoid , Receptors, Notch , Synovial Membrane , Humans , Arthritis, Rheumatoid/metabolism , Receptors, Notch/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/metabolism , Macrophages/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Synoviocytes/metabolism , Endothelial Cells/metabolism , Antirheumatic Agents/therapeutic use , Antirheumatic Agents/pharmacology , Signal Transduction , Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics
13.
Adv Sci (Weinh) ; 11(35): e2310285, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39013081

ABSTRACT

Successful dental pulp regeneration is closely associated with rapid revascularization and angiogenesis, processes driven by the Jagged1(JAG1)/Notch signaling pathway. However, soluble Notch ligands have proven ineffective in activating this pathway. To overcome this limitation, a Notch signaling hydrogel is developed by indirectly immobilizing JAG1, aimed at precisely directing the regeneration of vascularized pulp tissue. This hydrogel displays favorable mechanical properties and biocompatibility. Cultivating dental pulp stem cells (DPSCs) and endothelial cells (ECs) on this hydrogel significantly upregulate Notch target genes and key proangiogenic markers expression. Three-dimensional (3D) culture assays demonstrate Notch signaling hydrogels improve effectiveness by facilitating encapsulated cell differentiation, enhancing their paracrine functions, and promoting capillary lumen formation. Furthermore, it effectively communicates with the Wnt signaling pathway, creating an odontoinductive microenvironment for pulp-dentin complex formation. In vivo studies show that short-term transplantation of the Notch signaling hydrogel accelerates angiogenesis, stabilizes capillary-like structures, and improves cell survival. Long-term transplantation further confirms its capability to promote the formation of pulp-like tissues rich in blood vessels and peripheral nerve-like structures. In conclusion, this study introduces a feasible and effective hydrogel tailored to specifically regulate the JAG1/Notch signaling pathway, showing potential in advancing regenerative strategies for dental pulp tissue.


Subject(s)
Dental Pulp , Hydrogels , Neovascularization, Physiologic , Receptors, Notch , Regeneration , Signal Transduction , Dental Pulp/cytology , Dental Pulp/metabolism , Dental Pulp/blood supply , Receptors, Notch/metabolism , Animals , Regeneration/physiology , Regeneration/drug effects , Neovascularization, Physiologic/physiology , Neovascularization, Physiologic/drug effects , Mice , Humans , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Cell Differentiation/drug effects , Stem Cells/metabolism , Cells, Cultured
14.
Immunity ; 57(9): 2157-2172.e7, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39079536

ABSTRACT

Stroke leads to persistently high risk for recurrent vascular events caused by systemic atheroprogression that is driven by endothelial cell (EC) activation. However, whether and how stroke induces sustained pro-inflammatory and proatherogenic endothelial alterations in systemic vessels remain poorly understood. We showed that brain ischemia induces persistent activation, the upregulation of adhesion molecule VCAM1, and increased senescence in peripheral ECs until 4 weeks after stroke onset. This aberrant EC activity resulted from sustained Notch1 signaling, which was triggered by increased circulating Notch1 ligands DLL1 and Jagged1 after stroke in mice and humans. Consequently, this led to increased myeloid cell adhesion and atheroprogression by generating a senescent, pro-inflammatory endothelium. Notch1- or VCAM1-blocking antibodies and the genetic ablation of endothelial Notch1 reduced atheroprogression after stroke. Our findings revealed a systemic machinery that induces the persistent activation of peripheral ECs after stroke, which paves the way for therapeutic interventions or the prevention of recurrent vascular events following stroke.


Subject(s)
Atherosclerosis , Brain Ischemia , Calcium-Binding Proteins , Endothelial Cells , Receptor, Notch1 , Animals , Humans , Male , Mice , Atherosclerosis/metabolism , Atherosclerosis/immunology , Brain Ischemia/metabolism , Calcium-Binding Proteins/metabolism , Cell Adhesion , Cellular Senescence , Endothelial Cells/metabolism , Jagged-1 Protein/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptor, Notch1/metabolism , Signal Transduction , Stroke/metabolism , Stroke/immunology , Vascular Cell Adhesion Molecule-1/metabolism
15.
PLoS One ; 19(7): e0307943, 2024.
Article in English | MEDLINE | ID: mdl-39074091

ABSTRACT

Notch ligands [jagged (JAG) and, delta-like (DLL) families] and receptors [NOTCH family] are key regulators of Notch signaling. NOTCH signaling contributes to vascular development, tissue homeostasis, angiogenesis, and cancer progression. To elucidate the universal functions of the JAG, DLL, and NOTCH families and their connections with various biological functions, we examined 15 types of cancer using The Cancer Genome Atlas clinical database. We selected the differentially expressed genes (DEGs), which were positively correlated to the JAG, DLL, and NOTCH families in each cancer. We selected positive and negative hallmark signatures across cancer types. These indicated biological features associated with angiogenesis, hypoxia, KRAS signaling, cell cycle, and MYC targets by gene ontology and gene set enrichment analyses using DEGs. Furthermore, we analyzed single-cell RNA sequencing data to examine the expression of JAG, DLL, and NOTCH families and enrichment of hallmark signatures. Positive signatures identified using DEGs, such as KRAS signaling and hypoxia, were enriched in clusters with high expression of JAG, DLL, and NOTCH families. We subsequently validated the correlation between the JAG, DLL, and NOTCH families and clinical stages, including treatment response, metastasis, and recurrence. In addition, we performed survival analysis to identify hallmark signatures that critically affect patient survival when combining the expression of JAG, DLL, and NOTCH families. By combining the DEG enrichment and hallmark signature enrichment in survival analysis, we suggested unexplored regulatory functions and synergistic effects causing synthetic lethality. Taken together, our observations demonstrate the functions of JAG, DLL, and NOTCH families in cancer malignancy and provide insights into their molecular regulatory mechanisms.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms , Receptors, Notch , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Computer Simulation , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Clinical Relevance
16.
Environ Toxicol ; 39(9): 4417-4430, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38842024

ABSTRACT

Gliomas are the most prevalent primary malignant brain tumors worldwide. Growing evidences indicate that circular RNAs (circRNAs) play an important role in the regulation of biological behavior of tumors. We aimed to investigate the role and mechanism of circVCAN in glioma. RNase R treatment was utilized to assess the cyclic properties of circVCAN. CircVCAN, miR-488-3p, and myocyte enhancer factor 2C (MEF2C) levels in glioma tissues and cells were detected by reverse transcription real-time polymerase chain reaction (RT-qPCR), and the localization of them in glioma cells was determined with fluorescence in situ hybridization. Furthermore, a variety of biologically functional assessments were used to validate the role of circVCAN in glioma. The regulatory mechanisms of circVCAN, miR-488-3p, and MEF2C were further confirmed by double luciferase reporter gene assay, RNA immunoprecipitation and RNA pull-down assay, and the binding of MEF2C to JAGGED1 was revealed by chromatin immunoprecipitation. Additionally, a xenograft tumor model was constructed to demonstrate the effect of circVCAN on tumor growth in vivo. Our results indicated that circVCAN was more stable than its linear RNA and was significantly upregulated in gliomas. CircVCAN overexpression stimulated glioma cells to proliferate and metastasize, but circVCAN silencing exerted the opposite effect. Meanwhile, silencing circVCAN inhibited tumor growth in vivo. Moreover, we found that circVCAN interacted with miR-488-3p to regulate MEF2C expression, and miR-488-3p inhibition or MEF2C overexpression reversed the inhibitory effect on malignant bio-behaviors mediated by circVCAN knockdown in glioma cells. MEF2C promoted the transcription of JAGGED1, and circVCAN knockdown reduced the binding between MEF2C and JAGGED1. Collectively, circVCAN is a carcinogenic circRNA in glioma, and the circVCAN/miR-488-3p/MEF2C-JAGGED1 axis could serve as a potential target for the management of glioma.


Subject(s)
Brain Neoplasms , Glioma , Jagged-1 Protein , MEF2 Transcription Factors , MicroRNAs , RNA, Circular , Animals , Humans , Male , Mice , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Versicans/genetics , Versicans/metabolism
17.
Sci Rep ; 14(1): 13577, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866828

ABSTRACT

Zinc finger MIZ-type containing 1 (ZMIZ1) is a transcriptional coactivator related to the protein inhibitors of activated STATs (PIAS) family. Mounting evidence suggests that ZMIZ1 plays a crucial role in the occurrence and development of cancers. The function of ZMIZ1 in tongue squamous cell carcinoma (TSCC) and the mechanisms underpinning its role in this disease have not been fully clarified. We performed qualitative ZMIZ1 protein expression analyses using immunohistochemistry in 20 patient-derived, paraffin-embedded TSCC tissue sections. We used RNAi to knock down ZMIZ1 expression in the CAL-27 TSCC cell line and quantified the impact of ZMIZ1 knock down on proliferation, migration and apoptosis via CCK-8, scratch assay and flow cytometry, respectively. We used qRT-PCR and western blotting to investigate the role of ZMIZ1 in this cell line. Finally, we established a model of lung metastasis in nude mice to replicate the in vitro results. ZMIZ1 protein was significantly more abundant in TSCC case tissue samples. ZMIZ1 knockdown reduced the invasion and metastases of TSCC tumor cells and promoted apoptosis. ZMIZ1 knockdown was associated with the down-regulation of Notch signaling pathway related factors Jagged1 and Notch1, and invasion and metastasis related factors MKP-1, SSBP2 and MMP7 in vitro and in vivo, at the mRNA level. In vitro and in vivo data suggest that knock down of ZMIZ1 may inhibit TSCC invasion and metastasis by modulating Notch signaling. ZMIZ1 inhibition may therefore represent a new therapeutic target for TSCC.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Proliferation , Receptor, Notch1 , Signal Transduction , Tongue Neoplasms , Animals , Female , Humans , Male , Mice , Middle Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice, Nude , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Tongue Neoplasms/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Xenograft Model Antitumor Assays
18.
Iran J Kidney Dis ; 18(3): 159-167, 2024 05.
Article in English | MEDLINE | ID: mdl-38904340

ABSTRACT

INTRODUCTION: Shenqi pill (SQP) can be used to treat various kidney related diseases, but its exact mechanism of action remains unclear. We intended to analyze the role and mechanism of SQP on renal interstitial fibrosis (RIF). METHODS: After performing unilateral ureteral obstruction (UUO) surgery following the Institutional Animal Care and Use Committee guidelines, all rats were assigned into the sham group, UUO group, UUO + SQP 1.5 g/kg, UUO + SQP 3 g/kg, and UUO + SQP 6 g/kg groups. After treatment with SQP for 4 weeks, the appearance of kidney, serum creatinine (SCr), and blood urea nitrogen (BUN) levels were monitored in each group. The pathological injury, extracellular matrix (ECM), and Notch1 pathway-related protein levels were measured using H&E staining, Masson staining, immunohistochemistry, and Western blot, respectively. RESULTS: SQP could obviously ameliorate the appearance of the kidney as well as the levels of SCr and BUN in UUO rats (SCr: 67.6 ± 4.64 µM, 59.66 ± 4.96 µM, 48.76 ± 4.44 µM, 40.43 ± 3.02 µM for UUO, low, medium, and high SQP treatment groups; BUN: 9.09 ± 0.97 mM, 7.72 ± 0.61 mM, 5.42 ± 0.42 mM, 4.24 ± 0.34 mM for UUO, low, medium, and high SQP treatment groups; P < .05). SQP also effectively mitigated renal tissue injury in UUO rats (P < .05). Moreover, we uncovered that SQP significantly inhibited Collagen I, α-SMA, Collagen IV, TGF-B1, Notch1, and Jag1 protein expressions in UUO rats kidney (P < .05). CONCLUSION: Our data elucidated that SQP can alleviate RIF, and the mechanism may be related to the Notch1/Jag1 pathway. DOI: 10.52547/ijkd.7703.


Subject(s)
Blood Urea Nitrogen , Drugs, Chinese Herbal , Fibrosis , Jagged-1 Protein , Kidney , Rats, Sprague-Dawley , Receptor, Notch1 , Signal Transduction , Ureteral Obstruction , Animals , Drugs, Chinese Herbal/pharmacology , Male , Receptor, Notch1/metabolism , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/complications , Ureteral Obstruction/pathology , Rats , Signal Transduction/drug effects , Jagged-1 Protein/metabolism , Disease Models, Animal , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Kidney Diseases/prevention & control , Kidney Diseases/metabolism , Creatinine/blood , Transforming Growth Factor beta1/metabolism , Actins/metabolism
19.
Nat Commun ; 15(1): 5152, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886396

ABSTRACT

In many cancers, a stem-like cell subpopulation mediates tumor initiation, dissemination and drug resistance. Here, we report that cancer stem cell (CSC) abundance is transcriptionally regulated by C-terminally phosphorylated p27 (p27pT157pT198). Mechanistically, this arises through p27 co-recruitment with STAT3/CBP to gene regulators of CSC self-renewal including MYC, the Notch ligand JAG1, and ANGPTL4. p27pTpT/STAT3 also recruits a SIN3A/HDAC1 complex to co-repress the Pyk2 inhibitor, PTPN12. Pyk2, in turn, activates STAT3, creating a feed-forward loop increasing stem-like properties in vitro and tumor-initiating stem cells in vivo. The p27-activated gene profile is over-represented in STAT3 activated human breast cancers. Furthermore, mammary transgenic expression of phosphomimetic, cyclin-CDK-binding defective p27 (p27CK-DD) increases mammary duct branching morphogenesis, yielding hyperplasia and microinvasive cancers that can metastasize to liver, further supporting a role for p27pTpT in CSC expansion. Thus, p27pTpT interacts with STAT3, driving transcriptional programs governing stem cell expansion or maintenance in normal and cancer tissues.


Subject(s)
Breast Neoplasms , Cyclin-Dependent Kinase Inhibitor p27 , Hyperplasia , Neoplastic Stem Cells , STAT3 Transcription Factor , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Humans , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Female , Phosphorylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Hyperplasia/metabolism , Mice , Gene Expression Regulation, Neoplastic , Cell Self Renewal/genetics , Cell Line, Tumor , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/cytology , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics
20.
Curr Eye Res ; 49(10): 1098-1106, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38783634

ABSTRACT

PURPOSE: Microglia-related inflammation is closely linked to the pathogenesis of retinal diseases. The primary objective of this research was to investigate the impact and mechanism of M1 phenotype microglia on the barrier function of retina microvascular endothelial cells. METHODS: Quantitative polymerase chain reactions and western blot techniques were utilized to analysis the mRNA and protein expressions of M1 and M2 markers of human microglial clone 3 cell line (HMC3), as well as the levels of Notch ligands and receptors under the intervention of lipopolysaccharide (LPS) or interleukin (IL)-4. ELISA was utilized to detect the pro-inflammatory and anti-inflammatory cytokines from HMC3 cells. The cellular tight junction and apoptosis of human retinal microvascular endothelial cells (HRMECs) were assessed by western blot and fluorescein isothiocyanate-dextran permeability assay. The inhibitors of Notch1 and RNA interference (RNAi) targeting Jagged1 were used to assess their contribution to the barrier function of vascular endothelial cells. RESULTS: Inducible nitric oxide synthase (iNOS) and IL-1ß were considerably elevated in LPS-treated HMC3, while CD206 and Arg-1 markedly elevated under IL-4 stimulation. The conditioned medium derived from LPS-treated HMC3 cells promoted permeability, diminished the expression of zonula occludens-1 and Occludin, and elevated the expression of Cleaved caspase-3 in HRMECs. RNAi targeting Jagged1 or Notch1 inhibitor could block M1 HMC3 polarization and maintain barrier function of HRMECs. CONCLUSION: Our findings suggest that Jagged1-Notch1 signaling pathway induces M1 microglial cells to disrupt the barrier function of HRMECs, which may lead to retinal diseases.


Subject(s)
Blotting, Western , Jagged-1 Protein , Microglia , Receptor, Notch1 , Retinal Vessels , Signal Transduction , Humans , Microglia/metabolism , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Signal Transduction/physiology , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Retinal Vessels/cytology , Retinal Vessels/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Endothelial Cells/metabolism , Blood-Retinal Barrier , Apoptosis , Real-Time Polymerase Chain Reaction , Endothelium, Vascular/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL