Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
1.
Oral Oncol ; 156: 106953, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39004024

ABSTRACT

The study by Feeney et al. provides critical insights into the prognostic implications of NOTCH pathway activation in adenoid cystic carcinoma (ACC), particularly after disease recurrence. Utilizing both next-generation sequencing and immunohistochemistry, the research delineates the survival outcomes between NOTCH-activated and non-activated ACC groups, highlighting poorer outcomes in the former. The findings advocate for the targeted therapeutic approach and suggest a potential for personalized treatment strategies, emphasizing the need for further research into NOTCH pathway inhibitors and their clinical applications.


Subject(s)
Carcinoma, Adenoid Cystic , Neoplasm Recurrence, Local , Receptors, Notch , Signal Transduction , Carcinoma, Adenoid Cystic/metabolism , Carcinoma, Adenoid Cystic/mortality , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Adenoid Cystic/genetics , Humans , Receptors, Notch/metabolism , Receptors, Notch/genetics , Neoplasm Recurrence, Local/metabolism , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/mortality , Salivary Gland Neoplasms/genetics , Prognosis
2.
Article in English | MEDLINE | ID: mdl-38946043

ABSTRACT

Radiation therapy (RT) is a common treatment for lung cancer. Still, it can lead to irreversible loss of pulmonary function and a significant reduction in quality of life for one-third of patients. Preexisting comorbidities, such as chronic obstructive pulmonary disease (COPD), are frequent in patients with lung cancer and further increase the risk of complications. Because lung stem cells are crucial for the regeneration of lung tissue following injury, we hypothesized that airway stem cells from patients with COPD with lung cancer might contribute to increased radiation sensitivity. We used the air-liquid interface model, a three-dimensional (3D) culture system, to compare the radiation response of primary human airway stem cells from healthy and patients with COPD. We found that COPD-derived airway stem cells, compared to healthy airway stem cell cultures, exhibited disproportionate pathological mucociliary differentiation, aberrant cell cycle checkpoints, residual DNA damage, reduced survival of stem cells and self-renewal, and terminally differentiated cells post-irradiation, which could be reversed by blocking the Notch pathway using small-molecule γ-secretase inhibitors. Our findings shed light on the mechanisms underlying the increased radiation sensitivity of COPD and suggest that airway stem cells reflect part of the pathological remodeling seen in lung tissue from patients with lung cancer receiving thoracic RT.

3.
J Mol Cell Biol ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085037

ABSTRACT

Drosophila melanogaster crystal cells are a specialized type of blood cells for innate immune process upon injury. Under normal conditions, crystal cells rarely proliferate and constitute a small proportion of fly blood cells. Notch signaling has been known to guide the cell fate determination of crystal cells and maintain their survival. Here, we reported that protein phosphatase V (PpV), the unique catalytic subunit of protein phosphatase 6 in Drosophila, is a novel regulator of crystal cell proliferation and integrity. We found that PpV proteins highly accumulated in crystal cells in the larval hematopoietic organ termed the lymph gland. Silencing PpV using RNA interference led to increased crystal cell proliferation in a Notch-independent manner and induced crystal cell rupture dependent on Notch signaling. Moreover, additive PpV prevented the rupture of crystal cells in lymph glands upon a needle injury, suggesting the involvement of PpV in wound healing. Altogether, our results indicated that PpV plays a dual role in lymph glands, preventing crystal cell proliferation to limit the cell number, as well as inhibiting crystal cell rupture to maintain their survival.

4.
Inflammation ; 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39078585

ABSTRACT

Doxorubicin (DOX) is an effective chemotherapeutic drug; however, its cardiotoxicity and resistance compromise its therapeutic index. The Notch pathway was reported to contribute to DOX cancer resistance. The role of Notch pathway in DOX cardiotoxicity has not been identified yet. Notch receptors are characterized by their extracellular (NECD) and intracellular (NICD) domains (NICD). The γ-secretase enzyme helps in the release of NICD. Dibenzazepine (DBZ) is a γ-secretase inhibitor. The present study investigated the effect of Notch pathway inhibition on DOX cardiotoxicity. Twenty-four male Wistar rats were divided into four groups: control group, DOX group, acute cardiotoxicity was induced by a single dose of DOX (20 mg/kg) i.p., DOX (20 mg/kg) plus DBZ group, and DBZ group. The third and fourth groups received i.p. injection of DBZ daily for 14 days at 2 mg/kg dose. DOX cardiotoxicity increased the level of serum creatine kinase-MB and cardiac troponin I, and it was confirmed by the histopathological examination. Moreover, the antioxidants glutathione peroxidase and superoxide dismutase levels were markedly decreased, and the inflammatory markers, inducible nitric oxide synthase, nuclear factor-kB, and tumor necrosis factor-α were markedly increased. Furthermore, DOX increased BAX protein and downregulated BCL-2. In addition, DOX upregulated Notch pathway-related parameters: Hes1 and Hey1 mRNA levels, and increased Hes1 protein levels. DBZ ameliorated DOX-induced cardiotoxicity, evidenced by reducing the cardiac injury biomarkers, improving cardiac histopathological changes, correcting antioxidant levels, and reducing inflammatory and apoptotic proteins. Our study indicates the protective effect of Notch inhibitor against DOX-induced cardiotoxicity.

5.
Med Mol Morphol ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38987402

ABSTRACT

Primary cultured odontoblasts rapidly lose their tissue-specific phenotype. To identify transcription factors (TF) that are important for the maintenance of the odontoblast phenotype, primary cultures of C57BL/6 J mouse dental mesenchymal cells (DMC) were isolated, and expression of TF and odontoblast marker genes in cells immediately after isolation and 2 days after culture were comprehensively evaluated and compared using RNA-sequencing (RNA-seq). The expression of odontoblast markers in mouse dental mesenchymal cells decreased rapidly after isolation. In addition, the expression of Hedgehog-related, Notch-related, and immediate- early gene (IEG)-related transcription factors significantly decreased. Forced expression of these genes in lentiviral vectors, together with fibroblast growth factor 4 (FGF4), fibroblast growth factor 9 (FGF9), and the Wnt pathway activator CHIR99021, significantly induced the expression of odontogenic marker genes. These results indicate, for the first time, that Notch signaling and early genes may be important for maintaining odontoblast cultures. Furthermore, simultaneous stimulation of FGF, Wnt, Hedgehog, Notch pathways, and IEG transcription factors cooperatively promoted the maintenance of the odontoblast phenotype. These results suggest that the Hedgehog and Notch signaling pathways may play an important role in maintaining odontoblast phenotypes, in addition to FGF and Wnt signaling.

6.
Dev Cell ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38944040

ABSTRACT

We describe a next-generation Drosophila protein interaction map-"DPIM2"-established from affinity purification-mass spectrometry of 5,805 baits, covering the largest fraction of the Drosophila proteome. The network contains 32,668 interactions among 3,644 proteins, organized into 632 clusters representing putative functional modules. Our analysis expands the pool of known protein interactions in Drosophila, provides annotation for poorly studied genes, and postulates previously undescribed protein interaction relationships. The predictive power and functional relevance of this network are probed through the lens of the Notch signaling pathway, and we find that newly identified members of complexes that include known Notch modifiers can also modulate Notch signaling. DPIM2 allows direct comparisons with a recently published human protein interaction network, defining the existence of functional interactions conserved across species. Thus, DPIM2 defines a valuable resource for predicting protein co-complex memberships and functional associations as well as generates functional hypotheses regarding specific protein interactions.

7.
J Cancer ; 15(11): 3466-3480, 2024.
Article in English | MEDLINE | ID: mdl-38817852

ABSTRACT

Background: Cancer development involves alterations in key cellular pathways, with aspartate ß-hydroxylase (ASPH) emerging as an important player in tumorigenesis. ASPH is upregulated in various cancer types, where it promotes cancer progression mainly by regulating the Notch1 and SRC pathways. Methods: This study explored the responses of various human cervical, pharyngeal, and breast tumor cell lines to second- and third-generation ASPH inhibitors (MO-I-1151 and MO-I-1182) using proliferation, migration, and invasion assays; western blotting; and cell cycle analysis. Results: ASPH inhibition significantly reduced cell proliferation, migration, and invasion and disrupted both the canonical and noncanonical Notch1 pathways. The noncanonical pathway was particularly mediated by AKT signaling. Cell cycle analysis revealed a marked reduction in cyclin D1 expression, further confirming the inhibitory effect of ASPH inhibitors on cell proliferation. Additional analysis revealed G0/G1 arrest and restricted progression into S phase, highlighting the regulatory impact of ASPH inhibitors on the cell cycle. Furthermore, ASPH inhibition induced distinctive alterations in nuclear morphology. The high heterogeneity in the responses of individual tumor cell lines to ASPH inhibitors, both quantitatively and qualitatively, underscores the complex network of mechanisms that are regulated by ASPH and influence the efficacy of ASPH inhibition. The effects of ASPH inhibitors on Notch1 pathway activity, cyclin D1 expression, and nuclear morphology contribute to the understanding of the multifaceted effects of these inhibitors on cancer cell behavior. Conclusion: This study not only suggests that ASPH inhibitors are effective against tumor cell progression, in part through the induction of cell cycle arrest, but also highlights the diverse and heterogeneous effects of these inhibitors on the behavior of tumor cells of different origins.

8.
Stem Cells Transl Med ; 13(7): 661-677, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38709826

ABSTRACT

Loss of cochlear hair cells (HCs) leads to permanent hearing loss in mammals, and regenerative medicine is regarded as an ideal strategy for hearing recovery. Limited genetic and pharmaceutical approaches for HC regeneration have been established, and the existing strategies cannot achieve recovery of auditory function. A promising target to promote HC regeneration is MEK/ERK signaling because dynamic shifts in its activity during the critical stages of inner ear development have been observed. Here, we first showed that MEK/ERK signaling is activated specifically in supporting cells (SCs) after aminoglycoside-induced HC injury. We then selected 4 MEK/ERK signaling inhibitors, and PD0325901 (PD03) was found to induce the transdifferentiation of functional supernumerary HCs from SCs in the neonatal mammalian cochlear epithelium. We next found that PD03 facilitated the generation of HCs in inner ear organoids. Through genome-wide high-throughput RNA sequencing and verification, we found that the Notch pathway is the downstream target of MEK/ERK signaling. Importantly, delivery of PD03 into the inner ear induced mild HC regeneration in vivo. Our study thus reveals the importance of MEK/ERK signaling in cell fate determination and suggests that PD03 might serve as a new approach for HC regeneration.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , MAP Kinase Signaling System , Receptors, Notch , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , MAP Kinase Signaling System/drug effects , Mice , Receptors, Notch/metabolism , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Labyrinth Supporting Cells/metabolism
9.
Pathol Res Pract ; 257: 155282, 2024 May.
Article in English | MEDLINE | ID: mdl-38608371

ABSTRACT

Cancer is a group of diseases marked by unchecked cell proliferation and the ability for the disease to metastasize to different body areas. Enhancements in treatment and early detection are crucial for improved outcomes. LncRNAs are RNA molecules that encode proteins and have a length of more than 200 nucleotides. LncRNAs are crucial for chromatin architecture, gene regulation, and other cellular activities that impact both normal growth & pathological processes, even though they are unable to code for proteins. LncRNAs have emerged as significant regulators in the study of cancer biology, with a focus on their intricate function in the Notch signaling pathway. The imbalance of this pathway is often linked to a variety of malignancies. Notch signaling is essential for cellular functions like proliferation, differentiation, and death. The cellular response is shaped by these lncRNAs through their modulation of essential Notch pathway constituents such as receptors, ligands, and downstream effectors around it. Furthermore, a variety of cancer types exhibit irregular expression of Notch-related lncRNAs, underscoring their potential use as therapeutic targets and diagnostic markers. Gaining an understanding of the molecular processes behind the interaction between the Notch pathway and lncRNAs will help you better understand the intricate regulatory networks that control the development of cancer. This can open up new possibilities for individualized treatment plans and focused therapeutic interventions. The intricate relationships between lncRNAs & the Notch pathway in cancer are examined in this review.


Subject(s)
Neoplasms , RNA, Long Noncoding , Receptors, Notch , Signal Transduction , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction/genetics , Gene Expression Regulation, Neoplastic/genetics , Animals
10.
Mol Oncol ; 18(6): 1510-1530, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459621

ABSTRACT

The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Nuclear Receptor Interacting Protein 1 , Transcription Factor HES-1 , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Nuclear Receptor Interacting Protein 1/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics
11.
Reprod Biol ; 24(2): 100878, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38490111

ABSTRACT

It was reported previously that in adult males disruption of both androgen and Notch signaling impairs spermatid development and germ cell survival in rodent seminiferous epithelium. To explain the molecular mechanisms of these effects, we focused on the interaction between Notch signaling and androgen receptor (AR) in Sertoli cells and investigate its role in the control of proteins involved in apical ectoplasmic specializations, actin remodeling during spermiogenesis, and induction of germ cell apoptosis. First, it was revealed that in rat testicular explants ex vivo both testosterone and Notch signaling modulate AR expression and cooperate in the regulation of spermiogenesis-related genes (Nectin2, Afdn, Arp2, Eps8) and apoptosis-related genes (Fasl, Fas, Bax, Bcl2). Further, altered expression of these genes was found following exposure of Sertoli cells (TM4 cell line) and germ cells (GC-2 cell line) to ligands for Notch receptors (Delta-like1, Delta-like4, and Jagged1) and/or Notch pathway inhibition. Finally, direct interactions of Notch effector, Hairy/enhancer-of-split related with YRPW motif protein 1, and the promoter of Ar gene or AR protein were revealed in TM4 Sertoli cells. In conclusion, Notch pathway activity in Sertoli and germ cells regulates genes related to germ cell development and apoptosis acting both directly and indirectly by influencing androgen signaling in Sertoli cells.


Subject(s)
Androgens , Apoptosis , Receptors, Androgen , Receptors, Notch , Seminiferous Epithelium , Sertoli Cells , Signal Transduction , Spermatogenesis , Male , Animals , Apoptosis/physiology , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction/physiology , Rats , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Sertoli Cells/physiology , Androgens/metabolism , Spermatogenesis/physiology , Cell Line , Germ Cells/metabolism , Testosterone/metabolism , Rats, Wistar
12.
J Surg Res ; 297: 63-70, 2024 May.
Article in English | MEDLINE | ID: mdl-38447337

ABSTRACT

INTRODUCTION: Diabetic foot ulcer (DFU) is a severe complication that threatens the daily lives of patients with diabetes and represents a serious challenge to the global health system. Considering that impaired wound healing is the leading cause of DFU, exploring the mechanism of diabetic wound healing is beneficial for improving DFU treatment. Resveratrol (RES) is a native polyphenol with various pharmacological characteristics, and recent studies have indicated an accelerated function of RES in diabetic wound healing. As human dermal fibroblasts (HDFs) play a significant role in diabetic wound healing, this study aimed to elucidate the regulatory mechanism of RES in HDFs. METHODS: To mimic diabetic wound healing in vitro, the HDFs were stimulated with high glucose (HG). Our findings revealed that RES reversed HG-induced suppression of HDF proliferation and migration caused by HG. RES inhibits the Notch signaling pathway. More importantly, we demonstrated that the activation of the Notch pathway abrogated the effects of RES on HG-induced HDFs. RESULTS: In vivo assays also illustrated that RES contributed to wound healing in diabetic mice by blocking the Notch pathway. CONCLUSIONS: In conclusion, RES improved diabetic wound healing by targeting the Notch pathway, which offers novel insights into DFU therapy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Foot , Humans , Mice , Animals , Resveratrol/pharmacology , Diabetes Mellitus, Experimental/metabolism , Wound Healing , Skin/metabolism
13.
Dig Dis Sci ; 69(3): 821-834, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38172445

ABSTRACT

BACKGROUND: Glutamate-rich WD repeat containing 1 (GRWD1) is over-expressed in a variety of malignant tumors and is considered to be a potential oncogene. However, its mechanism of action in gastric cancer (GC) is still unclear. METHODS: Data analysis, Immunohistochemistry, and Western Blot (WB) were performed to verify the expression of GRWD1 in GC and para-cancerous tissues. The association between GRWD1 expression and tumor size, tissue differentiation, lymph node metastasis, TNM stage, and prognosis was analyzed according to the high and low expression levels of GRWD1. The relationship between GRWD1 and Notch pathway was verified by data analysis and WB. The effects of GRWD1 on the proliferation, migration, and invasion of GC cells were verified by cell proliferation, migration, and invasion assays. We confirmed that the high expression of GRWD1 promoted the proliferation of GC cells in vivo through the tumor formation assay in nude mice. RESULTS: The expression of GRWD1 was higher in GC tissues than in para-cancerous tissues, and its expression was positively correlated with tumor size, lymph node metastasis, and TNM stage, but negatively correlated with differentiation grade and prognosis. GRWD1 over-expression increased ADAM metallopeptidase domain 17 (ADAM17) expression and promoted Notch1 intracellular domain (NICD) release to promote GC cell proliferation, migration, and invasion in vitro. Results from animal studies have shown that high GRWD1 expression could promote GC cell proliferation in vivo by activating the Notch signaling pathway. CONCLUSION: GRWD1 promotes GC progression through ADAM17-dependent Notch signaling, and GRWD1 may be a novel tumor marker and therapeutic target.


Subject(s)
ADAM17 Protein , Carrier Proteins , Stomach Neoplasms , Animals , Mice , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lymphatic Metastasis , Mice, Nude , Neoplasm Invasiveness , Signal Transduction , Stomach Neoplasms/pathology , Up-Regulation , Carrier Proteins/metabolism , ADAM17 Protein/metabolism
14.
Int J Mol Med ; 53(3)2024 03.
Article in English | MEDLINE | ID: mdl-38240118

ABSTRACT

The senolytics dasatinib and quercetin (DQ) alleviate age­related disorders. However, limited information is available regarding the effects of DQ on diabetic kidney disease (DKD). The present study aimed to explore the effects of DQ on DKD and its potential molecular mechanism(s). Dasatinib (5 mg/kg) and quercetin (50 mg/kg) were administered to diabetic db/db mice by gavage for 20 weeks. Body weight, urine albumin­creatinine ratio (ACR), serum creatinine (Scr), and blood urea nitrogen (BUN) were recorded at the indicated time periods. Periodic acid­Schiff and Masson's staining were performed to assess the histopathological changes of kidney tissues. Immunohistochemical analysis, immunofluorescence and western blotting were performed to evaluate the expression levels of extracellular matrix (ECM) proteins, autophagic and podocyte differentiation­related proteins. In addition, mouse podocytes were administered with high­glucose, DQ and 3­methyladenine (3­MA), and the expression levels of autophagic and podocyte differentiation­related proteins were measured. Moreover, following overexpression of the Notch intracellular domain (NICD), the expression levels of NICD, autophagic and podocyte differentiation­related proteins were further assessed. DQ significantly reduced the body weight, blood glucose, ACR, Scr and BUN levels and improved the histopathological changes induced in diabetic db/db mice. In addition, DQ caused a significant downregulation of the expression levels of the ECM proteins, improved autophagy and induced an upregulation of the expression levels of podocyte differentiation­related proteins. Administration of 3­MA to mice significantly reduced podocyte differentiation, and overexpression of NICD could reverse the effects of DQ on autophagy and podocyte differentiation in vitro. The present study suggests that DQ protects against DKD by activation of autophagy to alleviate podocyte dedifferentiation via the Notch pathway.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Mice , Animals , Diabetic Nephropathies/metabolism , Podocytes/metabolism , Dasatinib/metabolism , Dasatinib/pharmacology , Quercetin/pharmacology , Quercetin/therapeutic use , Senotherapeutics , Autophagy , Body Weight , Diabetes Mellitus/pathology
15.
bioRxiv ; 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-38260427

ABSTRACT

Organisms adjust their physiology to cope with environmental fluctuations and maintain fitness. These adaptations occur via genetic changes over multiple generations or through acclimation, a set of reversible phenotypic changes that confer resilience to the individual. Aquatic organisms are subject to dramatic seasonal fluctuations in water salinity, which can affect the function of lateral line mechanosensory hair cells. To maintain hair cell function when salinity decreases, ion-regulating cells, Neuromast-associated ionocytes (Nm ionocytes), increase in number and invade lateral line neuromasts. How environmental changes trigger this adaptive differentiation of Nm ionocytes and how these cells are specified is still unknown. Here, we identify Nm ionocyte progenitors as foxi3a/foxi3b-expressing skin cells and show that their differentiation is associated with sequential activation of different Notch pathway components, which control ionocyte survival. We demonstrate that new Nm ionocytes are rapidly specified by absolute salinity levels, independently of stress response pathways. We further show that Nm ionocyte differentiation is selectively triggered by depletion of specific ions, such as Ca2+ and Na+/Cl-, but not by low K+ levels, and is independent of media osmolarity. Finally, we demonstrate that hair cell activity plays a role in Nm ionocyte recruitment and that systemic factors are not necessary for Nm ionocyte induction. In summary, we have identified how environmental changes activate a signaling cascade that triggers basal skin cell progenitors to differentiate into Nm ionocytes and invade lateral line organs. This adaptive behavior is an example of physiological plasticity that may prove essential for survival in changing climates.

16.
J Biochem Mol Toxicol ; 38(1): e23628, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38229317

ABSTRACT

This study aimed to explore the mechanism by which postembryonic renal ADAMTS18 methylation influences obstructive renal fibrosis in rats. After exposure to transforming growth factor (TGF)-ß1 during the embryonic period, analysis of postembryonic renal ADAMTS18 methylation and expression levels was conducted. Histological analysis was performed to assess embryonic kidney lesions and damage. Western blot analysis was used to determine the expression of renal fibrosis markers. Rats with ureteral obstruction and a healthy control group were selected. The methylation levels of ADAMTS18 in the different groups were analyzed. Western blot analysis and immunohistochemistry were performed to analyze the expression of renal fibrosis markers, and kidney-related indicators were measured. Treatment with TGF-ß1 resulted in abnormal development of the postembryonic kidney, which was characterized by rough kidney surfaces with mild depressions and irregularities on the outer surface. TGF-ß1 treatment significantly promoted ADAMTS18 methylation and activated the protein kinase B (AKT)/Notch pathway. Ureteral obstruction was induced to establish a renal hydronephrosis model, which led to renal fibrotic injury in newborn rats. Overexpression of the ADAMTS18 gene alleviated renal fibrosis. The western blot results showed that compared to that in the control group, the expression of renal fibrosis markers was significantly decreased after ADAMTS18 overexpression, and there was a thicker renal parenchymal tissue layer and significantly reduced p-AKT/AKT and Notch1 levels. TGF-ß1 can induce ADAMTS18 gene methylation in the postembryonic kidney, and the resulting downregulation of ADAMTS18 expression has long-term effects on kidney development, potentially leading to increased susceptibility to obstructive renal fibrosis. This mechanism may involve activation of the AKT/Notch pathway. Reversing ADAMTS18 gene methylation may reverse this process.


Subject(s)
ADAMTS Proteins , Kidney Diseases , Ureteral Obstruction , Animals , Rats , Fibrosis , Kidney , Kidney Diseases/metabolism , Methylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , ADAMTS Proteins/genetics
17.
Curr Mol Med ; 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38213137

ABSTRACT

BACKGROUND: The emergence of treatment resistance has hindered the efficacy of targeted therapies used to treat patients with hepatocellular carcinoma (HCC). OBJECTIVE: This study aimed to explore the mechanism of organoids constructed from lenvatinib-resistant HCC cells. METHODS: Hep3B cell and human HCC organoids were cultured and identified using hematoxylin and eosin staining and Immunohistochemistry. Lenvatinib-sensitive/ resistant Hep3B cells were constructed using lenvatinib (0, 0.1, 1, and 10 µM) and lenvatinib (0, 1, 10, and 100 µM). qRT-PCR and flow cytometry were utilized to determine HCC stem cell markers CD44, CD90, and CD133 expressions. Transcriptome sequencing was performed on organoids.-Western blot evaluated Notch pathwayrelated proteins (NOTCH1 and Jagged) expressions. Furthermore, DAPT, an inhibitor of the Notch pathway, was used to investigate the effects of lenvatinib on resistance or stemness in organoids and human HCC tissues. RESULTS: The organoids were successfully cultivated. With the increase of lenvatinib concentration, sensitive cell organoids were markedly degraded and ATP activity was gradually decreased, while there was no significant change in ATP activity of resistant cell organoids. CD44 expressions were elevated after lenvatinib treatment compared with the control group. KEGG showed that lenvatinib treatment of organoids constructed from Hep3B cells mainly activated the Notch pathway. Compared with the control group, NOTCH1 and Jagged expressions elevated, and ATP activity decreased after lenvatinib treatment. However, ATP activity was notably decreased after DAPT treatment. Moreover, DAPT inhibited lenvatinib resistance and the increase in the expressions of CD44 caused by lenvatinib. Besides, 100 µM lenvatinib significantly inhibited the growth and ATP activity of human HCC organoids, and DAPT increased the inhibitory effect of lenvatinib. CONCLUSION: Lenvatinib regulated resistance and stemness in organoids via the Notch pathway.

18.
Poult Sci ; 103(1): 103171, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37925772

ABSTRACT

Vaccination is still the main method of preventing most infectious diseases, but there are inefficiencies and inaccuracies in immunization. Studies have reported that Atractylodis macrocephalae Koidz. polysaccharides (RAMP) have immunomodulatory effects, but the mechanisms involved in whether they can modulate the immune response in chickens are not yet clear. The aim of this study was to investigate the effect of RAMP on lymphocytes functions by analyzing cell proliferation, cell cycle, mRNA expression of cytokines and CD4 +/CD8 + ratio. To identify potential molecules involved in immune regulation, we performed a comprehensive transcriptome profiling of chicken lymphocytes. In addition, the adjuvant effect of RAMP was evaluated by detecting indicators of hemagglutination inhibition. When lymphocytes were cultured with RAMP in vitro, the proliferation rate of lymphocytes was increased (P < 0.01), more cells in S phase and G2/M phase (P < 0.01) and the mRNA expression of IFN-γ was upregulated (P < 0.05), while the mRNA expression of TGF-ß (P < 0.01) and IL-4 (P < 0.05) was downregulated and the CD4 +/CD8 + ratio was increased (P < 0.05). Transcriptomic results showed that RAMP increased the expression of HIST1H46 (P < 0.05) and CENPP (P < 0.05). Validation of qPCR showed that RAMP may play an important role in regulating cellular immunity by downregulating the Notch pathway. The results also showed that RAMP could increase the serum Newcastle disease virus antibody levels in chickens. These data suggest that RAMP could enhance immune function of lymphocytes and was a candidate vaccine adjuvant in chickens.


Subject(s)
Chickens , Cytokines , Animals , Chickens/genetics , Cytokines/genetics , Cytokines/metabolism , Immunity, Cellular , Polysaccharides/pharmacology , RNA, Messenger
19.
J Leukoc Biol ; 115(2): 322-333, 2024 01 19.
Article in English | MEDLINE | ID: mdl-37726110

ABSTRACT

Scavenger receptor A (SRA) is preferentially expressed in macrophages and implicated as a multifunctional pattern recognition receptor for innate immunity. Hepatic macrophages play a primary role in the pathogenesis of alcoholic liver disease. Herein, we observed that SRA expression was significantly increased in the liver tissues of mice with alcohol-related liver injury. SRA-deficient (SRA-/-) mice developed more severe alcohol-induced liver disease than wild-type mice. Enhanced liver inflammation existed in alcohol-challenged SRA-/- mice and was associated with increased Notch activation in hepatic macrophages compared with wild-type control animals. Mechanistically, SRA directly bound with Notch1 and suppressed its S-glutathionylation, thereby inhibiting Notch pathway activation. Further, we determined that the SRA interacted with thioredoxin-1 (Trx-1), a redox-active protein. SRA inhibited Trx-1 dimerization and facilitated the interaction of Trx-1 with Notch1. Application of a Trx-1-specific inhibitory agent during macrophage stimulation abolished SRA-mediated regulation of the Notch pathway and its downstream targets. In summary, our study revealed that SRA plays a critical role in macrophage inflammatory response by targeting Notch1 for its glutathionylation. SRA-mediated negative regulation of Notch activation might serve as a novel therapeutic strategy for alcohol-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Mice , Animals , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Scavenger Receptors, Class A/metabolism , Macrophages/metabolism , Receptors, Scavenger/metabolism , Liver/metabolism , Immunologic Factors , Ethanol/toxicity , Thioredoxins/genetics , Thioredoxins/metabolism , Mice, Inbred C57BL , Mice, Knockout
20.
J Biol Chem ; 300(1): 105522, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043798

ABSTRACT

Notch signaling plays a critical role in cell fate decisions in all cell types. Furthermore, gain-of-function mutations in NOTCH1 have been uncovered in many human cancers. Disruption of Notch signaling has recently emerged as an attractive disease treatment strategy. However, the nuclear interaction landscape of the oncoprotein NOTCH1 remains largely unexplored. We therefore employed here a proximity-dependent biotin identification approach to identify in vivo protein associations with the nuclear Notch1 intracellular domain in live cells. We identified a large set of previously reported and unreported proteins that associate with NOTCH1, including general transcription and elongation factors, DNA repair and replication factors, coactivators, corepressors, and components of the NuRD and SWI/SNF chromatin remodeling complexes. We also found that Notch1 intracellular domain associates with protein modifiers and components of other signaling pathways that may influence Notch signal transduction and protein stability such as USP7. We further validated the interaction of NOTCH1 with histone deacetylase 1 or GATAD2B using protein network analysis, proximity-based ligation, in vivo cross-linking and coimmunoprecipitation assays in several Notch-addicted cancer cell lines. Through data mining, we also revealed potential drug targets for the inhibition of Notch signaling. Collectively, these results provide a valuable resource to uncover the mechanisms that fine-tune Notch signaling in tumorigenesis and inform therapeutic targets for Notch-addicted tumors.


Subject(s)
Carcinogenesis , Neoplasms , Oncogene Proteins , Receptor, Notch1 , Humans , Cell Differentiation , Cell Line , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Signal Transduction , Ubiquitin-Specific Peptidase 7/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Neoplasms/genetics , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL